1
|
Das S, Pal U, Saha-Dasgupta T, Leong S. Combating Fuel Biocontamination: Tailored Antimicrobial Peptides and an Innovative Delivery Strategy. ACS APPLIED BIO MATERIALS 2025. [PMID: 40400296 DOI: 10.1021/acsabm.5c00474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2025]
Abstract
Microbial invasion and subsequent fuel biocontamination have long posed significant challenges, leading to a significant infrastructural damage. The lack of systematic data on the correlation between environmental parameters and microbial growth has hampered the development of targeted solutions to date. To address this challenge, this study reports a targeted strategy to inactivate and control the proliferation of commonly identified fuel-contaminating microbial clusters through the development of synthetic peptides that can be delivered directly to fuel samples. From a library of short peptides which was designed based on the indolicidin template peptides, three unique sequences were found to have good broad-spectrum activity toward a range of microbes such as Bacillus, Sphingomonas, and Hormoconis, with P17, showing the highest killing potential. The structural analyses of the peptides based on circular dichroism spectroscopy revealed the helical propensity of the peptides in SDS micelles and a random flexible structure in solution. The peptides showed stability under biological conditions and minimal cytotoxicity against mammalian cells. This study presents an innovative method to effectively address fuel biocontamination using short peptides coupled with a potentially scalable protocol to administer the peptides to fuel samples.
Collapse
Affiliation(s)
- Swagata Das
- Food, Chemical and Biotechnology Cluster, Singapore Institute of Technology, Singapore 138683, Singapore
| | - Uttam Pal
- Technical Research Centre, S. N. Bose National Centre for Basic Sciences, Kolkata 700106, India
| | | | - Susanna Leong
- Food, Chemical and Biotechnology Cluster, Singapore Institute of Technology, Singapore 138683, Singapore
| |
Collapse
|
2
|
Lee WH, Zygiel EM, Lee CH, Oglesby AG, Nolan EM. Calprotectin-mediated survival of Staphylococcus aureus in coculture with Pseudomonas aeruginosa occurs without nutrient metal sequestration. mBio 2025; 16:e0384624. [PMID: 40152583 PMCID: PMC12077171 DOI: 10.1128/mbio.03846-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 02/05/2025] [Indexed: 03/29/2025] Open
Abstract
Pseudomonas aeruginosa and Staphylococcus aureus are bacterial pathogens of major clinical concern that cause polymicrobial infections in diverse patient populations. Human calprotectin (CP; S100A8/S100A9 heterooligomer, MRP8/MRP14 heterooligomer) is a host-defense protein that contributes to nutritional immunity by sequestering multiple nutrient metal ions including Mn(II), Fe(II), and Zn(II). Here, we examine the consequences of metal availability and CP treatment on cocultures of P. aeruginosa and S. aureus. We report that CP elicits Fe-starvation responses in both P. aeruginosa and S. aureus in coculture, including the upregulation of genes involved in Fe uptake by both organisms. Moreover, analyses of pseudomonal metabolites in coculture supernatants further demonstrate Fe-starvation responses, showing that CP treatment leads to increased siderophore levels and reduced phenazine levels. Consistent with prior studies, growth under conditions of Fe depletion accelerated P. aeruginosa killing of S. aureus in coculture, but treatment with CP promoted S. aureus survival. Treatment with CP site variants lacking functional transition-metal-binding sites and metalated CP also enhanced S. aureus survival in coculture with P. aeruginosa, revealing that this consequence of CP treatment is independent of its canonical metal-sequestering function. Thus, the protective effects of CP treatment during coculture appear to override the observed Fe-starvation effects that make P. aeruginosa more virulent toward S. aureus. This work highlights an unappreciated facet of how CP contributes to host-pathogen and pathogen-pathogen interactions that are relevant to human infectious disease. IMPORTANCE The current working model that describes how the innate immune protein calprotectin (CP) protects the host against bacterial pathogens focuses on its capacity to sequester multiple essential metal nutrients in a process called nutritional immunity. Our study further explores this function by focusing on the effects of metal availability and CP treatment on the dynamics of Pseudomonas aeruginosa and Staphylococcus aureus grown in coculture. These two bacterial pathogens are of significant clinical concern and colocalize with CP at infection sites. This work reveals that CP modulates P. aeruginosa/S. aureus coculture dynamics in a manner that is independent of its ability to sequester nutrient metal ions. This surprising result is important because it demonstrates that CP has metal-independent function and thus contributes to the host-pathogen and pathogen-pathogen interactions in ways that are not accounted for in the current working model focused on metal sequestration.
Collapse
Affiliation(s)
- Wei H. Lee
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Emily M. Zygiel
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Celis H. Lee
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Amanda G. Oglesby
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland, USA
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, Maryland, USA
| | - Elizabeth M. Nolan
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| |
Collapse
|
3
|
Liu Q, Wu Q, Liu J, Xu T, Liu J, Wu Q, Malakar PK, Zhu Y, Zhao Y, Zhang Z. New Insights into the Mediation of Biofilm Formation by Three Core Extracellular Polysaccharide Biosynthesis Pathways in Pseudomonas aeruginosa. Int J Mol Sci 2025; 26:3780. [PMID: 40332422 PMCID: PMC12027665 DOI: 10.3390/ijms26083780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2025] [Revised: 03/31/2025] [Accepted: 04/08/2025] [Indexed: 05/08/2025] Open
Abstract
Pseudomonas aeruginosa biofilms, driven by extracellular polysaccharides (EPSs), exacerbate pathogenicity and drug resistance, posing critical threats to public health. While EPS biosynthesis pathways are central to biofilm formation, their distinct contributions and regulatory dynamics remain incompletely understood. Here, we systematically dissect the roles of three core EPS pathways-Psl, Pel, and alginate-in biofilm architecture and function using multi-omics approaches. Key findings reveal Psl as the dominant regulator of biofilm elasticity and thickness, with its deletion disrupting chemotaxis, quorum sensing, and 3',5'-Cyclic GMP (c-di-GMP)/amino acid metabolism. Pel redundantly enhances biofilm biomass, but elevates flagellar synthesis efficiency when Psl is absent. Alginate exhibited negligible transcriptional or metabolic influence on biofilms. These insights clarify hierarchical EPS contributions and highlight Psl as a priority target for therapeutic strategies to dismantle biofilm-mediated resistance.
Collapse
Affiliation(s)
- Qianhui Liu
- College of Food Science and Technology, Shanghai Ocean University, 999# Hu Cheng Huan Road, Shanghai 201306, China; (Q.L.); (Q.W.); (J.L.); (T.X.); (J.L.); (Q.W.); (P.K.M.); (Y.Z.)
- International Research Center for Food and Health, Shanghai Ocean University, 999# Hu Cheng Huan Road, Shanghai 201306, China
| | - Qian Wu
- College of Food Science and Technology, Shanghai Ocean University, 999# Hu Cheng Huan Road, Shanghai 201306, China; (Q.L.); (Q.W.); (J.L.); (T.X.); (J.L.); (Q.W.); (P.K.M.); (Y.Z.)
- International Research Center for Food and Health, Shanghai Ocean University, 999# Hu Cheng Huan Road, Shanghai 201306, China
| | - Jiawen Liu
- College of Food Science and Technology, Shanghai Ocean University, 999# Hu Cheng Huan Road, Shanghai 201306, China; (Q.L.); (Q.W.); (J.L.); (T.X.); (J.L.); (Q.W.); (P.K.M.); (Y.Z.)
- International Research Center for Food and Health, Shanghai Ocean University, 999# Hu Cheng Huan Road, Shanghai 201306, China
| | - Tianming Xu
- College of Food Science and Technology, Shanghai Ocean University, 999# Hu Cheng Huan Road, Shanghai 201306, China; (Q.L.); (Q.W.); (J.L.); (T.X.); (J.L.); (Q.W.); (P.K.M.); (Y.Z.)
- International Research Center for Food and Health, Shanghai Ocean University, 999# Hu Cheng Huan Road, Shanghai 201306, China
| | - Jing Liu
- College of Food Science and Technology, Shanghai Ocean University, 999# Hu Cheng Huan Road, Shanghai 201306, China; (Q.L.); (Q.W.); (J.L.); (T.X.); (J.L.); (Q.W.); (P.K.M.); (Y.Z.)
- International Research Center for Food and Health, Shanghai Ocean University, 999# Hu Cheng Huan Road, Shanghai 201306, China
| | - Qin Wu
- College of Food Science and Technology, Shanghai Ocean University, 999# Hu Cheng Huan Road, Shanghai 201306, China; (Q.L.); (Q.W.); (J.L.); (T.X.); (J.L.); (Q.W.); (P.K.M.); (Y.Z.)
- International Research Center for Food and Health, Shanghai Ocean University, 999# Hu Cheng Huan Road, Shanghai 201306, China
| | - Pradeep K. Malakar
- College of Food Science and Technology, Shanghai Ocean University, 999# Hu Cheng Huan Road, Shanghai 201306, China; (Q.L.); (Q.W.); (J.L.); (T.X.); (J.L.); (Q.W.); (P.K.M.); (Y.Z.)
- International Research Center for Food and Health, Shanghai Ocean University, 999# Hu Cheng Huan Road, Shanghai 201306, China
| | - Yongheng Zhu
- College of Food Science and Technology, Shanghai Ocean University, 999# Hu Cheng Huan Road, Shanghai 201306, China; (Q.L.); (Q.W.); (J.L.); (T.X.); (J.L.); (Q.W.); (P.K.M.); (Y.Z.)
- International Research Center for Food and Health, Shanghai Ocean University, 999# Hu Cheng Huan Road, Shanghai 201306, China
| | - Yong Zhao
- College of Food Science and Technology, Shanghai Ocean University, 999# Hu Cheng Huan Road, Shanghai 201306, China; (Q.L.); (Q.W.); (J.L.); (T.X.); (J.L.); (Q.W.); (P.K.M.); (Y.Z.)
- International Research Center for Food and Health, Shanghai Ocean University, 999# Hu Cheng Huan Road, Shanghai 201306, China
- Laboratory of Quality & Safety Risk Assessment for Aquatic Products on Storage and Preservation (Shanghai), Ministry of Agriculture and Rural Affairs, 999# Hu Cheng Huan Road, Shanghai 201306, China
- Shanghai Engineering Research Center of Aquatic-Product Processing & Preservation, 999# Hu Cheng Huan Road, Shanghai 201306, China
| | - Zhaohuan Zhang
- College of Food Science and Technology, Shanghai Ocean University, 999# Hu Cheng Huan Road, Shanghai 201306, China; (Q.L.); (Q.W.); (J.L.); (T.X.); (J.L.); (Q.W.); (P.K.M.); (Y.Z.)
- International Research Center for Food and Health, Shanghai Ocean University, 999# Hu Cheng Huan Road, Shanghai 201306, China
- Laboratory of Quality & Safety Risk Assessment for Aquatic Products on Storage and Preservation (Shanghai), Ministry of Agriculture and Rural Affairs, 999# Hu Cheng Huan Road, Shanghai 201306, China
- Shanghai Engineering Research Center of Aquatic-Product Processing & Preservation, 999# Hu Cheng Huan Road, Shanghai 201306, China
| |
Collapse
|
4
|
Hanstein S, Grochow T, Mötzing M, Fietz SA, Hoffmann R, Baums CG, Kähl S. Comparative evaluation of antimicrobial peptides: effect on formation, metabolic activity and viability of Klebsiella pneumoniae biofilms. Front Microbiol 2025; 16:1548362. [PMID: 40291808 PMCID: PMC12021911 DOI: 10.3389/fmicb.2025.1548362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 03/19/2025] [Indexed: 04/30/2025] Open
Abstract
Introduction Klebsiella pneumoniae (K. pneumoniae) is a major human nosocomial infectious agent and an important veterinary pathogen, frequently resistant to various antibiotics. It causes diseases such as pneumonia, urinary tract infections, surgical wound infections and septicemia. Biofilm formation of K. pneumoniae promotes persistent infection and contributes to resistance against antimicrobial agents. The objective of this study was to comparatively evaluate the effect of selected AMPs on the formation, metabolic activity and viability of Klebsiella pneumoniae biofilms of veterinary and human origin. Methods Biofilm formation of three K. pneumoniae strains was quantified using the crystal violet assay and visualized by scanning electron microscopy (SEM). The inhibitory effects of eight different AMPs on the formation and metabolic activity of K. pneumoniae biofilms, as well as on planktonic growth, were examined using crystal violet, resazurin and broth microdilution assays, respectively. The effect on living and dead bacteria in mature biofilms was investigated using the fluorescent dyes SYTO™ 9 and propidium iodide. In addition, the distribution of rhodamine B-labeled peptide DJK-5 in mature biofilms of strain 17349 was visualized by confocal laser scanning microscopy (CLSM). Results Biofilm formation was confirmed for all three K. pneumoniae strains. Depending on the strain, we found that planktonic growth was affected by the AMPs DJK-5, DJK-6, Onc72, and Onc112. Biofilm formation of all three strains was inhibited by hbD3, LL-37, DJK-5, and DJK-6, with biofilm mass reduced to less than 40% of the untreated control. In addition to the inhibition of biofilm formation, a reduction in the metabolic activity of the biofilm-associated bacteria was also observed. These four AMPs also showed an effect on mature biofilms by reducing the number of both viable and dead bacteria in 22 h-old biofilms. Rhodamine B-labeled DJK-5 took 7 h to visibly accumulate in the planktonic bacteria. Multi-layered biofilm aggregations were mainly negative for rhodamine B-labeled DJK-5, even 44 h after AMP treatment, indicating that certain parts of mature K. pneumoniae biofilms are not accessible for this AMP. Conclusion In conclusion, we found differences in the effect of AMPs on biofilms including both increases and decreases in biofilm mass and viability.
Collapse
Affiliation(s)
- Sophia Hanstein
- Institute of Bacteriology and Mycology, Center for Infectious Diseases, Faculty of Veterinary Medicine, Leipzig University, Leipzig, Germany
| | - Thomas Grochow
- Institute of Veterinary Anatomy, Histology and Embryology, Faculty of Veterinary Medicine, Leipzig University, Leipzig, Germany
| | - Marina Mötzing
- Institute of Bioanalytical Chemistry, Faculty of Chemistry, Leipzig University, Leipzig, Germany
- Center for Biotechnology and Biomedicine, Leipzig University, Leipzig, Germany
| | - Simone A. Fietz
- Institute of Veterinary Anatomy, Histology and Embryology, Faculty of Veterinary Medicine, Leipzig University, Leipzig, Germany
| | - Ralf Hoffmann
- Institute of Bioanalytical Chemistry, Faculty of Chemistry, Leipzig University, Leipzig, Germany
- Center for Biotechnology and Biomedicine, Leipzig University, Leipzig, Germany
| | - Christoph G. Baums
- Institute of Bacteriology and Mycology, Center for Infectious Diseases, Faculty of Veterinary Medicine, Leipzig University, Leipzig, Germany
| | - Sophie Kähl
- Institute of Bacteriology and Mycology, Center for Infectious Diseases, Faculty of Veterinary Medicine, Leipzig University, Leipzig, Germany
| |
Collapse
|
5
|
Rizakos T, Geddes-McAlister J. Remodeling of the neutrophil proteome upon exposure to Pseudomonas aeruginosa biofilms. Microbiol Resour Announc 2025; 14:e0114724. [PMID: 40079579 PMCID: PMC11984127 DOI: 10.1128/mra.01147-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 01/17/2025] [Indexed: 03/15/2025] Open
Abstract
Neutrophils play important roles within the innate immune system as the first line of defense against invading pathogens. Pathogens counteract these defenses through multiple mechanisms, including protective biofilm formation. Herein, we assessed proteome remodeling between Pseudomonas aeruginosa biofilms and HL-60 cells to provide new insights into host defense responses.
Collapse
Affiliation(s)
- Tia Rizakos
- Molecular and Cellular Biology Department, University Of Guelph, Guelph, Ontario, Canada
| | | |
Collapse
|
6
|
de Sousa T, Silva C, Igrejas G, Hébraud M, Poeta P. The Interactive Dynamics of Pseudomonas aeruginosa in Global Ecology. J Basic Microbiol 2025; 65:e70004. [PMID: 39972634 DOI: 10.1002/jobm.70004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 01/20/2025] [Accepted: 02/03/2025] [Indexed: 02/21/2025]
Abstract
Pseudomonas aeruginosa is an opportunistic bacterium widely distributed in both natural and urban environments, playing a crucial role in global microbial ecology. This article reviews the interactive dynamics of P. aeruginosa across different ecosystems, highlighting its capacity for adaptation and resistance in response to environmental and therapeutic pressures. We analyze the mechanisms of antibiotic resistance, including the presence of resistance genes and efflux systems, which contribute to its persistence in both clinical and nonclinical settings. The interconnection between human, animal, and environmental health, within the context of the One Health concept, is discussed, emphasizing the importance of monitoring and sustainable management practices to mitigate the spread of resistance. Through a holistic approach, this work offers insights into the influence of P. aeruginosa on public health and biodiversity.
Collapse
Affiliation(s)
- Telma de Sousa
- Department of Genetics and Biotechnology, University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal
- Functional Genomics and Proteomics Unit, University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal
- LAQV-REQUIMTE, Faculty of Sciences and Technology, Universidade NOVA de Lisboa, Caparica, Portugal
- Microbiology and Antibiotic Resistance Team (MicroART), Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal
| | - Catarina Silva
- Microbiology and Antibiotic Resistance Team (MicroART), Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal
| | - Gilberto Igrejas
- Department of Genetics and Biotechnology, University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal
- Functional Genomics and Proteomics Unit, University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal
- LAQV-REQUIMTE, Faculty of Sciences and Technology, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Michel Hébraud
- INRAE, Université Clermont Auvergne, UMR Microbiologie Environnement Digestif Santé (MEDiS), Saint-Genès-Champanelle, France
| | - Patrícia Poeta
- LAQV-REQUIMTE, Faculty of Sciences and Technology, Universidade NOVA de Lisboa, Caparica, Portugal
- Microbiology and Antibiotic Resistance Team (MicroART), Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal
- CECAV-Veterinary and Animal Research Center, University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), University of Lisboa, Lisboa, Portugal
| |
Collapse
|
7
|
Palková Z, Váchová L. Cell differentiation, aging, and death in spatially organized yeast communities: mechanisms and consequences. Cell Death Differ 2025:10.1038/s41418-025-01485-9. [PMID: 40158069 DOI: 10.1038/s41418-025-01485-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 02/12/2025] [Accepted: 03/17/2025] [Indexed: 04/01/2025] Open
Abstract
Cell death is a natural part of the development of multicellular organisms and is central to their physiological and pathological states. However, the existence of regulated cell death in unicellular microorganisms, including eukaryotic and prokaryotic microbes, has been a topic of debate. One reason for the continued debate is the lack of obvious benefit from cell death in the context of a single cell. However, unicellularity is relative, as most of these microbes dwell in communities of varying complexities, often with complicated spatial organization. In these spatially organized microbial communities, such as yeast and bacterial colonies and biofilms growing on solid surfaces, cells differentiate into specialized types, and the whole community often behaves like a simple multicellular organism. As these communities develop and age, cell death appears to offer benefits to the community as a whole. This review explores the potential roles of cell death in spatially organized communities of yeasts and draws analogies to similar communities of bacteria. The natural dying processes in microbial cell communities are only partially understood and may result from suicidal death genes, (self-)sabotage (without death effectors), or from non-autonomous mechanisms driven by interactions with other differentiated cells. We focus on processes occurring during the stratification of yeast colonies, the formation of the extracellular matrix in biofilms, and discuss potential roles of cell death in shaping the organization, differentiation, and overall physiology of these microbial structures.
Collapse
Affiliation(s)
- Zdena Palková
- Department of Genetics and Microbiology, Faculty of Science, BIOCEV, Charles University, Prague, Czech Republic.
| | - Libuše Váchová
- Department of Genetics and Microbiology, Faculty of Science, BIOCEV, Charles University, Prague, Czech Republic
- Institute of Microbiology of the Czech Academy of Sciences, BIOCEV, Prague, Czech Republic
| |
Collapse
|
8
|
Reichhardt C, Matwichuk ML, Lewerke LT, Jacobs HM, Yan J, Parsek MR. Non-disruptive matrix turnover is a conserved feature of biofilm aggregate growth in paradigm pathogenic species. mBio 2025; 16:e0393524. [PMID: 39982068 PMCID: PMC11898600 DOI: 10.1128/mbio.03935-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 01/23/2025] [Indexed: 02/22/2025] Open
Abstract
Bacteria form multicellular aggregates called biofilms. A crucial component of these aggregates is a protective matrix that holds the community together. Biofilm matrix composition varies depending upon bacterial species but typically includes exopolysaccharides (EPS), proteins, and extracellular DNA. Pseudomonas aeruginosa is a model organism for the study of biofilms, and in non-mucoid biofilms, it uses the structurally distinct EPS Psl and Pel, the EPS-binding protein CdrA, and eDNA as key matrix components. An interesting phenomenon that we and others have observed is that the periphery of a biofilm aggregate can be EPS-rich and contain very few cells. In this study, we investigated two possible models of assembly and dynamics of this EPS-rich peripheral region: (i) newly synthesized EPS is inserted and incorporated into the existing EPS-rich region at the periphery during biofilm aggregate growth or (ii) EPS is continuously turned over and newly synthesized EPS is deposited at the outermost edge of the aggregate. Our results support the latter model. Specifically, we observed that new EPS is continually deposited at the aggregate periphery, which is necessary for continued aggregate growth but not aggregate stability. We made similar observations in another paradigm biofilm-forming species, Vibrio cholerae. This pattern of deposition raises the question of how EPS is retained. Specifically, for P. aeruginosa biofilms, the matrix adhesin CdrA is thought to retain EPS. However, current thinking is that cell-associated CdrA is responsible for this retention, and it is not clear how CdrA might function in the relatively cell-free aggregate periphery. We observed that CdrA is enzymatically degraded during aggregate growth without negatively impacting biofilm stability and that cell-free CdrA can partially maintain aggregation and Psl retention. Overall, this study shows that the matrix of P. aeruginosa biofilms undergoes both continuous synthesis of matrix material and matrix turnover to accommodate biofilm aggregate growth and that cell-free matrix can at least partially maintain biofilm aggregation and EPS localization. Furthermore, our similar observations for V. cholerae biofilms suggest that our findings may represent basic principles of aggregate assembly in bacteria. IMPORTANCE Here, we show that, to accommodate growing cellular biomass, newly produced Psl is deposited over existing Psl at the periphery of biofilm aggregates. We demonstrated that V. cholerae employs a similar mechanism with its biofilm matrix EPS, VPS. In addition, we found that the protease LasB is present in the biofilm matrix, resulting in degradation of CdrA to lower molecular weight cell-free forms. We then show that the released forms of CdrA are retained in the matrix and remain functional. Together, our findings support that the P. aeruginosa biofilm matrix is dynamic during the course of aggregate growth and that other species may employ similar mechanisms to remodel their matrix.
Collapse
Affiliation(s)
- Courtney Reichhardt
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| | | | - Lincoln T. Lewerke
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| | - Holly M. Jacobs
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| | - Jing Yan
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, Connecticut, USA
| | - Matthew R. Parsek
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
9
|
Guo M, Ling X, He L, Gou Y, Li Z, Li W. NapR Regulates the Expression of Phosphoserine Aminotransferase SerC to Modulate Biofilm Formation and Resistance to Serine Stress of Mycobacteria. Int J Mol Sci 2025; 26:2181. [PMID: 40076802 PMCID: PMC11899882 DOI: 10.3390/ijms26052181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 02/26/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
Mycobacterium tuberculosis is a formidable pathogen capable of establishing persistent infections within macrophages. To survive and thrive within the host environment, it has evolved intricate regulatory networks, including a diverse array of transcription factors that enable adaptation to various stresses encountered within the host. However, the mechanisms by which transcription factors regulate biofilm formation in M. tuberculosis remain incompletely understood. This study aimed to investigate the role of serC, encoding phosphoserine aminotransferase, and its regulation by NapR, a transcription factor, in mycobacterial physiology. NapR regulates serC through directly binding to its promoter. Notably, the regulatory effect and corresponding phenotypes vary due to distinct binding affinities of NapR for the serC promoter in different mycobacterial species. In Mycobacterium smegmatis, NapRMsm positively regulates biofilm formation, growth on solid media, and the transition from microcolonies to microcolonies by activating serCMsm. In the BCG vaccine, on the contrary, NapRBCG represses serCBCG, thus negatively regulating colony size and alleviating the growth inhibition caused by high concentrations of serine. Furthermore, proteomic analysis suggested NapR serves as a global transcriptional regulator in BCG vaccine strains by simultaneously modulating four metabolic pathways. These findings underscore the complex and strain-specific regulatory mechanisms governing serine metabolism in mycobacteria and provide valuable insights into the interplay between metabolism, gene regulation, and bacterial physiology.
Collapse
Affiliation(s)
| | | | | | | | | | - Weihui Li
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Life Science and Technology, Guangxi University, Nanning 530004, China
| |
Collapse
|
10
|
Ebrahim H, Haldenby S, Moore M, Dashti A, Floyd R, Fothergill J. Genotypic and phenotypic analyses of two distinct sets of Pseudomonas aeruginosa urinary tract isolates. J Med Microbiol 2025; 74:001971. [PMID: 40013918 PMCID: PMC11868659 DOI: 10.1099/jmm.0.001971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 01/18/2025] [Indexed: 02/28/2025] Open
Abstract
Introduction. Urinary tract infections (UTIs) are associated with a high burden of morbidity, mortality and cost. Pseudomonas aeruginosa employs a myriad of virulence factors, including biofilm formation and motility mechanisms, to cause infections including persistent UTIs. P. aeruginosa is highly resistant to antibiotics, and the World Health Organization has identified it as a pathogen for which novel antimicrobials are urgently required.Gap statement. Genotypic and phenotypic characterization of P. aeruginosa from UTIs is underreported. In addition, the rise of antimicrobial resistance (AMR) is a cause for concern, particularly in many countries where surveillance is severely lacking.Aim. To identify genotypic and phenotypic characteristics of P. aeruginosa UTI isolates sourced from the UK and the state of Kuwait, with an emphasis on genotypic diversity and AMR.Methods. Twenty-three P. aeruginosa UTI isolates were sourced from the UK and Kuwait. To establish the phenotypes of UK isolates, growth analysis, biofilm formation assays, motility assays and antibiotic disc diffusion assays were performed. Whole-genome sequencing, antimicrobial susceptibility assays and in silico detection of AMR-associated genes were conducted on both sets of isolates.Results. In terms of their phenotypic characteristics and genomic composition, the UTI isolates varied. Multiple resistance genes are associated with resistance to various classes of antibiotics, such as aminoglycosides, fluoroquinolones and β-lactams, particularly in isolates from Kuwait. Extreme antibiotic resistance was detected in the isolates obtained from Kuwait, indicating that the country may be an antibiotic resistance hotspot.Conclusion. This study highlights that isolates from UTIs are diverse and can display extremely high resistance. Surveillance in countries such as Kuwait is currently limited, and this study suggests the need for greater surveillance.
Collapse
Affiliation(s)
- H. Ebrahim
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
- School of Arts and Sciences, American International University, Kuwait
| | - S. Haldenby
- Centre for Genomic Research, Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| | - M.P. Moore
- School of Life Sciences, University of Warwick, Coventry, UK
| | - A.A. Dashti
- Department of Medical Laboratory Sciences, Faculty of Allied Health Science, Health Sciences Centre, Kuwait University, Kuwait
| | - R.V. Floyd
- School of Life Sciences, University of Liverpool, Liverpool, UK
| | - J.L. Fothergill
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| |
Collapse
|
11
|
Xue Y, Kang X. Time-resolved compositional and dynamics analysis of biofilm maturation and dispersal via solid-state NMR spectroscopy. NPJ Biofilms Microbiomes 2025; 11:21. [PMID: 39880834 PMCID: PMC11779841 DOI: 10.1038/s41522-025-00655-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 01/20/2025] [Indexed: 01/31/2025] Open
Abstract
Dispersal plays a crucial role in the development and ecology of biofilms. While extensive studies focused on elucidating the molecular mechanisms governing this process, few have characterized the associated temporal changes in composition and structure. Here, we employed solid-state nuclear magnetic resonance (NMR) techniques to achieve time-resolved characterization of Bacillus subtilis biofilms over a 5-day period. The mature biofilm, established within 48 h, undergoes significant degradation in following 72 h. The steepest decline of proteins precedes that of exopolysaccharides, likely reflecting their distinct spatial distribution. Exopolysaccharide sugar units display clustered temporal patterns, suggesting the presence of distinct polysaccharide types. A sharp rise in aliphatic carbon signals on day 4 probably corresponds to a surge in biosurfactant production. Different dynamic regimes respond differently to dispersal: the mobile domain exhibits increased rigidity, while the rigid domain remains stable. These findings provide novel insights and perspectives on the complex process of biofilm dispersal.
Collapse
Affiliation(s)
- Yi Xue
- Institute of Drug Discovery Technology, Ningbo University, Ningbo, 315211, Zhejiang, China
| | - Xue Kang
- Institute of Drug Discovery Technology, Ningbo University, Ningbo, 315211, Zhejiang, China.
| |
Collapse
|
12
|
Zellner AA, Wirtz DC, Schildberg FA. In Vitro Efficacy of Phage Therapy Against Common Biofilm-forming Pathogens in Orthopedics and Trauma Surgery. ZEITSCHRIFT FUR ORTHOPADIE UND UNFALLCHIRURGIE 2025. [PMID: 39832775 DOI: 10.1055/a-2436-7394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Formation of biofilms by bacteria is a major challenge in a clinical setting. The importance of these biofilms increases in specialties where foreign bodies and prosthetic material are used. Orthopaedics is such a speciality and phage therapy could offer additional therapeutic options when dealing with biofilm infections.We conducted a systematic literature review using the PubMed database. We searched for phage activity against biofilms of the most common pathogens found in orthopaedics.The results of the systematic review were broken down into different categories and discussed accordingly. We concentrated on the time the biofilms were allowed to mature, and the surface they were grown on. In addition, we checked the efficacy of bacteriophages compared to antibiotics and when applied simultaneously with antibiotics. We also investigated the source of the phages, how they were tested for sensibility against the biofilms, as well the conditions (pH, temperature) under which they remained active and stable.The data suggests that the in vitro efficacy of phages does not change under a wide spectrum of temperature and pH. To further explore the use of bacteriophages in orthopaedics, we need further studies that test biofilms which matured for several weeks on surfaces that are common in arthroplasty and traumatology.
Collapse
Affiliation(s)
- Alberto Alfieri Zellner
- Klinik und Poliklinik für Orthopädie und Unfallchirurgie, Universitätsklinikum Bonn, Bonn, Deutschland
| | - Dieter Christian Wirtz
- Klinik und Poliklinik für Orthopädie und Unfallchirurgie, Universitätsklinikum Bonn, Bonn, Deutschland
| | | |
Collapse
|
13
|
Zhao T, Lei F, Zhang Z, Wang D, Ma LZ. Extracellular aminopeptidase regulates exopolysaccharide production of Pseudomonas aeruginosa via quorum sensing. THE ISME JOURNAL 2025; 19:wraf038. [PMID: 40037286 PMCID: PMC12034520 DOI: 10.1093/ismejo/wraf038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 02/15/2025] [Accepted: 02/25/2025] [Indexed: 03/06/2025]
Abstract
The biofilm matrix primarily consists of proteins, exopolysaccharides, and extracellular DNA. Pseudomonas aeruginosa aminopeptidase is one of the most abundant matrix proteins in P. aeruginosa biofilms and plays a crucial role in modulating biofilm development. In a previous study, we have revealed that the loss of aminopeptidase enhances the attachment ability of P. aeruginosa. However, the mechanism by which aminopeptidase affects attachment remains unclear. In this study, we demonstrate that aminopeptidase is the primary protein associated with the matrix exopolysaccharide Psl. The loss of aminopeptidase leads to increased production of Psl, resulting in enhanced attachment of P. aeruginosa. Further investigation shows that aminopeptidase represses the transcription of the psl operon through the LasI/LasR quorum-sensing system, rather than via other known psl regulators or the cyclic-di-GMP signaling molecule. Aminopeptidase inhibits the transcription of lasI via the short peptides cleaved from the proform of aminopeptidase during its activation, which results in reduced biosynthesis of the quorum-sensing signaling molecule, N-(3-oxododecanoyl)-L-homoserine lactone, further decreasing the production of Psl. In conclusion, our study reveals an interplay between two key matrix components via quorum-sensing signal, suggesting a mechanism by which bacteria control initial attachment and exopolysaccharide production in response to cell density.
Collapse
Affiliation(s)
- Tianhu Zhao
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
- College of Life Sciences, University of Chinese Academy of Sciences, No. 1 Yanqihu East Rd, Huairou District, Beijing 101408, PR China
| | - Fanglin Lei
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
- School of Life Sciences, Yunnan University, Kunming 650091, China
| | - Zhenyu Zhang
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
- College of Life Sciences, University of Chinese Academy of Sciences, No. 1 Yanqihu East Rd, Huairou District, Beijing 101408, PR China
| | - Di Wang
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Luyan Z Ma
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
- Medical School, University of Chinese Academy of Sciences, No. 1 Yanqihu East Rd, Huairou District, Beijing 101408, PR China
- College of Life Sciences, University of Chinese Academy of Sciences, No. 1 Yanqihu East Rd, Huairou District, Beijing 101408, PR China
| |
Collapse
|
14
|
Kalia M, Sauer K. Distinct transcriptome and traits of freshly dispersed Pseudomonas aeruginosa cells. mSphere 2024; 9:e0088424. [PMID: 39601567 DOI: 10.1128/msphere.00884-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 11/04/2024] [Indexed: 11/29/2024] Open
Abstract
Bacteria assume two distinct lifestyles: the planktonic and biofilm modes of growth. Additionally, dispersion has emerged as a third phenotype, accompanied by the distinct phenotypes and the unique expression of >600 genes. Here, we asked whether the distinct phenotype of dispersed cells is already apparent within minutes of egressing from the biofilm. We used RNA-seq to show that the physiology of freshly dispersed cells from Pseudomonas aeruginosa biofilms is highly different from those of planktonic and biofilm cells, apparent by dispersed cells uniquely expressing 194 genes. Unique and differentially expressed genes relative to planktonic or biofilm cells include genes associated with type IV pili, pyoverdine, type III and type VI secretion systems, and antibiotic resistance that are downregulated in dispersed cells, whereas the transcript abundance of genes involved in swimming motility, Hxc type II secretion system and various other virulence factors, and metabolic and energy-generating pathways are increased, indicative of dispersion coinciding with an awakening and re-energizing of dispersed cells, and a switch in virulence, further apparent by freshly dispersed cells significantly subverting engulfment by macrophages. The findings suggest that dispersed cells display a distinct phenotype within minutes of egressing from the biofilm, with freshly dispersed cells already capable of efficiently evading phagocytosis. IMPORTANCE Dispersion is considered a transitionary phenotype, enabling bacteria to switch between the communal, biofilm lifestyle, where cells share resources and are protected from harmful conditions to the planktonic state. Here, we demonstrate that within minutes of leaving the biofilm, dispersed cells express genes and display phenotypic traits that are distinct from biofilms and planktonic cells. Our findings suggest that dispersed cells quickly adapt to a less structured and protected but more nutrient-rich environment, with this trade-off in environment coinciding with an awakening and a switch in virulence, specifically a switch from directly intoxicating host cells and potential competitors toward more broadly active virulence factors and strategies of evasion. To our knowledge, this is the first report of dispersed cells' distinct (trade-off) phenotype and their enhanced resilience so soon after egressing from the biofilm.
Collapse
Affiliation(s)
- Manmohit Kalia
- Department of Biological Sciences, Binghamton University, Binghamton, New York, USA
- Binghamton Biofilm Research Center, Binghamton University, Binghamton, New York, USA
| | - Karin Sauer
- Department of Biological Sciences, Binghamton University, Binghamton, New York, USA
- Binghamton Biofilm Research Center, Binghamton University, Binghamton, New York, USA
| |
Collapse
|
15
|
Salim A, Sathishkumar P. Therapeutic efficacy of chitosan-based hybrid nanomaterials to treat microbial biofilms and their infections - A review. Int J Biol Macromol 2024; 283:137850. [PMID: 39577523 DOI: 10.1016/j.ijbiomac.2024.137850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 11/11/2024] [Accepted: 11/17/2024] [Indexed: 11/24/2024]
Abstract
Antimicrobial resistance, the biggest issue facing the global healthcare sector, quickly emerged and spread due to the frequent use of antibiotics in regular treatments. The investigation of polymer-based nanomaterials as possible antibiofilm treatment agents is prompted by the growing ineffectiveness of conventional therapeutic techniques against these resistant bacteria species. So far, several articles have been published on microbial biofilm eradication using various polymer-based nanomaterials due to their therapeutic efficacy and biocompatibility nature. Despite their potential, a comprehensive review of the chitosan-based hybrid nanomaterials to treat microbial biofilms and their infections is lacking. This review provides a comprehensive investigation of the current state of therapeutic efficacy, various nanoformulations, advantages, limitations, and regulations of chitosan-based hybrid nanomaterials for biofilm treatment. Special attention is given to the application of chitosan-based nanomaterials in wound care, urinary tract infections, and dental biofilms are discussed, highlighting their role in managing biofilm-associated complications. Researchers will be better able to comprehend and develop unique, marketable chitosan-based nanomaterials with increased activity to treat biofilm infections in near future with the aid of this review.
Collapse
Affiliation(s)
- Anisha Salim
- Green Lab, Department of Prosthodontics, Saveetha Dental College and Hospital, SIMATS, Saveetha University, Chennai 600 077, Tamil Nadu, India
| | - Palanivel Sathishkumar
- Green Lab, Department of Prosthodontics, Saveetha Dental College and Hospital, SIMATS, Saveetha University, Chennai 600 077, Tamil Nadu, India.
| |
Collapse
|
16
|
Graham CL, Bryant J, Roper DI, Banzhaf M. Membrane staining and phospholipid tracking in Pseudomonas aeruginosa PAO1 using the phosphatidylcholine mimic propargyl-choline. Access Microbiol 2024; 6:000690.v3. [PMID: 39610693 PMCID: PMC11604180 DOI: 10.1099/acmi.0.000690.v3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 11/04/2024] [Indexed: 11/30/2024] Open
Abstract
The use of membrane-specific dyes for in vivo fluorescent microscopy is commonplace. However, most of these reagents are non-specific and cannot track specific lipid species movement, instead often acting as non-covalent lipid-associated probes or requiring the uptake of whole lipids and acyl tails into the membrane. This issue has been solved in eukaryotic cell biology by the use of click-chemistry-liable phospholipid headgroup pulse labels. Here, we describe a method for in vivo phospholipid labelling by fluorescent imaging in Pseudomonas aeruginosa using a phosphatidylcholine mimic, 'propargyl-choline' (PCho). This click-chemistry-liable headgroup mimic is visible by microscopy and allows the covalent labelling of lipids. Fluorescence of the cell membranes, visible in heterogeneous patches, is dependent on PCho concentration and is localized in the membrane fraction of cells, demonstrating that it is suitable for membrane labelling and cell imaging.
Collapse
Affiliation(s)
| | - Jack Bryant
- School of Biosciences, University of Birmingham, Birmingham, UK
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | - David I. Roper
- School of Life Sciences, University of Warwick, Coventry, UK
| | - Manuel Banzhaf
- School of Biosciences, University of Birmingham, Birmingham, UK
- Newcastle University Biosciences Institute, Newcastle University, Newcastle, UK
| |
Collapse
|
17
|
Squyres GR, Newman DK. Real-time high-resolution microscopy reveals how single-cell lysis shapes biofilm matrix morphogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.13.618105. [PMID: 39463994 PMCID: PMC11507769 DOI: 10.1101/2024.10.13.618105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
During development, multiscale patterning requires that cells organize their behavior in space and time. Bacteria in biofilms must similarly dynamically pattern their behavior with a simpler toolkit. Like in eukaryotes, morphogenesis of the extracellular matrix is essential for biofilm development, but how it is patterned has remained unclear. Here, we explain how the architecture of eDNA, a key matrix component, is controlled by single cell lysis events during Pseudomonas aeruginosa biofilm development. We extend single-cell imaging methods to capture complete biofilm development, characterizing the stages of biofilm development and visualizing eDNA matrix morphogenesis. Mapping the spatiotemporal distribution of single cell lysis events reveals that cell lysis is restricted to a specific biofilm zone. Simulations indicate that this patterning couples cell lysis to growth, more uniformly distributing eDNA throughout the biofilm. Finally, we find that patterning of cell lysis is organized by nutrient gradients that act as positioning cues.
Collapse
Affiliation(s)
- Georgia R. Squyres
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125
| | - Dianne K. Newman
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125
- Division of Geological and Planetary Sciences, California Institute of Technology, Pasadena, CA 91125
| |
Collapse
|
18
|
Chen KZM, Vu LM, Vollmer AC. Cultivation in long-term simulated microgravity is detrimental to pyocyanin production and subsequent biofilm formation ability of Pseudomonas aeruginosa. Microbiol Spectr 2024; 12:e0021124. [PMID: 39162544 PMCID: PMC11448113 DOI: 10.1128/spectrum.00211-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 06/27/2024] [Indexed: 08/21/2024] Open
Abstract
Pseudomonas aeruginosa forms aggregates known as biofilms. Previous studies have shown that when P. aeruginosa is cultivated in space, thicker and structurally different biofilms are formed than from those grown on Earth. We investigated how microgravity, simulated in a laboratory setting, influenced the growth, colonization, and virulence potentials of a P. aeruginosa PA14 wild-type strain, as well as two surface attachment-defective (sad) mutants altered at crucial biofilm-forming steps: flgK and pelA. Using high-aspect ratio rotating-wall vessel (HARV) bioreactors, P. aeruginosa bacteria were grown to stationary phase under prolonged (6 days) exposure to simulated microgravity or normal gravity conditions. After the exposure, the capacity of the culture to form biofilms was measured. Additionally, pigment (pyocyanin) formed by each culture during the incubation was extracted and quantified. We demonstrate that the first prolonged exposure to low-shear modeled microgravity (LSMMG) and without nutrient replenishment significantly diminishes wild-type P. aeruginosa PA14 biofilm formation abilities after exposure and pyocyanin production during exposure, while the mutant strains exhibit differing outcomes for both properties. IMPORTANCE Given plans for humans to engage in prolonged space travel, we investigated biofilm and pigment/virulence factor formation in Pseudomonas aeruginosa when cultivated in microgravity. These bacteria are opportunistic pathogens in immunocompromised individuals. Previous studies of space travelers have shown some immune system diminutions. Hence, our studies shed some light on how prolonged cultivation of bacteria in simulated microgravity conditions affect their growth characteristics.
Collapse
Affiliation(s)
| | - Linda My Vu
- Department of Biology, Swarthmore College, Swarthmore, Pennsylvania, USA
- Department of Microbial Pathogenesis, University of Maryland—Baltimore, Baltimore, Maryland, USA
| | - Amy Cheng Vollmer
- Department of Biology, Swarthmore College, Swarthmore, Pennsylvania, USA
| |
Collapse
|
19
|
da Cruz Nizer WS, Allison KN, Adams ME, Vargas MA, Ahmed D, Beaulieu C, Raju D, Cassol E, Howell PL, Overhage J. The role of exopolysaccharides Psl and Pel in resistance of Pseudomonas aeruginosa to the oxidative stressors sodium hypochlorite and hydrogen peroxide. Microbiol Spectr 2024; 12:e0092224. [PMID: 39194290 PMCID: PMC11448232 DOI: 10.1128/spectrum.00922-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 07/01/2024] [Indexed: 08/29/2024] Open
Abstract
Pseudomonas aeruginosa is well-known for its antimicrobial resistance and the ability to survive in harsh environmental conditions due to an abundance of resistance mechanisms, including the formation of biofilms and the production of exopolysaccharides. Exopolysaccharides are among the major components of the extracellular matrix in biofilms and aggregates of P. aeruginosa. Although their contribution to antibiotic resistance has been previously shown, their roles in resistance to oxidative stressors remain largely elusive. Here, we studied the function of the exopolysaccharides Psl and Pel in the resistance of P. aeruginosa to the commonly used disinfectants and strong oxidizing agents NaOCl and H2O2. We observed that the simultaneous inactivation of Psl and Pel in P. aeruginosa PAO1 mutant strain ∆pslA pelF resulted in a significant increase in susceptibility to both NaOCl and H2O2. Further analyses revealed that Pel is more important for oxidative stress resistance in P. aeruginosa and that the form of Pel (i.e., cell-associated or cell-free) did not affect NaOCl susceptibility. Additionally, we show that Psl/Pel-negative strains are protected against oxidative stress in co-culture biofilms with P. aeruginosa PAO1 WT. Taken together, our results demonstrate that the EPS matrix and, more specifically, Pel exhibit protective functions against oxidative stressors such as NaOCl and H2O2 in P. aeruginosa. IMPORTANCE Biofilms are microbial communities of cells embedded in a self-produced polymeric matrix composed of polysaccharides, proteins, lipids, and extracellular DNA. Biofilm bacteria have been shown to possess unique characteristics, including increased stress resistance and higher antimicrobial tolerance, leading to failures in bacterial eradication during chronic infections or in technical settings, including drinking and wastewater industries. Previous studies have shown that in addition to conferring structure and stability to biofilms, the polysaccharides Psl and Pel are also involved in antibiotic resistance. This work provides evidence that these biofilm matrix components also contribute to the resistance of Pseudomonas aeruginosa to oxidative stressors including the widely used disinfectant NaOCl. Understanding the mechanisms by which bacteria escape antimicrobial agents, including strong oxidants, is urgently needed in the fight against antimicrobial resistance and will help in developing new strategies to eliminate resistant strains in any environmental, industrial, and clinical setting.
Collapse
Affiliation(s)
| | - Kira N. Allison
- Department of Health Sciences, Carleton University, Ottawa, Ontario, Canada
| | - Madison E. Adams
- Department of Health Sciences, Carleton University, Ottawa, Ontario, Canada
| | - Mario A. Vargas
- Program in Medicine, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Duale Ahmed
- Department of Health Sciences, Carleton University, Ottawa, Ontario, Canada
| | - Carole Beaulieu
- Department of Health Sciences, Carleton University, Ottawa, Ontario, Canada
| | - Deepa Raju
- Program in Medicine, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Edana Cassol
- Department of Health Sciences, Carleton University, Ottawa, Ontario, Canada
| | - P. Lynne Howell
- Program in Medicine, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Joerg Overhage
- Department of Health Sciences, Carleton University, Ottawa, Ontario, Canada
| |
Collapse
|
20
|
Ren Y, You X, Zhu R, Li D, Wang C, He Z, Hu Y, Li Y, Liu X, Li Y. Mutation of Pseudomonas aeruginosa lasI/rhlI diminishes its cytotoxicity, oxidative stress, inflammation, and apoptosis on THP-1 macrophages. Microbiol Spectr 2024; 12:e0414623. [PMID: 39162513 PMCID: PMC11448257 DOI: 10.1128/spectrum.04146-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 06/27/2024] [Indexed: 08/21/2024] Open
Abstract
The management of Pseudomonas aeruginosa (P. aeruginosa) infections presents a substantial challenge to clinics and public health, emphasizing the urgent need for innovative strategies to address this issue. Quorum sensing (QS) is an intercellular communication mechanism that coordinates bacterial activities involved in various virulence mechanisms, such as acquiring host nutrients, facilitating biofilm formation, enhancing motility, secreting virulence factors, and evading host immune responses, all of which play a crucial role in the colonization and infection of P. aeruginosa. The LasI/R and RhlI/R sub-systems dominate in the QS system of P. aeruginosa. Macrophages play a pivotal role in the host's innate immune response to P. aeruginosa invasion, particularly through phagocytosis as the initial host defense mechanism. This study investigated the effects of P. aeruginosa's QS system on THP-1 macrophages. Mutants of PAO1 with lasI/rhlI deletion, as well as their corresponding complemented strains, were obtained, and significant downregulation of QS-related genes was observed in the mutants. Furthermore, the ΔlasI and ΔlasIΔrhlI mutants exhibited significantly attenuated virulence in terms of biofilm formation, extracellular polymeric substances synthesis, bacterial adhesion, motility, and virulence factors production. When infected with ΔlasI and ΔlasIΔrhlI mutants, THP-1 macrophages exhibited enhanced scavenging ability against the mutants and demonstrated resistance to cytotoxicity, oxidative stress, inflammatory response, and apoptosis induced by the culture supernatants of these mutant strains. These findings offer novel insights into the mechanisms underlying how the lasI/rhlI mutation attenuates cytotoxicity, oxidative stress, inflammation, and apoptosis in macrophages induced by P. aeruginosa.IMPORTANCEP. aeruginosa is classified as one of the ESKAPE pathogens and poses a global public health concern. The QS system of this versatile pathogen contributes to a broad spectrum of virulence, thereby constraining therapeutic options for serious infections. This study illustrated that the lasI/rhlI mutation of the QS system plays a prominent role in attenuating the virulence of P. aeruginosa by affecting bacterial adhesion, biofilm formation, extracellular polymeric substances synthesis, bacterial motility, and virulence factors' production. Notably, THP-1 macrophages infected with mutant strains exhibited increased phagocytic activity in eliminating intracellular bacteria and enhanced resistance to cytotoxicity, oxidative stress, inflammation, and apoptosis. These findings suggest that targeted intervention toward the QS system is anticipated to diminish the pathogenicity of P. aeruginosa to THP-1 macrophages.
Collapse
Affiliation(s)
- Yanying Ren
- Dazhou integrated Traditional Chinese Medicine & Western Medicine Hospital, Dazhou Second People's Hospital, Dazhou, China
- Henan University of Chinese Medicine, Zhengzhou, China
| | - Xiaojuan You
- Henan University of Chinese Medicine, Zhengzhou, China
- Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Chinese Medicine, Zhenghzhou, China
| | - Rui Zhu
- Henan University of Chinese Medicine, Zhengzhou, China
- Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Chinese Medicine, Zhenghzhou, China
| | - Dengzhou Li
- Henan University of Chinese Medicine, Zhengzhou, China
- Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Chinese Medicine, Zhenghzhou, China
| | - Chunxia Wang
- Henan University of Chinese Medicine, Zhengzhou, China
- Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Chinese Medicine, Zhenghzhou, China
| | - Zhiqiang He
- Henan University of Chinese Medicine, Zhengzhou, China
- Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Chinese Medicine, Zhenghzhou, China
| | - Yue Hu
- Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Chinese Medicine, Zhenghzhou, China
| | - Yifan Li
- Henan University of Chinese Medicine, Zhengzhou, China
| | - Xinwei Liu
- Henan University of Chinese Medicine, Zhengzhou, China
- Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Chinese Medicine, Zhenghzhou, China
| | - Yongwei Li
- Henan University of Chinese Medicine, Zhengzhou, China
- Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Chinese Medicine, Zhenghzhou, China
- The Key Laboratory of Pathogenic Microbes &Antimicrobial Resistance Surveillance of Zhengzhou, Zhengzhou, China
- Henan Engineering Research Center for Identification of Pathogenic Microbes, Zhengzhou, China
- Henan Provincial Key Laboratory of Antibiotics-Resistant Bacterial Infection Prevention & Therapy with Traditional Chinese Medicine, Zhengzhou, China
| |
Collapse
|
21
|
Rimal B, Chang JD, Liu C, Kim H, Aderotoye O, Zechmann B, Kim SJ. Scanning Electron Microscopy and Energy-Dispersive X-ray Spectroscopy of Staphylococcus aureus Biofilms. ACS OMEGA 2024; 9:37610-37620. [PMID: 39281927 PMCID: PMC11391442 DOI: 10.1021/acsomega.4c01168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 06/06/2024] [Accepted: 07/01/2024] [Indexed: 09/18/2024]
Abstract
Understanding the dynamics of biofilm formation and its elemental composition is crucial for developing effective strategies against biofilm-associated infections. In this study, we employed scanning electron microscopy (SEM) and energy-dispersive X-ray spectroscopy (EDS) to investigate the morphological changes and elemental compositions of Staphylococcus aureus biofilms. SEM images revealed distinct stages of biofilm development, from initial aggregation to the formation of mature and aged biofilms. EDS analysis consistently showed elevated levels of sodium (Na), oxygen (O), and phosphorus (P) in the biofilm matrix, indicating its high negative charge and the presence of anionic biopolymers. The incorporation of extracellular DNA (eDNA) into the biofilm matrix, leading to significant retention of sodium ions, underscored the importance of electrostatic interactions in biofilm formation and stability. Our findings highlight the potential of EDS analysis in quantifying elemental compositions and elucidating the role of anionic biopolymers in biofilm development.
Collapse
Affiliation(s)
- Binayak Rimal
- Institute of Biomedical Studies, Baylor University, Waco, Texas 76798, United States
| | - James D Chang
- Department of Chemistry and Biochemistry, Baylor University, Waco, Texas 76798, United States
| | - Chengyin Liu
- Department of Chemistry, Howard University, Washington, District of Columbia 20059, United States
| | - Haley Kim
- Department of Chemistry, University of Maryland, College Park, Maryland 20742, United States
| | - Oluwatobi Aderotoye
- Department of Chemistry, Howard University, Washington, District of Columbia 20059, United States
| | - Bernd Zechmann
- Center for Microscopy and Imaging, Baylor University, Waco, Texas 76798, United States
| | - Sung Joon Kim
- Department of Chemistry, Howard University, Washington, District of Columbia 20059, United States
| |
Collapse
|
22
|
Mishra A, Tabassum N, Aggarwal A, Kim YM, Khan F. Artificial Intelligence-Driven Analysis of Antimicrobial-Resistant and Biofilm-Forming Pathogens on Biotic and Abiotic Surfaces. Antibiotics (Basel) 2024; 13:788. [PMID: 39200087 PMCID: PMC11351874 DOI: 10.3390/antibiotics13080788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/14/2024] [Accepted: 08/19/2024] [Indexed: 09/01/2024] Open
Abstract
The growing threat of antimicrobial-resistant (AMR) pathogens to human health worldwide emphasizes the need for more effective infection control strategies. Bacterial and fungal biofilms pose a major challenge in treating AMR pathogen infections. Biofilms are formed by pathogenic microbes encased in extracellular polymeric substances to confer protection from antimicrobials and the host immune system. Biofilms also promote the growth of antibiotic-resistant mutants and latent persister cells and thus complicate therapeutic approaches. Biofilms are ubiquitous and cause serious health risks due to their ability to colonize various surfaces, including human tissues, medical devices, and food-processing equipment. Detection and characterization of biofilms are crucial for prompt intervention and infection control. To this end, traditional approaches are often effective, yet they fail to identify the microbial species inside biofilms. Recent advances in artificial intelligence (AI) have provided new avenues to improve biofilm identification. Machine-learning algorithms and image-processing techniques have shown promise for the accurate and efficient detection of biofilm-forming microorganisms on biotic and abiotic surfaces. These advancements have the potential to transform biofilm research and clinical practice by allowing faster diagnosis and more tailored therapy. This comprehensive review focuses on the application of AI techniques for the identification of biofilm-forming pathogens in various industries, including healthcare, food safety, and agriculture. The review discusses the existing approaches, challenges, and potential applications of AI in biofilm research, with a particular focus on the role of AI in improving diagnostic capacities and guiding preventative actions. The synthesis of the current knowledge and future directions, as described in this review, will guide future research and development efforts in combating biofilm-associated infections.
Collapse
Affiliation(s)
- Akanksha Mishra
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144001, Punjab, India;
| | - Nazia Tabassum
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea; (N.T.); (Y.-M.K.)
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea
| | - Ashish Aggarwal
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144001, Punjab, India;
| | - Young-Mog Kim
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea; (N.T.); (Y.-M.K.)
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea
- Department of Food Science and Technology, Pukyong National University, Busan 48513, Republic of Korea
| | - Fazlurrahman Khan
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea; (N.T.); (Y.-M.K.)
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea
- Institute of Fisheries Science, Pukyong National University, Busan 48513, Republic of Korea
- International Graduate Program of Fisheries Science, Pukyong National University, Busan 48513, Republic of Korea
| |
Collapse
|
23
|
Gong F, Xin S, Liu X, He C, Yu X, Pan L, Zhang S, Gao H, Xu J. Multiple biological characteristics and functions of intestinal biofilm extracellular polymers: friend or foe? Front Microbiol 2024; 15:1445630. [PMID: 39224216 PMCID: PMC11367570 DOI: 10.3389/fmicb.2024.1445630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
The gut microbiota is vital to human health, and their biofilms significantly impact intestinal immunity and the maintenance of microbial balance. Certain pathogens, however, can employ biofilms to elude identification by the immune system and medical therapy, resulting in intestinal diseases. The biofilm is formed by extracellular polymorphic substances (EPS), which shield microbial pathogens from the host immune system and enhance its antimicrobial resistance. Therefore, investigating the impact of extracellular polysaccharides released by pathogens that form biofilms on virulence and defence mechanisms is crucial. In this review, we provide a comprehensive overview of current pathogenic biofilm research, deal with the role of extracellular polymers in the formation and maintenance of pathogenic biofilm, and elaborate different prevention and treatment strategies to provide an innovative approach to the treatment of intestinal pathogen-based diseases.
Collapse
Affiliation(s)
- Fengrong Gong
- Department of Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Shuzi Xin
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xiaohui Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Chengwei He
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xinyi Yu
- Department of Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Luming Pan
- Department of Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Sitian Zhang
- Department of Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Han Gao
- Department of Clinical Laboratory, Aerospace Center Hospital, Beijing, China
| | - Jingdong Xu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| |
Collapse
|
24
|
Walton B, Abbondante S, Marshall ME, Dobruchowska JM, Alvi A, Gallagher LA, Vallikat N, Zhang Z, Wozniak DJ, Yu EW, Boons GJ, Pearlman E, Rietsch A. A biofilm-tropic Pseudomonas aeruginosa bacteriophage uses the exopolysaccharide Psl as receptor. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.12.607380. [PMID: 39185188 PMCID: PMC11343166 DOI: 10.1101/2024.08.12.607380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Bacteria in nature can exist in multicellular communities called biofilms. Biofilms also form in the course of many infections. Pseudomonas aeruginosa infections frequently involve biofilms, which contribute materially to the difficulty to treat these infections with antibiotic therapy. Many biofilm-related characteristics are controlled by the second messenger, cyclic-di-GMP, which is upregulated on surface contact. Among these factors is the exopolysaccharide Psl, which is a critically important component of the biofilm matrix. Here we describe the discovery of a P. aeruginosa bacteriophage, which we have called Clew-1, that directly binds to and uses Psl as a receptor. While this phage does not efficiently infect planktonically growing bacteria, it can disrupt P. aeruginosa biofilms and replicate in biofilm bacteria. We further demonstrate that the Clew-1 can reduce the bacterial burden in a mouse model of P. aeruginosa keratitis, which is characterized by the formation of a biofilm on the cornea. Due to its reliance on Psl for infection, Clew-1 does not actually form plaques on wild-type bacteria under standard in vitro conditions. This argues that our standard isolation procedures likely exclude bacteriophage that are adapted to using biofilm markers for infection. Importantly, the manner in which we isolated Clew-1 can be easily extended to other strains of P. aeruginosa and indeed other bacterial species, which will fuel the discovery of other biofilm-tropic bacteriophage and expand their therapeutic use.
Collapse
Affiliation(s)
- Brenna Walton
- Dept. of Molecular Biology and Microbiology, Case Western Reserve University, Cleveland, OH, U.S.A
| | - Serena Abbondante
- Dept. of Ophthalmology, University of California, Irvine, CA, U.S.A
- Institute of Immunology, University of California, Irvine, CA, U.S.A
| | - Michaela Ellen Marshall
- Dept. of Ophthalmology, University of California, Irvine, CA, U.S.A
- Institute of Immunology, University of California, Irvine, CA, U.S.A
| | - Justyna M. Dobruchowska
- Dept. of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, NL
| | - Amani Alvi
- Dept. of Molecular Biology and Microbiology, Case Western Reserve University, Cleveland, OH, U.S.A
| | | | - Nikhil Vallikat
- Dept. of Molecular Biology and Microbiology, Case Western Reserve University, Cleveland, OH, U.S.A
| | - Zhemin Zhang
- Dept. of Pharmacology, Case Western Reserve University, Cleveland, OH, U.S.A
| | - Daniel J. Wozniak
- Dept. of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, U.S.A
- Dept. of Microbiology, The Ohio State University, Columbus, OH, U.S.A
| | - Edward W. Yu
- Dept. of Pharmacology, Case Western Reserve University, Cleveland, OH, U.S.A
| | - Geert-Jan Boons
- Dept. of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, NL
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, U.S.A
- Dept. of Chemistry, University of Georgia, Athens, GA, U.S.A
| | - Eric Pearlman
- Dept. of Ophthalmology, University of California, Irvine, CA, U.S.A
- Institute of Immunology, University of California, Irvine, CA, U.S.A
| | - Arne Rietsch
- Dept. of Molecular Biology and Microbiology, Case Western Reserve University, Cleveland, OH, U.S.A
| |
Collapse
|
25
|
David A, Tahrioui A, Tareau AS, Forge A, Gonzalez M, Bouffartigues E, Lesouhaitier O, Chevalier S. Pseudomonas aeruginosa Biofilm Lifecycle: Involvement of Mechanical Constraints and Timeline of Matrix Production. Antibiotics (Basel) 2024; 13:688. [PMID: 39199987 PMCID: PMC11350761 DOI: 10.3390/antibiotics13080688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 09/01/2024] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen causing acute and chronic infections, especially in immunocompromised patients. Its remarkable adaptability and resistance to various antimicrobial treatments make it difficult to eradicate. Its persistence is enabled by its ability to form a biofilm. Biofilm is a community of sessile micro-organisms in a self-produced extracellular matrix, which forms a scaffold facilitating cohesion, cell attachment, and micro- and macro-colony formation. This lifestyle provides protection against environmental stresses, the immune system, and antimicrobial treatments, and confers the capacity for colonization and long-term persistence, often characterizing chronic infections. In this review, we retrace the events of the life cycle of P. aeruginosa biofilm, from surface perception/contact to cell spreading. We focus on the importance of extracellular appendages, mechanical constraints, and the kinetics of matrix component production in each step of the biofilm life cycle.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Sylvie Chevalier
- Univ Rouen Normandie, Univ Caen Normandie, Normandie Univ, CBSA UR 4312, F-76000 Rouen, France
| |
Collapse
|
26
|
Çevikbaş H, Ulusoy S, Kaya Kinaytürk N. Exploring rose absolute and phenylethyl alcohol as novel quorum sensing inhibitors in Pseudomonas aeruginosa and Chromobacterium violaceum. Sci Rep 2024; 14:15666. [PMID: 38977845 PMCID: PMC11231148 DOI: 10.1038/s41598-024-66888-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 07/05/2024] [Indexed: 07/10/2024] Open
Abstract
Inter-cellular signaling, referred to as quorum sensing (QS), regulates the production of virulence factors in numerous gram-negative bacteria, such as the human pathogens Pseudomonas aeruginosa and Chromobacterium violaceum. QS inhibition may provide an opportunity for the treatment of bacterial infections. This represents the initial study to examine the antibiofilm and antivirulence capabilities of rose absolute and its primary component, phenylethyl alcohol. QS inhibition was assessed by examining extracellular exopolysaccharide synthesis, biofilm development, and swarming motility in P. aeruginosa PAO1, along with violacein production in C. violaceum ATCC 12472. Molecular docking analysis was conducted to explore the mechanism by which PEA inhibits QS. Our results indicate that rose absolute and PEA caused decrease in EPS production (60.5-33.5%), swarming motility (94.7-64.5%), and biofilm formation (98.53-55.5%) in the human pathogen P. aeruginosa PAO1. Violacein production decreased by 98.1% and 62.5% with an absolute (0.5 v/v %) and PEA (2 mM). Moreover, the molecular docking analysis revealed a promising competitive interaction between PEA and AHLs. Consequently, this study offers valuable insights into the potential of rose absolute and PEA as inhibitors of QS in P. aeruginosa and C. violaceum.
Collapse
Affiliation(s)
- Halime Çevikbaş
- Faculty of Engineering and Natural Sciences, Biology Department, Süleyman Demirel University, Isparta, 32260, Turkey
| | - Seyhan Ulusoy
- Faculty of Engineering and Natural Sciences, Biology Department, Süleyman Demirel University, Isparta, 32260, Turkey.
| | - Neslihan Kaya Kinaytürk
- Faculty of Arts and Science, Nanoscience and Nanotechnology Department, Mehmet Akif Ersoy University, Burdur, 15100, Turkey
| |
Collapse
|
27
|
Crabbé A. Moving Beyond Antibiotics: Exploring the Potential of an Antivirulence Monoclonal Antibody against Pseudomonas aeruginosa in Bronchiectasis. Am J Respir Crit Care Med 2024; 210:8-9. [PMID: 38780075 PMCID: PMC11197072 DOI: 10.1164/rccm.202405-0974ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 05/19/2024] [Indexed: 05/25/2024] Open
Affiliation(s)
- Aurélie Crabbé
- Laboratory of Pharmaceutical Microbiology Ghent University Ghent, Belgium
| |
Collapse
|
28
|
Quaye JA, Wood KE, Snelgrove C, Ouedraogo D, Gadda G. An active site mutation induces oxygen reactivity in D-arginine dehydrogenase: A case of superoxide diverting protons. J Biol Chem 2024; 300:107381. [PMID: 38762175 PMCID: PMC11193025 DOI: 10.1016/j.jbc.2024.107381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/09/2024] [Accepted: 05/13/2024] [Indexed: 05/20/2024] Open
Abstract
Enzymes are potent catalysts that increase biochemical reaction rates by several orders of magnitude. Flavoproteins are a class of enzymes whose classification relies on their ability to react with molecular oxygen (O2) during catalysis using ionizable active site residues. Pseudomonas aeruginosa D-arginine dehydrogenase (PaDADH) is a flavoprotein that oxidizes D-arginine for P. aeruginosa survival and biofilm formation. The crystal structure of PaDADH reveals the interaction of the glutamate 246 (E246) side chain with the substrate and at least three other active site residues, establishing a hydrogen bond network in the active site. Additionally, E246 likely ionizes to facilitate substrate binding during PaDADH catalysis. This study aimed to investigate how replacing the E246 residue with leucine affects PaDADH catalysis and its ability to react with O2 using steady-state kinetics coupled with pH profile studies. The data reveal a gain of O2 reactivity in the E246L variant, resulting in a reduced flavin semiquinone species and superoxide (O2•-) during substrate oxidation. The O2•- reacts with active site protons, resulting in an observed nonstoichiometric slope of 1.5 in the enzyme's log (kcat/Km) pH profile with D-arginine. Adding superoxide dismutase results in an observed correction of the slope to 1.0. This study demonstrates how O2•- can alter the slopes of limbs in the pH profiles of flavin-dependent enzymes and serves as a model for correcting nonstoichiometric slopes in elucidating reaction mechanisms of flavoproteins.
Collapse
Affiliation(s)
- Joanna A Quaye
- Department of Chemistry, Georgia State University, Atlanta, Georgia, USA
| | - Kendall E Wood
- Biology Department, Morehouse College, Atlanta, Georgia, USA
| | - Claire Snelgrove
- The Gwinnett School of Mathematics, Science, and Technology, Lawrenceville, Georgia, USA
| | - Daniel Ouedraogo
- Department of Chemistry, Georgia State University, Atlanta, Georgia, USA
| | - Giovanni Gadda
- Department of Chemistry, Georgia State University, Atlanta, Georgia, USA; Department of Biology, Georgia State University, Atlanta, Georgia, USA; Department of the Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia, USA.
| |
Collapse
|
29
|
Rolon ML, Voloshchuk O, Bartlett KV, LaBorde LF, Kovac J. Multi-species biofilms of environmental microbiota isolated from fruit packing facilities promoted tolerance of Listeria monocytogenes to benzalkonium chloride. Biofilm 2024; 7:100177. [PMID: 38304489 PMCID: PMC10832383 DOI: 10.1016/j.bioflm.2024.100177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 01/06/2024] [Accepted: 01/08/2024] [Indexed: 02/03/2024] Open
Abstract
Listeria monocytogenes may survive and persist in food processing environments due to formation of complex multi-species biofilms of environmental microbiota that co-exists in these environments. This study aimed to determine the effect of selected environmental microbiota on biofilm formation and tolerance of L. monocytogenes to benzalkonium chloride in formed biofilms. The studied microbiota included bacterial families previously shown to co-occur with L. monocytogenes in tree fruit packing facilities, including Pseudomonadaceae, Xanthomonadaceae, Microbacteriaceae, and Flavobacteriaceae. Biofilm formation ability and the effect of formed biofilms on the tolerance of L. monocytogenes to benzalkonium chloride was measured in single- and multi-family assemblages. Biofilms were grown statically on polystyrene pegs submerged in a R2A broth. Biofilm formation was quantified using a crystal violet assay, spread-plating, confocal laser scanning microscopy, and its composition was assessed using amplicon sequencing. The concentration of L. monocytogenes in biofilms was determined using the most probable number method. Biofilms were exposed to the sanitizer benzalkonium chloride, and the death kinetics of L. monocytogenes were quantified using a most probable number method. A total of 8, 8, 6, and 3 strains of Pseudomonadaceae, Xanthomonadaceae, Microbacteriaceae, and Flavobacteriaceae, respectively, were isolated from the environmental microbiota of tree fruit packing facilities and were used in this study. Biofilms formed by Pseudomonadaceae, Xanthomonadaceae, and all multi-family assemblages had significantly higher concentration of bacteria, as well as L. monocytogenes, compared to biofilms formed by L. monocytogenes alone. Furthermore, multi-family assemblage biofilms increased the tolerance of L. monocytogenes to benzalkonium chloride compared to L. monocytogenes mono-species biofilms and planktonic multi-family assemblages. These findings suggest that L. monocytogenes control strategies should focus not only on assessing the efficacy of sanitizers against L. monocytogenes, but also against biofilm-forming microorganisms that reside in the food processing built environment, such as Pseudomonadaceae or Xanthomonadaceae.
Collapse
Affiliation(s)
- M. Laura Rolon
- Department of Food Science, The Pennsylvania State University, University Park, PA, 16802, USA
- One Health Microbiome Center, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Olena Voloshchuk
- Department of Food Science, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Katelyn V. Bartlett
- Department of Food Science, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Luke F. LaBorde
- Department of Food Science, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Jasna Kovac
- Department of Food Science, The Pennsylvania State University, University Park, PA, 16802, USA
- One Health Microbiome Center, The Pennsylvania State University, University Park, PA, 16802, USA
| |
Collapse
|
30
|
Weaver AA, Jia J, Cutri AR, Madukoma CS, Vaerewyck CM, Bohn PW, Shrout JD. Alkyl quinolones mediate heterogeneous colony biofilm architecture that improves community-level survival. J Bacteriol 2024; 206:e0009524. [PMID: 38564677 PMCID: PMC11025328 DOI: 10.1128/jb.00095-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 03/11/2024] [Indexed: 04/04/2024] Open
Abstract
Bacterial communities exhibit complex self-organization that contributes to their survival. To better understand the molecules that contribute to transforming a small number of cells into a heterogeneous surface biofilm community, we studied acellular aggregates, structures seen by light microscopy in Pseudomonas aeruginosa colony biofilms using light microscopy and chemical imaging. These structures differ from cellular aggregates, cohesive clusters of cells important for biofilm formation, in that they are visually distinct from cells using light microscopy and are reliant on metabolites for assembly. To investigate how these structures benefit a biofilm community we characterized three recurrent types of acellular aggregates with distinct geometries that were each abundant in specific areas of these biofilms. Alkyl quinolones (AQs) were essential for the formation of all aggregate types with AQ signatures outside the aggregates below the limit of detection. These acellular aggregates spatially sequester AQs and differentiate the biofilm space. However, the three types of aggregates showed differing properties in their size, associated cell death, and lipid content. The largest aggregate type co-localized with spatially confined cell death that was not mediated by Pf4 bacteriophage. Biofilms lacking AQs were absent of localized cell death but exhibited increased, homogeneously distributed cell death. Thus, these AQ-rich aggregates regulate metabolite accessibility, differentiate regions of the biofilm, and promote survival in biofilms.IMPORTANCEPseudomonas aeruginosa is an opportunistic pathogen with the ability to cause infection in the immune-compromised. It is well established that P. aeruginosa biofilms exhibit resilience that includes decreased susceptibility to antimicrobial treatment. This work examines the self-assembled heterogeneity in biofilm communities studying acellular aggregates, regions of condensed matter requiring alkyl quinolones (AQs). AQs are important to both virulence and biofilm formation. Aggregate structures described here spatially regulate the accessibility of these AQs, differentiate regions of the biofilm community, and despite their association with autolysis, correlate with improved P. aeruginosa colony biofilm survival.
Collapse
Affiliation(s)
- Abigail A. Weaver
- Department of Civil and Environmental Engineering and Earth Sciences, University of Notre Dame, Notre Dame, Indiana, USA
| | - Jin Jia
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana, USA
| | - Allison R. Cutri
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana, USA
| | - Chinedu S. Madukoma
- Department of Civil and Environmental Engineering and Earth Sciences, University of Notre Dame, Notre Dame, Indiana, USA
| | - Catherine M. Vaerewyck
- Department of Civil and Environmental Engineering and Earth Sciences, University of Notre Dame, Notre Dame, Indiana, USA
| | - Paul W. Bohn
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana, USA
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana, USA
| | - Joshua D. Shrout
- Department of Civil and Environmental Engineering and Earth Sciences, University of Notre Dame, Notre Dame, Indiana, USA
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, USA
| |
Collapse
|
31
|
Yaeger LN, Ranieri MRM, Chee J, Karabelas-Pittman S, Rudolph M, Giovannoni AM, Harvey H, Burrows LL. A genetic screen identifies a role for oprF in Pseudomonas aeruginosa biofilm stimulation by subinhibitory antibiotics. NPJ Biofilms Microbiomes 2024; 10:30. [PMID: 38521769 PMCID: PMC10960818 DOI: 10.1038/s41522-024-00496-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 03/05/2024] [Indexed: 03/25/2024] Open
Abstract
Biofilms are surface-associated communities of bacteria that grow in a self-produced matrix of polysaccharides, proteins, and extracellular DNA (eDNA). Sub-minimal inhibitory concentrations (sub-MIC) of antibiotics induce biofilm formation, potentially as a defensive response to antibiotic stress. However, the mechanisms behind sub-MIC antibiotic-induced biofilm formation are unclear. We show that treatment of Pseudomonas aeruginosa with multiple classes of sub-MIC antibiotics with distinct targets induces biofilm formation. Further, addition of exogenous eDNA or cell lysate failed to increase biofilm formation to the same extent as antibiotics, suggesting that the release of cellular contents by antibiotic-driven bacteriolysis is insufficient. Using a genetic screen for stimulation-deficient mutants, we identified the outer membrane porin OprF and the ECF sigma factor SigX as important. Similarly, loss of OmpA - the Escherichia coli OprF homolog - prevented sub-MIC antibiotic stimulation of E. coli biofilms. Our screen also identified the periplasmic disulfide bond-forming enzyme DsbA and a predicted cyclic-di-GMP phosphodiesterase encoded by PA2200 as essential for biofilm stimulation. The phosphodiesterase activity of PA2200 is likely controlled by a disulfide bond in its regulatory domain, and folding of OprF is influenced by disulfide bond formation, connecting the mutant phenotypes. Addition of reducing agent dithiothreitol prevented sub-MIC antibiotic biofilm stimulation. Finally, activation of a c-di-GMP-responsive promoter follows treatment with sub-MIC antibiotics in the wild-type but not an oprF mutant. Together, these results show that antibiotic-induced biofilm formation is likely driven by a signaling pathway that translates changes in periplasmic redox state into elevated biofilm formation through increases in c-di-GMP.
Collapse
Affiliation(s)
- Luke N Yaeger
- Biochemistry and Biomedical Sciences and the Michael G. DeGroote Centre for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| | - Michael R M Ranieri
- Biochemistry and Biomedical Sciences and the Michael G. DeGroote Centre for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| | - Jessica Chee
- Biochemistry and Biomedical Sciences and the Michael G. DeGroote Centre for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| | - Sawyer Karabelas-Pittman
- Biochemistry and Biomedical Sciences and the Michael G. DeGroote Centre for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| | - Madeleine Rudolph
- Biochemistry and Biomedical Sciences and the Michael G. DeGroote Centre for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| | - Alessio M Giovannoni
- Biochemistry and Biomedical Sciences and the Michael G. DeGroote Centre for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| | - Hanjeong Harvey
- Biochemistry and Biomedical Sciences and the Michael G. DeGroote Centre for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| | - Lori L Burrows
- Biochemistry and Biomedical Sciences and the Michael G. DeGroote Centre for Infectious Disease Research, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
32
|
Wu D, Zhang B, Shi S, Tang R, Qiao C, Li T, Jia J, Yang M, Si X, Wang Y, Sun X, Xiao D, Li F, Song H. Engineering extracellular electron transfer to promote simultaneous brewing wastewater treatment and chromium reduction. JOURNAL OF HAZARDOUS MATERIALS 2024; 465:133171. [PMID: 38147750 DOI: 10.1016/j.jhazmat.2023.133171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 11/06/2023] [Accepted: 12/01/2023] [Indexed: 12/28/2023]
Abstract
Microbial fuel cell (MFC) technology has been developed for wastewater treatment in the anodic chamber, and heavy metal reduction in the cathodic chamber. However, the limited extracellular electron transfer (EET) rate of exoelectrogens remained a constraint for practical applications of MFCs. Here, a MFC system that used the electricity derived from anodic wastewater treatment to drive cathodic Cr6+ reduction was developed, which enabled an energy self-sustained approach to efficiently address Cr6+ contamination. This MFC system was achieved by screening exoelectrogens with a superior EET rate, promoting the exoelectrogenic EET rate, and constructing a conductive bio-anode. Firstly, Shewanella algae-L3 was screened from brewing wastewater acclimatized sludge, which generated power density of 566.83 mW m-2. Secondly, to facilitate EET rate, flavin synthesis gene operon ribADEHC was overexpressed in engineered S. algae-L3F to increase flavins biosynthesis, which promoted the power density to 1233.21 mW m-2. Thirdly, to facilitate interface electron transfer, carbon nanotube (CNT) was employed to construct a S. algae-L3F-CNT bio-anode, which further enhanced power density to 3112.98 mW m-2. Lastly, S. algae-L3F-CNT bio-anode was used to harvest electrical energy from brewing wastewater to drive cathodic Cr6+ reduction in MFC, realizing 71.43% anodic COD removal and 98.14% cathodic Cr6+ reduction. This study demonstrated that enhanced exoelectrogenic EET could facilitate cathodic Cr6+ reduction in MFC.
Collapse
Affiliation(s)
- Deguang Wu
- Key Laboratory of Industrial Fermentation Microbiology (Ministry of Education), Tianjin Industrial Microbiology Key Lab, College of Biotechnology, Tianjin University of Science and Technology, Box 08, No. 29, 13ST. TEDA, Tianjin 300457, PR China
| | - Baocai Zhang
- Frontiers Science Center for Synthetic Biology (Ministry of Education), Key Laboratory of Systems Bioengineering, and School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, PR China
| | - Sicheng Shi
- Frontiers Science Center for Synthetic Biology (Ministry of Education), Key Laboratory of Systems Bioengineering, and School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, PR China
| | - Rui Tang
- Frontiers Science Center for Synthetic Biology (Ministry of Education), Key Laboratory of Systems Bioengineering, and School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, PR China
| | - Chunxiao Qiao
- Frontiers Science Center for Synthetic Biology (Ministry of Education), Key Laboratory of Systems Bioengineering, and School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, PR China
| | - Teng Li
- Frontiers Science Center for Synthetic Biology (Ministry of Education), Key Laboratory of Systems Bioengineering, and School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, PR China
| | - Jichao Jia
- Frontiers Science Center for Synthetic Biology (Ministry of Education), Key Laboratory of Systems Bioengineering, and School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, PR China
| | - Meiyi Yang
- Frontiers Science Center for Synthetic Biology (Ministry of Education), Key Laboratory of Systems Bioengineering, and School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, PR China
| | - Xiaoguang Si
- The Institute of Seawater Desalination and Multipurpose Utilization, Ministry of Natural Resources, Tianjin, PR China
| | - Yifei Wang
- College of Biological Engineering, Tianjin Agricultural University, Tianjin, PR China
| | - Xi Sun
- College of Biological Engineering, Tianjin Agricultural University, Tianjin, PR China
| | - Dongguang Xiao
- Key Laboratory of Industrial Fermentation Microbiology (Ministry of Education), Tianjin Industrial Microbiology Key Lab, College of Biotechnology, Tianjin University of Science and Technology, Box 08, No. 29, 13ST. TEDA, Tianjin 300457, PR China.
| | - Feng Li
- Frontiers Science Center for Synthetic Biology (Ministry of Education), Key Laboratory of Systems Bioengineering, and School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, PR China.
| | - Hao Song
- Frontiers Science Center for Synthetic Biology (Ministry of Education), Key Laboratory of Systems Bioengineering, and School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, PR China.
| |
Collapse
|
33
|
Bakalakos M, Ampadiotaki MM, Vlachos C, Sipsas N, Pneumaticos S, Vlamis J. Molecular Mechanisms of Biofilm Formation on Orthopaedic Implants: Review of the Literature. MAEDICA 2024; 19:129-136. [PMID: 38736937 PMCID: PMC11079743 DOI: 10.26574/maedica.2021.19.1.129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 05/14/2024]
Abstract
Orthopaedic implant-associated infections (OIAIs) is one of the most catastrophic complications following joint arthroplasty or fracture fixation. Given the increasing number of orthopaedic implants which are used annually, periprosthetic infections emerge as a global problem. Their diagnosis and consequent therapeutic management remain challenging for clinicians. Biofilm formation is a complex and only partially understood process that has not been extensively studied. Understanding the underlying mechanisms involved in biofilm formation is crucial in the amelioration of both diagnosis and therapeutic management of OIAIs. We performed a literature review of the molecular mechanisms of biofilm formation and discussed the four most common and thoroughly researched microbes of biofilm-related OIAIs.
Collapse
Affiliation(s)
- Matthaios Bakalakos
- 3rd Orthopaedic Department, National and Kapodistrian University of Athens, KAT General Hospital, Athens, Greece
| | | | - Christos Vlachos
- 3rd Orthopaedic Department, National and Kapodistrian University of Athens, KAT General Hospital, Athens, Greece
| | - Nikolaos Sipsas
- Infectious Diseases, National and Kapodistrian University of Athens, Athens, Greece
| | - Spiros Pneumaticos
- 3rd Orthopaedic Department, National and Kapodistrian University of Athens, KAT General Hospital, Athens, Greece
| | - John Vlamis
- 3rd Orthopaedic Department, National and Kapodistrian University of Athens, KAT General Hospital, Athens, Greece
| |
Collapse
|
34
|
Kameswaran S, Gujjala S, Zhang S, Kondeti S, Mahalingam S, Bangeppagari M, Bellemkonda R. Quenching and quorum sensing in bacterial bio-films. Res Microbiol 2024; 175:104085. [PMID: 37268165 DOI: 10.1016/j.resmic.2023.104085] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/24/2023] [Accepted: 05/25/2023] [Indexed: 06/04/2023]
Abstract
Quorum sensing (QS) is the ability of bacteria to monitor their population density and adjust gene expression accordingly. QS-regulated processes include host-microbe interactions, horizontal gene transfer, and multicellular behaviours (such as the growth and development of biofilm). The creation, transfer, and perception of bacterial chemicals known as autoinducers or QS signals are necessary for QS signalling (e.g. N-acylhomoserine lactones). Quorum quenching (QQ), another name for the disruption of QS signalling, comprises a wide range of events and mechanisms that are described and analysed in this study. In order to better comprehend the targets of the QQ phenomena that organisms have naturally developed and are currently being actively researched from practical perspectives, we first surveyed the diversity of QS-signals and QS-associated responses. Next, the mechanisms, molecular players, and targets related to QS interference are discussed, with a focus on natural QQ enzymes and compounds that function as QS inhibitors. To illustrate the processes and biological functions of QS inhibition in microbe-microbe and host-microbe interactions, a few QQ paradigms are described in detail. Finally, certain QQ techniques are offered as potential instruments in a variety of industries, including agriculture, medical, aquaculture, crop production, and anti-biofouling areas.
Collapse
Affiliation(s)
- Srinivasan Kameswaran
- Department of Botany, Vikrama Simhapuri University College, Kavali, Andhra Pradesh, India
| | - Sudhakara Gujjala
- Department of Biochemistry, Sri Krishnadevaray a University, Ananthapuram, Andhra Pradesh, India
| | - Shaoqing Zhang
- School of Chemistry and Civil Engineering, Shaoguan University, Shaoguan, 512005, PR China
| | - Suresh Kondeti
- Multi-Disciplinary Research Unit, Nizam's Institute of Medical Sciences, Hyderabad, 500082, India
| | - Sundararajan Mahalingam
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Manjunatha Bangeppagari
- Department of Cell Biology & Molecular Genetics, Sri Devaraj Urs Academy of Higher Education and Research (Deemed to Be University), Tamaka, Kolar, 563103, Karnataka, India
| | - Ramesh Bellemkonda
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
35
|
Böhning J, Tarafder AK, Bharat TA. The role of filamentous matrix molecules in shaping the architecture and emergent properties of bacterial biofilms. Biochem J 2024; 481:245-263. [PMID: 38358118 PMCID: PMC10903470 DOI: 10.1042/bcj20210301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 01/29/2024] [Accepted: 01/31/2024] [Indexed: 02/16/2024]
Abstract
Numerous bacteria naturally occur within spatially organised, multicellular communities called biofilms. Moreover, most bacterial infections proceed with biofilm formation, posing major challenges to human health. Within biofilms, bacterial cells are embedded in a primarily self-produced extracellular matrix, which is a defining feature of all biofilms. The biofilm matrix is a complex, viscous mixture primarily composed of polymeric substances such as polysaccharides, filamentous protein fibres, and extracellular DNA. The structured arrangement of the matrix bestows bacteria with beneficial emergent properties that are not displayed by planktonic cells, conferring protection against physical and chemical stresses, including antibiotic treatment. However, a lack of multi-scale information at the molecular level has prevented a better understanding of this matrix and its properties. Here, we review recent progress on the molecular characterisation of filamentous biofilm matrix components and their three-dimensional spatial organisation within biofilms.
Collapse
Affiliation(s)
- Jan Böhning
- Structural Studies Division, MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, U.K
| | - Abul K. Tarafder
- Structural Studies Division, MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, U.K
| | - Tanmay A.M. Bharat
- Structural Studies Division, MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, U.K
| |
Collapse
|
36
|
Ma Z, Xiao H, Li H, Lu X, Yan J, Nie H, Yin Q. Prodigiosin as an Antibiofilm Agent against the Bacterial Biofilm-Associated Infection of Pseudomonas aeruginosa. Pathogens 2024; 13:145. [PMID: 38392883 PMCID: PMC10891946 DOI: 10.3390/pathogens13020145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/21/2024] [Accepted: 01/23/2024] [Indexed: 02/25/2024] Open
Abstract
Pseudomonas aeruginosa is known to generate bacterial biofilms that increase antibiotic resistance. With the increase of multi-drug resistance in recent years, the formulation of a new therapeutic strategy has seemed urgent. Preliminary findings show that Prodigiosin (PG), derived from chromium-resistant Serratia marcescens, exhibited efficient anti-biofilm activity against Staphylococcus aureus. However, its anti-biofilm activity against P. aeruginosa remains largely unexplored. The anti-biofilm activity of PG against three clinical single drug-resistant P. aeruginosa was evaluated using crystal violet staining, and the viability of biofilms and planktonic cells were also assessed. A model of chronic lung infection was constructed to test the in vivo antibiofilm activity of PG. The results showed that PG inhibited biofilm formation and effectively inhibited the production of pyocyanin and extracellular polysaccharides in vitro, as well as moderated the expression of interleukins (IL-1β, IL-6, IL-10) and tumor necrosis factor (TNF-α) in vivo, which might be attributed to the downregulation of biofilm-related genes such as algA, pelA, and pslM. These findings suggest that PG could be a potential treatment for drug-resistant P aeruginosa and chronic biofilm infections.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Qi Yin
- Department of Health Laboratory Technology, School of Public Health, Chongqing Medical University, No. 61 Daxuecheng Middle Road, Shapingba District, Chongqing 401334, China
| |
Collapse
|
37
|
Hemmati J, Nazari M, Ahmadi A, Bayati M, Jalili M, Taheri M, Mohammadi Y, Asghari B. In vitro evaluation of biofilm phenotypic and genotypic characteristics among clinical isolates of Pseudomonas aeruginosa in Hamadan, West of Iran. J Appl Genet 2024; 65:213-222. [PMID: 38017355 DOI: 10.1007/s13353-023-00811-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 11/03/2023] [Accepted: 11/20/2023] [Indexed: 11/30/2023]
Abstract
Due to high antimicrobial resistance and biofilm-forming ability, Pseudomonas aeruginosa is one of the seriously life-threatening agents causing chronic and nosocomial infections. This study was performed to determine the antibiotic resistance pattern, biofilm formation, and frequency of biofilm-related genes in P. aeruginosa strains. In total, 123 P. aeruginosa isolates were collected from different clinical sources. Antimicrobial susceptibility testing (AST) was performed to detect multidrug-resistant P. aeruginosa (MDRPA) isolates. To evaluate the biofilm-forming isolates, the microtiter plate (MTP) method was carried out. Also, the prevalence of biofilm genotype patterns, including pslA, pslD, pelA, pelF, and algD genes, was detected by polymerases chain reaction (PCR). According to our findings, the highest resistance and susceptibility rates were found in ceftazidime with 74.7% (n = 92) and ciprofloxacin with 42.2% (n = 52), respectively. In our study, the highest level of antibiotic resistance belonged to wound isolates which meropenem had the most antibacterial activity against them. In total, 86.1% (n = 106) P. aeruginosa isolates were determined as MDRPA, of which 61.3% (n = 65) were able to form strong biofilm. The highest and lowest frequency of biofilm-related genes among biofilm producer isolates belonged to pelF with 82.1% (n = 101) and algD with 55.2% (n = 68), respectively. The findings of the conducted study indicate a significant relationship between MDRPA and biofilm genotypic/phenotypic patterns, suggesting the necessity of a careful surveillance program in hospital settings.
Collapse
Affiliation(s)
- Jaber Hemmati
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohsen Nazari
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Amjad Ahmadi
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Maral Bayati
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mahsa Jalili
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohammad Taheri
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Younes Mohammadi
- Department of Epidemiology, School of Public Health, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Babak Asghari
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
38
|
Bajrami D, Sarquis A, Ladero VM, Fernández M, Mizaikoff B. Rapid discrimination of Lentilactobacillus parabuchneri biofilms via in situ infrared spectroscopy. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 304:123391. [PMID: 37714102 DOI: 10.1016/j.saa.2023.123391] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 09/01/2023] [Accepted: 09/08/2023] [Indexed: 09/17/2023]
Abstract
Microbial contamination in food industry is a source of foodborne illnesses and biofilm-related diseases. In particular, biogenic amines (BAs) accumulated in fermented foods via lactic acid bacterial activity exert toxic effects on human health. Among these, biofilms of histamine-producer Lentilactobacillus parabuchneri strains adherent at food processing equipment surfaces can cause food spoilage and poisoning. Understanding the chain of contamination is closely related to elucidating molecular mechanisms of biofilm formation. In the present study, an innovative approach using integrated chemical sensing technologies is demonstrated to fundamentally understand the temporal behavior of biofilms at the molecular level by combining mid-infrared (MIR) spectroscopy and fluorescence sensing strategies. Using these concepts, the biofilm forming capacity of six cheese-isolated L. parabuchneri strains (IPLA 11151, 11150, 11129, 11125, 11122 and 11117) was examined. The cut-off values for the biofilm production ability of each strain were quantified using crystal violet (CV) assays. Real-time infrared attenuated total reflection spectroscopy (IR-ATR) combined with fluorescence quenching oxygen sensing provides insight into distinct molecular mechanisms for each strain. IR spectra showed significant changes in characteristic bands of amides, lactate, nucleic acids, and extracellular polymeric substances (i.e., lipopolysaccharides, phospholipids, phosphodiester, peptidoglycan, etc.), which are major contributors to biofilm maturation involved in the initial adhesion processes. Chemometric methods including principal component analysis and partial least square-discriminant analysis facilitated the rapid determination and classification of cheese isolated L. parabuchneri strains unambiguously differentiating the IR signatures based on their ability to produce biofilm. All biofilms were morphologically characterized by confocal laser scanning microscopy on relevant industrial equipment surfaces. In summary, this innovative approach combining MIR spectroscopy with luminescence sensing enables real-time insight into the molecular composition and formation of L. parabuchneri biofilms.
Collapse
Affiliation(s)
- Diellza Bajrami
- Institute of Analytical and Bioanalytical Chemistry, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Agustina Sarquis
- Dairy Research Institute (IPLA-CSIC), Paseo Rio Linares s/n, 33300 Villaviciosa, Spain
| | - Victor M Ladero
- Dairy Research Institute (IPLA-CSIC), Paseo Rio Linares s/n, 33300 Villaviciosa, Spain; Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - María Fernández
- Dairy Research Institute (IPLA-CSIC), Paseo Rio Linares s/n, 33300 Villaviciosa, Spain; Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain.
| | - Boris Mizaikoff
- Institute of Analytical and Bioanalytical Chemistry, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany; Hahn-Schickard, Sedanstrasse 14, 89077 Ulm, Germany.
| |
Collapse
|
39
|
Jung H. A pore-scale reactive transport modeling study for quorum sensing-driven biofilm dispersal in heterogeneous porous media. Math Biosci 2024; 367:109126. [PMID: 38070765 DOI: 10.1016/j.mbs.2023.109126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 10/26/2023] [Accepted: 12/06/2023] [Indexed: 12/17/2023]
Abstract
Microorganisms regulate the expression of energetically expensive phenotypes via a collective decision-making mechanism known as quorum sensing (QS). This study investigates the intricate dynamics of biofilm growth and QS-controlled biofilm dispersal in heterogeneous porous media, employing a pore-scale reactive transport modeling approach. Model simulations carried out under various fluid flow conditions and biofilm growth scenarios reveal that QS processes are influenced not only by the biomass density of biofilm colonies but also by a complex interplay between pore architecture, flow velocity, and the rates of biofilm growth and dispersal. This study demonstrates that pore architecture controls the initiation of QS processes and advection gives rise to oscillatory growth of biofilms. Such oscillation is suppressed if biofilm dynamics are in favor of sustaining a sufficiently high signal concentration, such as fast growth or slow dispersal rates. By establishing a mathematical framework, this study contributes to the fundamental understanding of QS-controlled biofilm dynamics in complex environments.
Collapse
Affiliation(s)
- Heewon Jung
- Department of Geological Sciences, Chungnam National University, Daejeon 34134, South Korea.
| |
Collapse
|
40
|
Cullom A, Spencer MS, Williams MD, Falkinham JO, Brown C, Edwards MA, Pruden A. Premise Plumbing Pipe Materials and In-Building Disinfectants Shape the Potential for Proliferation of Pathogens and Antibiotic Resistance Genes. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2023; 57:21382-21394. [PMID: 38071676 DOI: 10.1021/acs.est.3c05905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2023]
Abstract
In-building disinfectants are commonly applied to control the growth of pathogens in plumbing, particularly in facilities such as hospitals that house vulnerable populations. However, their application has not been well optimized, especially with respect to interactive effects with pipe materials and potential unintended effects, such as enrichment of antibiotic resistance genes (ARGs) across the microbial community. Here, we used triplicate convectively mixed pipe reactors consisting of three pipe materials (PVC, copper, and iron) for replicated simulation of the distal reaches of premise plumbing and evaluated the effects of incrementally increased doses of chlorine, chloramine, chlorine dioxide, and copper-silver disinfectants. We used shotgun metagenomic sequencing to characterize the resulting succession of the corresponding microbiomes over the course of 37 weeks. We found that both disinfectants and pipe material affected ARG and microbial community taxonomic composition both independently and interactively. Water quality and total bacterial numbers were not found to be predictive of pathogenic species markers. One result of particular concern was the tendency of disinfectants, especially monochloramine, to enrich ARGs. Metagenome assembly indicated that many ARGs were enriched specifically among the pathogenic species. Functional gene analysis was indicative of a response of the microbes to oxidative stress, which is known to co/cross-select for antibiotic resistance. These findings emphasize the need for a holistic evaluation of pathogen control strategies for plumbing.
Collapse
Affiliation(s)
- Abraham Cullom
- Civil and Environmental Engineering, Virginia Tech, 1145 Perry St., 418 Durham Hall, Blacksburg, Virginia 24061, United States
| | - Matheu Storme Spencer
- Civil and Environmental Engineering, Virginia Tech, 1145 Perry St., 418 Durham Hall, Blacksburg, Virginia 24061, United States
| | - Myra D Williams
- Department of Biological Sciences, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Joseph O Falkinham
- Department of Biological Sciences, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Connor Brown
- Department of Genetics, Bioinformatics, and Computational Biology, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Marc A Edwards
- Civil and Environmental Engineering, Virginia Tech, 1145 Perry St., 418 Durham Hall, Blacksburg, Virginia 24061, United States
| | - Amy Pruden
- Civil and Environmental Engineering, Virginia Tech, 1145 Perry St., 418 Durham Hall, Blacksburg, Virginia 24061, United States
| |
Collapse
|
41
|
Liu J, Huang T, Xu Z, Mao Y, Soteyome T, Liu G, Qu C, Yuan L, Ma Q, Zhou F, Seneviratne G. Sub-MIC streptomycin and tetracycline enhanced Staphylococcus aureus Guangzhou-SAU749 biofilm formation, an in-depth study on transcriptomics. Biofilm 2023; 6:100156. [PMID: 37779859 PMCID: PMC10539642 DOI: 10.1016/j.bioflm.2023.100156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 09/16/2023] [Accepted: 09/18/2023] [Indexed: 10/03/2023] Open
Abstract
Staphylococcus aureus is a major human pathogen, a potential "Super-bug" and a typical biofilm forming bacteria. With usage of large amount of antibiotics, the residual antibiotics in clinical settings further complicate the colonization, pathogenesis and resistance of S. aureus. This study aimed at investigating the phenotypical and global gene expression changes on biofilm formation of a clinical S. aureus isolate treated under different types of antibiotics. Firstly, an isolate Guangzhou-SAU749 was selected from a large sale of previously identified S. aureus isolates, which exhibited weak biofilm formation in terms of biomass and viability. Secondly, 9 commonly prescribed antibiotics for S. aureus infections treatment, together with 10 concentrations ranging from 1/128 to 4 minimum inhibitory concentration (MIC) with 2-fold serial dilution, were used as different antibiotic stress conditions. Then, biofilm formation of S. aureus Guangzhou-SAU749 at different stages including 8 h, 16 h, 24 h, and 48 h, was tested by crystal violet and MTS assays. Thirdly, the whole genome of S. aureus Guangzhou-SAU749 was investigated by genome sequencing on PacBio platform. Fourthly, since enhancement of biofilm formation occurred when treated with 1/2 MIC tetracycline (TCY) and 1/4 MIC streptomycin (STR) since 5 h, the relevant biofilm samples were selected and subjected to RNA-seq and bioinformatics analysis. Last, expression of two component system (TCS) and biofilm associated genes in 4 h, 8 h, 16 h, 24 h, and 48 h sub-MIC TCY and STR treated biofilm samples were performed by reverse transcription-quantitative polymerase chain reaction (RT-qPCR). Although most antibiotics lowered the biomass and cell viability of Guangzhou-SAU749 biofilm at concentrations higher than MIC, certain antibiotics including TCY and STR promoted biofilm formation at sub-MICs. Additionally, upon genome sequencing, RNA-seq and RT-qPCR on biofilm samples treated with sub-MIC of TCY and STR at key time points, genes lytR, arlR, hssR, tagA, clfB, atlA and cidA related to TCS and biofilm formation were identified to contribute to the enhanced biofilm formation, providing a theoretical basis for further controlling on S. aureus biofilm formation.
Collapse
Affiliation(s)
- Junyan Liu
- College of Light Industry and Food Science, Guangdong Provincial Key Laboratory of Lingnan Specialty Food Science and Technology, Academy of Contemporary Agricultural Engineering Innovations, Zhongkai University of Agriculture and Engineering, Guangzhou, 510225, China
- Key Laboratory of Green Processing and Intelligent Manufacturing of Lingnan Specialty Food, Ministry of Agriculture, Guangzhou, 510225, China
| | - Tengyi Huang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Zhenbo Xu
- Department of Laboratory Medicine, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Yuzhu Mao
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Vegetable Protein Processing Ministry of Education, South China University of Technology, Guangzhou, 510640, China
| | - Thanapop Soteyome
- Home Economics Technology, Rajamangala University of Technology Phra Nakhon, Bangkok, Thailand
| | - Gongliang Liu
- College of Light Industry and Food Science, Guangdong Provincial Key Laboratory of Lingnan Specialty Food Science and Technology, Academy of Contemporary Agricultural Engineering Innovations, Zhongkai University of Agriculture and Engineering, Guangzhou, 510225, China
- Key Laboratory of Green Processing and Intelligent Manufacturing of Lingnan Specialty Food, Ministry of Agriculture, Guangzhou, 510225, China
| | - Chunyun Qu
- College of Light Industry and Food Science, Guangdong Provincial Key Laboratory of Lingnan Specialty Food Science and Technology, Academy of Contemporary Agricultural Engineering Innovations, Zhongkai University of Agriculture and Engineering, Guangzhou, 510225, China
- Key Laboratory of Green Processing and Intelligent Manufacturing of Lingnan Specialty Food, Ministry of Agriculture, Guangzhou, 510225, China
| | - Lei Yuan
- School of Food Science and Engineering, Yangzhou University, Yangzhou, Jiangsu, 225127, PR China
| | - Qin Ma
- Sericultural & Agri-Food Research Institute Guangdong Academy of Agricultural Sciences/Key Laboratory of Functional Foods, Ministry of Agriculture /Guangdong Key Laboratory of Agricultural Products Processing, Guangzhou, 510610, China
| | - Fang Zhou
- The First Affiliated Hospital, Sun Yan-Sen University, Guangzhou, 510080, China
| | - Gamini Seneviratne
- National Institute of Fundamental Studies, Hantana road, Kandy, Sri Lanka
| |
Collapse
|
42
|
Yang J, Wu P, Weng Y, Lin Y, Chen Z, Yu F, Lv X, Ni L, Han J. Rational Design and Antimicrobial Potency Assessment of Abaecin Analogues. ACS Biomater Sci Eng 2023; 9:6698-6714. [PMID: 37988627 DOI: 10.1021/acsbiomaterials.3c01234] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2023]
Abstract
The widespread and escalating emergence of multidrug resistance is now recognized as one of the most severe global threats to human health. To address the urgent issue of drug-resistant bacteria and the limitation of effective clinical treatments, antimicrobial peptides (AMPs) have been developed as promising substituents of conventional antibiotics. In this study, rational design strategies were employed to acquire seven cationic and α-helical engineered peptides based on the original template of Abaecin. After investigation, we found that AC7 (LLRRWKKLFKKIIRWPRPLPNPGH) demonstrated potent and broad-spectrum antimicrobial activity. Additionally, it demonstrated low cytotoxicity and hemolysis while maintaining good stability. Notably, AC7 displays the antibacterial mechanism with superior abilities in cell membrane disruption and potential DNA binding in vitro, as well as effectively disrupting biofilms. Moreover, the murine skin wound model infected with drug-resistant Pseudomonas aeruginosa was employed to evaluate the anti-infective efficacy and therapeutic potential of AC7. It was observed that AC7 displays a remarkable capacity to inhibit wound colonization, reduce levels of inflammatory cytokines (TNF-α) and inflammatory cells (white blood cells (WBC), monocytes (MONO), lymphocytes (LYMPH), neutrophils (GRAN)), promote the levels of IL-10 and VEGF, and enhance wound healing. Overall, these findings demonstrate the potential of AC7 as a viable alternative to traditional antibiotics.
Collapse
Affiliation(s)
- Jie Yang
- Institute of Food Science and Technology, College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, China
| | - Peifen Wu
- Institute of Food Science and Technology, College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, China
| | - Yanlin Weng
- Institute of Food Science and Technology, College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, China
| | - Yayi Lin
- Institute of Food Science and Technology, College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, China
| | - Zhiying Chen
- Institute of Food Science and Technology, College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, China
| | - Fengfan Yu
- Institute of Food Science and Technology, College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, China
| | - Xucong Lv
- Institute of Food Science and Technology, College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, China
- Food Nutrition and Health Research Center, School of Advanced Manufacturing, Fuzhou University, Jinjiang, Fujian 362200, China
| | - Li Ni
- Institute of Food Science and Technology, College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, China
- Food Nutrition and Health Research Center, School of Advanced Manufacturing, Fuzhou University, Jinjiang, Fujian 362200, China
| | - Jinzhi Han
- Institute of Food Science and Technology, College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, China
- Food Nutrition and Health Research Center, School of Advanced Manufacturing, Fuzhou University, Jinjiang, Fujian 362200, China
| |
Collapse
|
43
|
Yu Y, Han F, Yang M, Zhang X, Chen Y, Yu M, Wang Y. Pseudomonas composti isolate from oyster digestive tissue specifically binds with norovirus GII.6 via Psl extracellular polysaccharide. Int J Food Microbiol 2023; 406:110369. [PMID: 37666026 DOI: 10.1016/j.ijfoodmicro.2023.110369] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 08/15/2023] [Accepted: 08/21/2023] [Indexed: 09/06/2023]
Abstract
Oysters are recognized as important vectors for human norovirus transmission in the environment. Whether norovirus binds to bacteria in oyster digestive tissues (ODTs) remains unknown. To shed light on this concern, ODT-54 and ODT-32, positive for histo-blood group antigen (HBGA) -like substances, were isolated from ODTs and identified as Pseudomonas composti and Enterobacter cloacae, respectively. The binding of noroviruses (GII.4 and GII.6 P domains) to bacterial cells (ODT-32 and ODT-54; in situ assay) as well as extracted extracellular polysaccharides (EPSs; in vitro assay) was analyzed by flow cytometry, confocal laser scanning microscopy, ELISA, and gene knock-out mutants. ODT-32 bound to neither GII.4 nor GII.6 P domains, while ODT-54 specifically binds with GII.6 P domain through Psl, an exopolysaccharide encoded by the polysaccharide synthesis locus (psl), identified based on gene annotation, gene transcription, Psl specific staining, and ELISAs. These findings attest that ODT bacteria specifically bind with certain norovirus genotypes in a strain-dependent manner, contributing to a better understanding of the transmission and enrichment of noroviruses in the environment.
Collapse
Affiliation(s)
- Yongxin Yu
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China; Laboratory of Quality and Safety Risk Assessment for Aquatic Products on Storage and Preservation (Shanghai), Ministry of Agriculture, China
| | - Feng Han
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China; Ministry of Agriculture and Rural Affairs, East China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shanghai, China
| | - Mingshu Yang
- College of Food Science and Engineering, Hainan Tropical Ocean University, Sanya, China
| | - Xiaoya Zhang
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Yunfei Chen
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Mingxia Yu
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Yongjie Wang
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China; Laboratory of Quality and Safety Risk Assessment for Aquatic Products on Storage and Preservation (Shanghai), Ministry of Agriculture, China.
| |
Collapse
|
44
|
Nystedt HL, Grønlien KG, Rolfsnes RR, Winther-Larsen HC, Løchen Økstad OA, Tønnesen HH. Neutral natural deep eutectic solvents as anti-biofilm agents. Biofilm 2023; 5:100114. [PMID: 37020863 PMCID: PMC10067762 DOI: 10.1016/j.bioflm.2023.100114] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 03/19/2023] [Accepted: 03/20/2023] [Indexed: 03/28/2023] Open
Abstract
Natural deep eutectic solvents (NADES) are a class of liquids with promising properties as components in pharmaceutical formulations, such as a low toxicity profile, biodegradability and versatility. Recently, their potential use as anti-biofilm agents has been proposed, due to their ability to solubilize and stabilize biological macromolecules. In the current work, the ability to break down biofilm matrix and the biofilm killing activity of three NADES of neutral pH were investigated against Staphylococcus aureus ATCC 6538 and Pseudomonas aeruginosa ATCC 9027 biofilms. The tested NADES were choline chloride:xylitol (ChX), choline chloride:glycerol (ChG) and betaine:sucrose (BS). Two of the NADES (ChX and ChG) significantly reduced the number of remaining viable cells of both bacterial species in pre-formed biofilm by 4-6 orders of magnitude, while the average biofilm biomass removal for all NADES was 27-67% (S. aureus) and 34-49% (P. aeruginosa). The tested NADES also inhibited biofilm formation of both bacterial species at concentrations at or below 0.5 x the minimal inhibitory concentration (MIC), possibly in part due to observed restrictions imposed by NADES on planktonic growth. These results demonstrate the potential value of neutral NADES as anti-biofilm agents in future antimicrobial preparations.
Collapse
Affiliation(s)
- Helene Liepelt Nystedt
- Section for Pharmaceutics and Social Pharmacy, Department of Pharmacy, University of Oslo, P.O. Box 1068 Blindern, NO-0316, Oslo, Norway
| | - Krister Gjestvang Grønlien
- Section for Pharmaceutics and Social Pharmacy, Department of Pharmacy, University of Oslo, P.O. Box 1068 Blindern, NO-0316, Oslo, Norway
| | - Rebekka Rekkedal Rolfsnes
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, P. O. Box 1068 Blindern, NO-0316, Oslo, Norway
| | - Hanne Cecilie Winther-Larsen
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, P. O. Box 1068 Blindern, NO-0316, Oslo, Norway
| | - Ole Andreas Løchen Økstad
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, P. O. Box 1068 Blindern, NO-0316, Oslo, Norway
| | - Hanne Hjorth Tønnesen
- Section for Pharmaceutics and Social Pharmacy, Department of Pharmacy, University of Oslo, P.O. Box 1068 Blindern, NO-0316, Oslo, Norway
| |
Collapse
|
45
|
Guo Y, Mao Z, Ran F, Sun J, Zhang J, Chai G, Wang J. Nanotechnology-Based Drug Delivery Systems to Control Bacterial-Biofilm-Associated Lung Infections. Pharmaceutics 2023; 15:2582. [PMID: 38004561 PMCID: PMC10674810 DOI: 10.3390/pharmaceutics15112582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/09/2023] [Accepted: 10/17/2023] [Indexed: 11/26/2023] Open
Abstract
Airway mucus dysfunction and impaired immunological defenses are hallmarks of several lung diseases, including asthma, cystic fibrosis, and chronic obstructive pulmonary diseases, and are mostly causative factors in bacterial-biofilm-associated respiratory tract infections. Bacteria residing within the biofilm architecture pose a complex challenge in clinical settings due to their increased tolerance to currently available antibiotics and host immune responses, resulting in chronic infections with high recalcitrance and high rates of morbidity and mortality. To address these unmet clinical needs, potential anti-biofilm therapeutic strategies are being developed to effectively control bacterial biofilm. This review focuses on recent advances in the development and application of nanoparticulate drug delivery systems for the treatment of biofilm-associated respiratory tract infections, especially addressing the respiratory barriers of concern for biofilm accessibility and the various types of nanoparticles used to combat biofilms. Understanding the obstacles facing pulmonary drug delivery to bacterial biofilms and nanoparticle-based approaches to combatting biofilm may encourage researchers to explore promising treatment modalities for bacterial-biofilm-associated chronic lung infections.
Collapse
Affiliation(s)
- Yutong Guo
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Zeyuan Mao
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Fang Ran
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Jihong Sun
- Department of Radiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
| | - Jingfeng Zhang
- The Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Ningbo 315000, China
| | - Guihong Chai
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Jian Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510180, China
| |
Collapse
|
46
|
Gheorghita AA, Wozniak DJ, Parsek MR, Howell PL. Pseudomonas aeruginosa biofilm exopolysaccharides: assembly, function, and degradation. FEMS Microbiol Rev 2023; 47:fuad060. [PMID: 37884397 PMCID: PMC10644985 DOI: 10.1093/femsre/fuad060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 10/04/2023] [Accepted: 10/25/2023] [Indexed: 10/28/2023] Open
Abstract
The biofilm matrix is a fortress; sheltering bacteria in a protective and nourishing barrier that allows for growth and adaptation to various surroundings. A variety of different components are found within the matrix including water, lipids, proteins, extracellular DNA, RNA, membrane vesicles, phages, and exopolysaccharides. As part of its biofilm matrix, Pseudomonas aeruginosa is genetically capable of producing three chemically distinct exopolysaccharides - alginate, Pel, and Psl - each of which has a distinct role in biofilm formation and immune evasion during infection. The polymers are produced by highly conserved mechanisms of secretion, involving many proteins that span both the inner and outer bacterial membranes. Experimentally determined structures, predictive modelling of proteins whose structures are yet to be solved, and structural homology comparisons give us insight into the molecular mechanisms of these secretion systems, from polymer synthesis to modification and export. Here, we review recent advances that enhance our understanding of P. aeruginosa multiprotein exopolysaccharide biosynthetic complexes, and how the glycoside hydrolases/lyases within these systems have been commandeered for antimicrobial applications.
Collapse
Affiliation(s)
- Andreea A Gheorghita
- Program in Molecular Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay St, Toronto, ON M5G 0A4, Canada
- Department of Biochemistry, University of Toronto, Medical Science Building, 1 King's College Cir, Toronto, ON M5S 1A8, Canada
| | - Daniel J Wozniak
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine, 776 Biomedical Research Tower, 460 W 12th Ave, Columbus, OH 43210, United States
- Department of Microbiology, The Ohio State University College, Biological Sciences Bldg, 105, 484 W 12th Ave, Columbus, OH 43210, United States
| | - Matthew R Parsek
- Department of Microbiology, University of Washington, Health Sciences Bldg, 1705 NE Pacific St, Seattle, WA 98195-7735, United States
| | - P Lynne Howell
- Program in Molecular Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay St, Toronto, ON M5G 0A4, Canada
- Department of Biochemistry, University of Toronto, Medical Science Building, 1 King's College Cir, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
47
|
Abdelghafar A, El-Ganiny A, Shaker G, Askoura M. A novel lytic phage exhibiting a remarkable in vivo therapeutic potential and higher antibiofilm activity against Pseudomonas aeruginosa. Eur J Clin Microbiol Infect Dis 2023; 42:1207-1234. [PMID: 37608144 PMCID: PMC10511388 DOI: 10.1007/s10096-023-04649-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 08/07/2023] [Indexed: 08/24/2023]
Abstract
BACKGROUND Pseudomonas aeruginosa is a nosocomial bacterium responsible for variety of infections. Inappropriate use of antibiotics could lead to emergence of multidrug-resistant (MDR) P. aeruginosa strains. Herein, a virulent phage; vB_PaeM_PS3 was isolated and tested for its application as alternative to antibiotics for controlling P. aeruginosa infections. METHODS Phage morphology was observed using transmission electron microscopy (TEM). The phage host range and efficiency of plating (EOP) in addition to phage stability were analyzed. One-step growth curve was performed to detect phage growth kinetics. The impact of isolated phage on planktonic cells and biofilms was assessed. The phage genome was sequenced. Finally, the therapeutic potential of vB_PaeM_PS3 was determined in vivo. RESULTS Isolated phage has an icosahedral head and a contractile tail and was assigned to the family Myoviridae. The phage vB_PaeM_PS3 displayed a broad host range, strong bacteriolytic ability, and higher environmental stability. Isolated phage showed a short latent period and large burst size. Importantly, the phage vB_PaeM_PS3 effectively eradicated bacterial biofilms. The genome of vB_PaeM_PS3 consists of 93,922 bp of dsDNA with 49.39% G + C content. It contains 171 predicted open reading frames (ORFs) and 14 genes as tRNA. Interestingly, the phage vB_PaeM_PS3 significantly attenuated P. aeruginosa virulence in host where the survival of bacteria-infected mice was markedly enhanced following phage treatment. Moreover, the colonizing capability of P. aeruginosa was markedly impaired in phage-treated mice as compared to untreated infected mice. CONCLUSION Based on these findings, isolated phage vB_PaeM_PS3 could be potentially considered for treating of P. aeruginosa infections.
Collapse
Affiliation(s)
- Aliaa Abdelghafar
- Department of Microbiology and Immunology, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| | - Amira El-Ganiny
- Department of Microbiology and Immunology, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| | - Ghada Shaker
- Department of Microbiology and Immunology, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| | - Momen Askoura
- Department of Microbiology and Immunology, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt.
| |
Collapse
|
48
|
Chen B, Liu H, Wang Z, Ma J, Shen Y. Effects of DJK-5 and chlorhexidine on exopolysaccharide volume and pH in oral biofilms. BMC Oral Health 2023; 23:705. [PMID: 37777729 PMCID: PMC10544135 DOI: 10.1186/s12903-023-03381-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 09/01/2023] [Indexed: 10/02/2023] Open
Abstract
BACKGROUND Exopolysaccharides (EPS) are essential constituents of the extracellular matrix within oral biofilms and are significantly influenced by the local microenvironment. This study aimed to investigate the impact of two distinct antimicrobial agents, DJK-5 and chlorhexidine (CHX), on the EPS volume and pH levels in oral biofilms. METHODS Oral biofilms obtained from two donors were cultured on hydroxyapatite discs for durations of 3 days, 1 week, 2 weeks, 3 weeks, and 4 weeks. Subsequently, these biofilms were subjected to treatment with 10 µg/mL DJK-5 or 2% CHX for 3 min. The impact of these antimicrobial treatments on factors such as the proportion of dead bacterial, in situ pH, and EPS volume within the biofilms was assessed using corresponding fluorescent probes. The examination was carried out utilizing confocal laser scanning microscopy, and the resulting images were analyzed with a focus on the upper and lower layers of the biofilm, respectively. RESULTS DJK-5 exhibited a more potent bactericidal effect compared to CHX across the 3-day to 4-week duration of the biofilm (P < 0.05). The biofilms were acidic, with the upper layer being less acidic than the lower layer (P < 0.05). Both antimicrobial agents increased the pH, but DJK-5 had a greater effect than CHX (P < 0.05). The volume of EPS was significantly lower in DJK-5 treated biofilms compared to that of CHX, regardless of age or layer (P < 0.05). CONCLUSION DJK-5 exhibited superior effectiveness in reducing viable bacteria and EPS volume, as well as in raising extracellular pH, as compared to chlorhexidine.
Collapse
Affiliation(s)
- Binwen Chen
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
- Department of Oral Biological & Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, Canada
| | - He Liu
- Department of Oral Biological & Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, Canada
| | - Zhejun Wang
- Department of Oral Biological & Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, Canada
| | - Jingzhi Ma
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China.
| | - Ya Shen
- Department of Oral Biological & Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, Canada.
| |
Collapse
|
49
|
El-Sapagh S, El-Shenody R, Pereira L, Elshobary M. Unveiling the Potential of Algal Extracts as Promising Antibacterial and Antibiofilm Agents against Multidrug-Resistant Pseudomonas aeruginosa: In Vitro and In Silico Studies including Molecular Docking. PLANTS (BASEL, SWITZERLAND) 2023; 12:3324. [PMID: 37765485 PMCID: PMC10537748 DOI: 10.3390/plants12183324] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 09/09/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023]
Abstract
Multidrug-resistant Pseudomonas aeruginosa poses a global challenge due to its virulence and biofilm-forming ability, leading to persistent infections. This study had a dual focus: first, it aimed to investigate the biofilm activity and antibiotic resistance profiles of Pseudomonas aeruginosa isolates obtained from a fish-rearing farm. Second, it explored the potential of algal extracts as effective antibacterial and antibiofilm agents. The study analyzed 23 isolates of P. aeruginosa from the farm, assessing antibiotic resistance and biofilm formation. The antimicrobial and antibiofilm activities of two algal extracts, Arthrospira platensis (cyanobacteria) acetone extract (AAE) and Polysiphonia scopulorum (Rhodophyta) methanol extract (PME), were tested individually and combined (COE). The effects on biofilm-related gene expression were examined. AAE, PME, and COE were evaluated for antimicrobial and antibiofilm properties. Biofilm-related gene expression was measured and the extracts were analyzed for physicochemical properties and toxicity. Most P. aeruginosa isolates (86.9%) were antibiotic-resistant and formed biofilms. AAE, PME, and COE displayed promising antibacterial and antibiofilm effects, with COE being particularly effective. COE reduced a key biofilm-related gene expression. The fatty acid content (56% in AAE and 34% in PME) correlated with the effects. Specific compounds, such as phytol, bromophenol, and dihydroxy benzaldehyde, contributed to the activities. The extracts showed favorable characteristics and interactions with FabZ protein amino acids. This study suggests the potential of algal extracts as antibacterial and antibiofilm agents against drug-resistant infections. Further exploration in clinical applications is warranted.
Collapse
Affiliation(s)
- Shimaa El-Sapagh
- Department of Botany and Microbiology, Faculty of Science, Tanta University, Tanta 31527, Egypt; (S.E.-S.); (R.E.-S.)
| | - Rania El-Shenody
- Department of Botany and Microbiology, Faculty of Science, Tanta University, Tanta 31527, Egypt; (S.E.-S.); (R.E.-S.)
| | - Leonel Pereira
- Department of Life Sciences, University of Coimbra, MARE-Marine and Environmental Sciences Centre/ARNET-Aquatic Research Network, 3000-456 Coimbra, Portugal;
| | - Mostafa Elshobary
- Department of Botany and Microbiology, Faculty of Science, Tanta University, Tanta 31527, Egypt; (S.E.-S.); (R.E.-S.)
| |
Collapse
|
50
|
Ghosh M, Raghav S, Ghosh P, Maity S, Mohela K, Jain D. Structural analysis of novel drug targets for mitigation of Pseudomonas aeruginosa biofilms. FEMS Microbiol Rev 2023; 47:fuad054. [PMID: 37771093 DOI: 10.1093/femsre/fuad054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 09/20/2023] [Accepted: 09/27/2023] [Indexed: 09/30/2023] Open
Abstract
Pseudomonas aeruginosa is an opportunistic human pathogen responsible for acute and chronic, hard to treat infections. Persistence of P. aeruginosa is due to its ability to develop into biofilms, which are sessile bacterial communities adhered to substratum and encapsulated in layers of self-produced exopolysaccharides. These biofilms provide enhanced protection from the host immune system and resilience towards antibiotics, which poses a challenge for treatment. Various strategies have been expended for combating biofilms, which involve inhibiting biofilm formation or promoting their dispersal. The current remediation approaches offer some hope for clinical usage, however, treatment and eradication of preformed biofilms is still a challenge. Thus, identifying novel targets and understanding the detailed mechanism of biofilm regulation becomes imperative. Structure-based drug discovery (SBDD) provides a powerful tool that exploits the knowledge of atomic resolution details of the targets to search for high affinity ligands. This review describes the available structural information on the putative target protein structures that can be utilized for high throughput in silico drug discovery against P. aeruginosa biofilms. Integrating available structural information on the target proteins in readily accessible format will accelerate the process of drug discovery.
Collapse
Affiliation(s)
- Moumita Ghosh
- Transcription Regulation Lab, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurugram Expressway, Faridabad, Haryana-121001, India
| | - Shikha Raghav
- Transcription Regulation Lab, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurugram Expressway, Faridabad, Haryana-121001, India
| | - Puja Ghosh
- Transcription Regulation Lab, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurugram Expressway, Faridabad, Haryana-121001, India
| | - Swagatam Maity
- Transcription Regulation Lab, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurugram Expressway, Faridabad, Haryana-121001, India
| | - Kavery Mohela
- Transcription Regulation Lab, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurugram Expressway, Faridabad, Haryana-121001, India
| | - Deepti Jain
- Transcription Regulation Lab, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurugram Expressway, Faridabad, Haryana-121001, India
| |
Collapse
|