1
|
Oladosu VI, Sauer K. FleQ finetunes the expression of a subset of BrlR-activated genes to enable antibiotic tolerance by Pseudomonas aeruginosa biofilms. J Bacteriol 2025; 207:e0050324. [PMID: 40304498 PMCID: PMC12096822 DOI: 10.1128/jb.00503-24] [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: 11/26/2024] [Accepted: 04/03/2025] [Indexed: 05/02/2025] Open
Abstract
The transcriptional regulator FleQ contributes to Pseudomonas aeruginosa biofilm formation by activating the expression and biosynthesis of matrix exopolysaccharides in a manner dependent on c-di-GMP. However, little is known about the role of FleQ in the antibiotic tolerance phenotype of P. aeruginosa biofilms. Inactivation of fleQ impaired biofilm formation and rendered biofilms susceptible to tobramycin and norfloxacin. The phenotypes were similar to biofilms inactivated in sagS encoding the orphan sensor SagS that promotes the switch from planktonic to biofilm growth via BfiSR and antibiotic tolerance via BrlR. While FleQ was found to contribute to biofilm formation independently of SagS and BfiSR, FleQ instead converged with SagS-dependent regulation at the level of BrlR. This was supported by multicopy expression of sagS failing to restore biofilm antibiotic tolerance by ΔfleQ to wild-type levels (and vice versa) and by biofilms formed by the ΔfleQΔsagS double mutant being as susceptible as ΔfleQ and ΔsagS biofilms. Increased antibiotic susceptibility was independent of BrlR abundance or BrlR DNA binding but coincided with significantly reduced transcript abundance of the BrlR-activated mexCD-oprJ and PA1874-77, encoding an ABC transporter previously shown to contribute to the tolerance of biofilms to tobramycin and norfloxacin. FleQ- dependent regulation of gene expression was indirect. Co-immunoprecipitation and BACTH assays indicated FleQ to interact with SagS via its HisKA-Rec domain, likely suggesting FleQ and SagS to likely work in concert to enable biofilm antibiotic tolerance, by finetuning the expression of BrlR activated genes.IMPORTANCEIn P. aeruginosa, FleQ inversely regulates the expression of genes encoding flagella and biofilm matrix components, including exopolysaccharide (Pel, Psl) in a manner dependent on the levels of c-di-GMP. Our findings expand on the role of FleQ from regulating the transition to the biofilm mode of growth to FleQ contributing to the antimicrobial tolerance phenotype of biofilms, by FleQ affecting the expression of PA1874-77, a downstream target of the SagS-dependent transcriptional regulator BrlR. Importantly, our findings suggest FleQ works in concert with SagS, likely via FleQ-SagS protein-protein interactions, to enable the formation of inherently tolerant P. aeruginosa biofilms.
Collapse
Affiliation(s)
- Victoria I. Oladosu
- 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
|
2
|
Volk M, Gundogdu O, Klančnik A. Temporal dynamics of gene expression during the development of Campylobacter jejuni biofilms. Microb Genom 2025; 11:001387. [PMID: 40327030 PMCID: PMC12056249 DOI: 10.1099/mgen.0.001387] [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: 12/03/2024] [Accepted: 02/26/2025] [Indexed: 05/07/2025] Open
Abstract
Campylobacter jejuni, an important pathogen of bacterial gastrointestinal infections, forms biofilms that enable its survival in different environments. C. jejuni biofilm development is still poorly understood, and thus, in this study, we characterized gene expression changes at different biofilm stages using RNA sequencing. Early biofilms (after 16 and 24 h) showed increased expressions of genes involved in cysteine and methionine metabolism, whereas mature biofilms (after 48 and 72 h) showed decreased expression of genes encoding capsular polysaccharides and lipooligosaccharides. Both early and mature biofilms showed increased expressions of genes involved in flagella formation, leucine metabolism and the oxidative stress response and decreased expressions of genes involved in energy metabolism, iron acquisition and transmembrane transport. This study provides insights into the molecular mechanisms underlying C. jejuni biofilm maturation, environmental resistance and the dynamic nature of gene expression during biofilm development.
Collapse
Affiliation(s)
- Manca Volk
- Department of Food Science and Technology, Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Ozan Gundogdu
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK
| | - Anja Klančnik
- Department of Food Science and Technology, Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
3
|
Salem MH, Azmy AF, Dishisha T, Dessouky N. Pseudomonas aeruginosa clinical isolates in Egypt: phenotypic, genotypic, and antibiofilm assessment of Pluronic F-127. BMC Microbiol 2025; 25:245. [PMID: 40281406 PMCID: PMC12023487 DOI: 10.1186/s12866-025-03946-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 04/02/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Virulence factors play an important role in developing bacterial resistance leading to the increased severity of Pseudomonas aeruginosa infections. Several genes encoding for virulence factors is coordinated by the quorum sensing (QS) system. In the present study, the prevalence of virulence genes, particularly those involved in controlling biofilm formation, and their correlation with antibiotic resistance patterns was investigated. The ability of the pathogens to form biofilm and the impact of Pluronic F-127 as a potential biofilm inhibitor was assessed. RESULTS A total of 118 P. aeruginosa clinical isolates were collected. The highest resistance rates were observed against ceftazidime (94%), while colistin was the most effective followed by polymyxin B with sensitivity rate 72% and 59%, respectively. Out of 118 isolates: 111 (94%) were biofilm producers, 24.6% of them were strong. The QS genes; lasR and rhlR, were detected in 85% and 89% of the isolates, respectively, toxA gene in 95% and ampC gene in 69% of the isolates. Pluronic F-127 was confirmed as a biofilm inhibitor in lowest concentration used 1.25 mg/ml which inhibits 78% of strong biofilm forming isolates and has better effect on detachment of established biofilm by 90% of biofilm forming isolates. CONCLUSION The ability of bacteria to form biofilms contributes greatly to the development of antibiotic resistance, which leads to the occurrence of persistent and chronic bacterial illnesses. Many isolates exhibited moderate to strong biofilm forming ability, which showed a high resistance pattern. The results demonstrated that Pluronic F-127 has a promising level of biofilm inhibition and detachment in most isolates. It has a chance to serve as a substitute means for combating biofilm formation.
Collapse
Affiliation(s)
- Mai Hamed Salem
- Department of Microbiology and Immunology, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Sciences and Technology, P.O. Box 77, Giza, Egypt.
| | - Ahmed F Azmy
- Department of Pharmaceutical Microbiology and Immunology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, 62511, Egypt
| | - Tarek Dishisha
- Department of Pharmaceutical Microbiology and Immunology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, 62511, Egypt
| | - Nesrein Dessouky
- Department of Microbiology and Immunology, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Sciences and Technology, P.O. Box 77, Giza, Egypt
| |
Collapse
|
4
|
Lavoie T, Daffinee KE, Vicent ML, LaPlante KL. Staphylococcus biofilm dynamics and antibiotic resistance: insights into biofilm stages, zeta potential dynamics, and antibiotic susceptibility. Microbiol Spectr 2025; 13:e0291524. [PMID: 40135862 PMCID: PMC12054104 DOI: 10.1128/spectrum.02915-24] [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: 12/10/2024] [Accepted: 01/16/2025] [Indexed: 03/27/2025] Open
Abstract
Staphylococcus spp. infections often involve biofilms, but standard antibiotic minimum inhibitory concentration (MIC) testing used to determine treatment evaluates planktonic bacterial growth only and does not account for biofilm presence, strength, or growth stage. To aid in determining a cost-effective method to solve this issue, we built upon in vitro methods initially published by Stepanovic et al. used to determine weak and strong biofilm formations. First, we determined 115 unique S. aureus isolate biofilms at 2, 4, 6, 8, 16, and 24 h to classify the hourly stages of biofilm development based on statistically significant final growth results (P < 0.001): stages one (0-6 h), two (6-16 h), three (16-24 h), and four (>24 h). Next, to further evaluate in vitro biofilm strength, electrostatic differences were measured through zeta (ζ)-potential for strong and weak biofilm producers at early and late stage-formed biofilms. The early stages of weak biofilm formers had a greater negative electrostatic charge when compared to strong biofilm formers. Meanwhile, strong biofilm formers began early stages with less negative charges before increasing the negative electrostatic charge by stage-four biofilm. At all time points, weak biofilm-forming isolate mean ζ-potentials were significantly more negative than strong biofilm formers (P = ≤0.04). Finally, to elucidate minimum eradication concentrations for biofilms, we treated stage-four biofilms with progressively higher concentrations of either daptomycin, vancomycin, or levofloxacin. Daptomycin was the only antibiotic to achieve ≥75% reduction in biofilm viability, seen at 32-256 μg/mL (64-512× MIC), and significantly reduced residual biofilm across all strong and weak biofilms. Biofilm findings showed an unexpected initial biofilm decrease in response to lower concentrations of antibiotics, followed by an increase in biofilm biomass at higher antibiotic concentrations. While higher antibiotic concentrations can be used to overcome bacterial resistance and eliminate infections, our results suggest that antimicrobial resistance is observed, regardless of bacterial biofilm strength, and that there may be an optimal treatment concentration window for achieving maximum kill. Our data add to the increasing evidence of biofilms' role in recurrent infections and the importance of antibiotic concentration.IMPORTANCEThis work is significant, as it addresses a critical gap in standard antibiotic testing by focusing on the unique characteristics of biofilm-forming Staphylococcus aureus infections, which are major contributors to recurrent and chronic infections. Unlike traditional MIC testing that evaluates planktonic bacteria, this study emphasizes the importance of biofilm presence, growth stages, and electrostatic properties in determining treatment strategies. By classifying biofilm development into distinct stages in an easily reproducible assay and measuring the biofilm zeta-potential for key differences and overall biofilm response to multiple standard antibiotic concentrations, this research provides valuable insights for the future of biofilm in vitro work. Furthermore, it highlights the efficacy of daptomycin in eradicating biofilm while identifying possibilities of optimal antibiotic concentration windows, a critical consideration for mitigating resistance and achieving effective infection control. These findings underscore the necessity of tailoring treatment to biofilm-specific dynamics, offering a path toward more effective therapeutic approaches for biofilm-associated infections.
Collapse
Affiliation(s)
- T. Lavoie
- College of Pharmacy, University of Rhode Island, Kingston, Rhode Island, USA
- Providence Veterans Affairs Medical Center, Providence, Rhode Island, USA
| | - K. E. Daffinee
- Providence Veterans Affairs Medical Center, Providence, Rhode Island, USA
| | - M. L. Vicent
- College of Pharmacy, University of Rhode Island, Kingston, Rhode Island, USA
- Providence Veterans Affairs Medical Center, Providence, Rhode Island, USA
| | - K. L. LaPlante
- College of Pharmacy, University of Rhode Island, Kingston, Rhode Island, USA
- Providence Veterans Affairs Medical Center, Providence, Rhode Island, USA
- Division of Infectious Diseases, Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| |
Collapse
|
5
|
Amann V, Kissmann AK, Firacative C, Rosenau F. Biofilm-Associated Candidiasis: Pathogenesis, Prevalence, Challenges and Therapeutic Options. Pharmaceuticals (Basel) 2025; 18:460. [PMID: 40283897 PMCID: PMC12030374 DOI: 10.3390/ph18040460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 03/11/2025] [Accepted: 03/21/2025] [Indexed: 04/29/2025] Open
Abstract
The rising prevalence of fungal infections, especially those caused by Candida species, presents a major risk to global health. With approximately 1.5 million deaths annually, the urgency for effective treatment options has never been greater. Candida spp. are the leading cause of invasive infections, significantly impacting immunocompromised patients and those in healthcare settings. C. albicans, C. parapsilosis and the emerging species C. auris are categorized as highly dangerous species because of their pathogenic potential and increasing drug resistance. This review comparatively describes the formation of microbial biofilms of both bacterial and fungal origin, including major pathogens, thereby creating a novel focus. Biofilms can further complicate treatment, as these structures provide enhanced resistance to antifungal therapies. Traditional antifungal agents, including polyenes, azoles and echinocandins, have shown effectiveness, yet resistance development continues to rise, necessitating the exploration of novel therapeutic approaches. Antimicrobial peptides (AMPs) such as the anti-biofilm peptides Pom-1 and Cm-p5 originally isolated from snails represent promising candidates due to their unique mechanisms of action and neglectable cytotoxicity. This review article discusses the challenges posed by Candida infections, the characteristics of important species, the role of biofilms in virulence and the potential of new therapeutic options like AMPs.
Collapse
Affiliation(s)
- Valerie Amann
- Institute of Pharmaceutical Biotechnology, Ulm University, 89081 Ulm, Germany; (V.A.); (A.-K.K.)
| | - Ann-Kathrin Kissmann
- Institute of Pharmaceutical Biotechnology, Ulm University, 89081 Ulm, Germany; (V.A.); (A.-K.K.)
| | - Carolina Firacative
- Studies in Translational Microbiology and Emerging Diseases (MICROS) Research Group, School of Medicine and Health Sciences, Universidad del Rosario, Bogota 111221, Colombia;
| | - Frank Rosenau
- Institute of Pharmaceutical Biotechnology, Ulm University, 89081 Ulm, Germany; (V.A.); (A.-K.K.)
| |
Collapse
|
6
|
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
|
7
|
Nour El-Din HT, Kettal M, Granados Maciel JC, Beaudoin G, Oktay U, Hrapovic S, Sad S, Dennis JJ, Peters DL, Chen W. Isolation, Characterization, and Genomic Analysis of Bacteriophages Against Pseudomonas aeruginosa Clinical Isolates from Early and Chronic Cystic Fibrosis Patients for Potential Phage Therapy. Microorganisms 2025; 13:511. [PMID: 40142404 PMCID: PMC11945000 DOI: 10.3390/microorganisms13030511] [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: 02/07/2025] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 03/28/2025] Open
Abstract
Pseudomonas aeruginosa is associated with both community and hospital-acquired infections. It colonizes the lungs of cystic fibrosis (CF) patients, establishing an ecological niche where it adapts and evolves from early to chronic stages, resulting in deteriorating lung function and frequent exacerbations. With antibiotics resistance on the rise, there is a pressing need for alternative personalized treatments (such as bacteriophage therapy) to combat P. aeruginosa infections. In this study, we aimed to isolate and characterize phages targeting both early and chronic P. aeruginosa isolates and evaluate their potential for phage therapy. Four highly virulent phages belonging to myoviral, podviral, and siphoviral morphotypes were isolated from sewage samples. These phages have a broad host range and effectively target 62.5% of the P. aeruginosa isolates with a positive correlation to the early isolates. All the phages have a virulence index of ≥0.90 (0.90-0.98), and one has a large burst size of 331 PFU/cell and a latency period of 30 min. All phages are stable under a wide range of temperature and pH conditions. Genomic analysis suggests the four phages are strictly lytic and devoid of identifiable temperate phage repressors and genes associated with antibiotic resistance and virulence. More significantly, two of the phages significantly delayed the onset of larval death when evaluated in a lethal Galleria mellonella infection model, suggesting their promise as phage therapy candidates for P. aeruginosa infections.
Collapse
Affiliation(s)
- Hanzada T. Nour El-Din
- Department of Immunobiology, Human Health Therapeutics Research Center, National Research Council Canada, Ottawa, ON K1N 5A2, Canada; (M.K.); (J.C.G.M.); (G.B.); (D.L.P.); (W.C.)
| | - Maryam Kettal
- Department of Immunobiology, Human Health Therapeutics Research Center, National Research Council Canada, Ottawa, ON K1N 5A2, Canada; (M.K.); (J.C.G.M.); (G.B.); (D.L.P.); (W.C.)
| | - José C. Granados Maciel
- Department of Immunobiology, Human Health Therapeutics Research Center, National Research Council Canada, Ottawa, ON K1N 5A2, Canada; (M.K.); (J.C.G.M.); (G.B.); (D.L.P.); (W.C.)
| | - Greg Beaudoin
- Department of Immunobiology, Human Health Therapeutics Research Center, National Research Council Canada, Ottawa, ON K1N 5A2, Canada; (M.K.); (J.C.G.M.); (G.B.); (D.L.P.); (W.C.)
| | - Umut Oktay
- Faculty of Science, Biological Sciences, University of Alberta, Edmonton, AB T6G 2R3, Canada; (U.O.); (J.J.D.)
| | - Sabahudin Hrapovic
- Aquatic and Crop Resource Development (ACRD) Research Center, National Research Council Canada, Montreal, QC H4P 2R2, Canada;
| | - Subash Sad
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1N 6N5, Canada;
| | - Jonathan J. Dennis
- Faculty of Science, Biological Sciences, University of Alberta, Edmonton, AB T6G 2R3, Canada; (U.O.); (J.J.D.)
| | - Danielle L. Peters
- Department of Immunobiology, Human Health Therapeutics Research Center, National Research Council Canada, Ottawa, ON K1N 5A2, Canada; (M.K.); (J.C.G.M.); (G.B.); (D.L.P.); (W.C.)
| | - Wangxue Chen
- Department of Immunobiology, Human Health Therapeutics Research Center, National Research Council Canada, Ottawa, ON K1N 5A2, Canada; (M.K.); (J.C.G.M.); (G.B.); (D.L.P.); (W.C.)
- Department of Biology, Brock University, St. Catharines, ON L2S 3A1, Canada
| |
Collapse
|
8
|
Buder C, Langkabel N, Kirse A, Kalusa M, Fietz SA, Meemken D. Nano-coating with silicon dioxide to reduce the occurrence of bacterial contamination in a pig abattoir drinking system. Folia Microbiol (Praha) 2025:10.1007/s12223-025-01243-x. [PMID: 39904879 DOI: 10.1007/s12223-025-01243-x] [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: 09/02/2024] [Accepted: 01/14/2025] [Indexed: 02/06/2025]
Abstract
A recently discovered source for infection of slaughter pigs, and thus entry for bacteria into the food chain, is the installed drinking equipment in lairage pens of pig abattoirs. To mitigate this, nano-coating of stainless steel, currently used in human medicine fields as well as in other parts of the food chain, appears as promising technology. In this study, silicon dioxide nano-coating was applied to six drinkers and installed for one and three months in a lairage of a pig abattoir, while results were compared with those of drinkers that had not been nano-coated. Laboratory examination of eight sample types related to the drinkers was conducted for total aerobic plate count, Enterobacteriaceae count, Pseudomonas spp. count, Salmonella presence, pathogenic Yersinia enterocolitica presence, Listeria monocytogenes presence and methicillin-resistant Staphylococcus aureus presence. The nipple drinker, which the pigs take into their mouth for drinking, was then examined using scanning electron microscopy and elemental analysis. The nano-coating did not produce statistically significant reductions in the loads or presence of these bacteria compared to the same but uncoated drinking equipment used under the same conditions. Further studies should focus on the implementation of combined methods, such as nano-coating and sanitary treatment, as well as modifications to the coating itself, to produce meaningful reductions of the bacterial loads on/in abattoir lairage drinking equipment.
Collapse
Affiliation(s)
- Celine Buder
- Institute of Food Safety and Food Hygiene, Working Group Meat Hygiene, School of Veterinary Medicine, Freie Universität Berlin, 14163, Berlin, Germany.
- Veterinary Centre for Resistance Research, School of Veterinary Medicine, Freie Universität Berlin, 14163, Berlin, Germany.
| | - Nina Langkabel
- Institute of Food Safety and Food Hygiene, Working Group Meat Hygiene, School of Veterinary Medicine, Freie Universität Berlin, 14163, Berlin, Germany
- Veterinary Centre for Resistance Research, School of Veterinary Medicine, Freie Universität Berlin, 14163, Berlin, Germany
| | - Alina Kirse
- Institute of Biometry, Epidemiology and Information Processing, WHO Collaborating Centre for Research and Training for Health at the Human-Animal-Environment Interface, University of Veterinary Medicine Hannover, 30559, Hannover, Germany
| | - Mirjam Kalusa
- Institute of Veterinary Anatomy, Histology and Embryology, Faculty of Veterinary Medicine, University of Leipzig, 04103, Leipzig, Germany
| | - Simone A Fietz
- Institute of Veterinary Anatomy, Histology and Embryology, Faculty of Veterinary Medicine, University of Leipzig, 04103, Leipzig, Germany
| | - Diana Meemken
- Institute of Food Safety and Food Hygiene, Working Group Meat Hygiene, School of Veterinary Medicine, Freie Universität Berlin, 14163, Berlin, Germany
- Veterinary Centre for Resistance Research, School of Veterinary Medicine, Freie Universität Berlin, 14163, Berlin, Germany
| |
Collapse
|
9
|
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
|
10
|
Gonzalez La Corte S, Stevens CA, Cárcamo-Oyarce G, Ribbeck K, Wingreen NS, Datta SS. Morphogenesis of bacterial cables in polymeric environments. SCIENCE ADVANCES 2025; 11:eadq7797. [PMID: 39823332 PMCID: PMC11740958 DOI: 10.1126/sciadv.adq7797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 12/17/2024] [Indexed: 01/19/2025]
Abstract
Many bacteria live in polymeric fluids, such as mucus, environmental polysaccharides, and extracellular polymers in biofilms. However, laboratory studies typically focus on cells in polymer-free fluids. Here, we show that interactions with polymers shape a fundamental feature of bacterial life-how they proliferate in space in multicellular colonies. Using experiments, we find that when polymer is sufficiently concentrated, cells generically and reversibly form large serpentine "cables" as they proliferate. By combining experiments with biophysical theory and simulations, we demonstrate that this distinctive form of colony morphogenesis arises from an interplay between polymer-induced entropic attraction between neighboring cells and their hindered ability to diffusely separate from each other in a viscous polymer solution. Our work thus reveals a pivotal role of polymers in sculpting proliferating bacterial colonies, with implications for how they interact with hosts and with the natural environment, and uncovers quantitative principles governing colony morphogenesis in such complex environments.
Collapse
Affiliation(s)
| | - Corey A. Stevens
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Gerardo Cárcamo-Oyarce
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Facultad de Ciencias Biológicas, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Katharina Ribbeck
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ned S. Wingreen
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Sujit S. Datta
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, USA
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, USA
| |
Collapse
|
11
|
Roque‐Borda CA, Primo LMDG, Medina‐Alarcón KP, Campos IC, Nascimento CDF, Saraiva MMS, Berchieri Junior A, Fusco‐Almeida AM, Mendes‐Giannini MJS, Perdigão J, Pavan FR, Albericio F. Antimicrobial Peptides: A Promising Alternative to Conventional Antimicrobials for Combating Polymicrobial Biofilms. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410893. [PMID: 39530703 PMCID: PMC11714181 DOI: 10.1002/advs.202410893] [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: 09/06/2024] [Revised: 10/23/2024] [Indexed: 11/16/2024]
Abstract
Polymicrobial biofilms adhere to surfaces and enhance pathogen resistance to conventional treatments, significantly contributing to chronic infections in the respiratory tract, oral cavity, chronic wounds, and on medical devices. This review examines antimicrobial peptides (AMPs) as a promising alternative to traditional antibiotics for treating biofilm-associated infections. AMPs, which can be produced as part of the innate immune response or synthesized therapeutically, have broad-spectrum antimicrobial activity, often disrupting microbial cell membranes and causing cell death. Many specifically target negatively charged bacterial membranes, unlike host cell membranes. Research shows AMPs effectively inhibit and disrupt polymicrobial biofilms and can enhance conventional antibiotics' efficacy. Preclinical and clinical research is advancing, with animal studies and clinical trials showing promise against multidrug-resistant bacteria and fungi. Numerous patents indicate increasing interest in AMPs. However, challenges such as peptide stability, potential cytotoxicity, and high production costs must be addressed. Ongoing research focuses on optimizing AMP structures, enhancing stability, and developing cost-effective production methods. In summary, AMPs offer a novel approach to combating biofilm-associated infections, with their unique mechanisms and synergistic potential with existing antibiotics positioning them as promising candidates for future treatments.
Collapse
Affiliation(s)
- Cesar Augusto Roque‐Borda
- Department of Biological SciencesSchool of Pharmaceutical SciencesUniversidade Estadual Paulista (UNESP)AraraquaraSao Paulo14800‐903Brazil
- iMed.ULisboa–Institute for Medicines ResearchFaculty of PharmacyUniversity of LisbonLisbon1649004Portugal
- Vicerrectorado de InvestigaciónUniversidad Católica de Santa MaríaArequipa04000Peru
| | - Laura Maria Duran Gleriani Primo
- Department of Biological SciencesSchool of Pharmaceutical SciencesUniversidade Estadual Paulista (UNESP)AraraquaraSao Paulo14800‐903Brazil
| | - Kaila Petronila Medina‐Alarcón
- Department of Clinical AnalysisSchool of Pharmaceutical SciencesUniversidade Estadual Paulista (UNESP)AraraquaraSao Paulo14800‐903Brazil
| | - Isabella C. Campos
- São Paulo State University (UNESP)School of Agricultural and Veterinarian SciencesJaboticabalSao Paulo14884‐900Brazil
| | - Camila de Fátima Nascimento
- São Paulo State University (UNESP)School of Agricultural and Veterinarian SciencesJaboticabalSao Paulo14884‐900Brazil
| | - Mauro M. S. Saraiva
- São Paulo State University (UNESP)School of Agricultural and Veterinarian SciencesJaboticabalSao Paulo14884‐900Brazil
| | - Angelo Berchieri Junior
- São Paulo State University (UNESP)School of Agricultural and Veterinarian SciencesJaboticabalSao Paulo14884‐900Brazil
| | - Ana Marisa Fusco‐Almeida
- Department of Clinical AnalysisSchool of Pharmaceutical SciencesUniversidade Estadual Paulista (UNESP)AraraquaraSao Paulo14800‐903Brazil
| | - Maria José Soares Mendes‐Giannini
- Department of Clinical AnalysisSchool of Pharmaceutical SciencesUniversidade Estadual Paulista (UNESP)AraraquaraSao Paulo14800‐903Brazil
| | - João Perdigão
- iMed.ULisboa–Institute for Medicines ResearchFaculty of PharmacyUniversity of LisbonLisbon1649004Portugal
| | - Fernando Rogério Pavan
- Department of Biological SciencesSchool of Pharmaceutical SciencesUniversidade Estadual Paulista (UNESP)AraraquaraSao Paulo14800‐903Brazil
| | - Fernando Albericio
- Peptide Science LaboratorySchool of Chemistry and PhysicsUniversity of KwaZulu‐NatalDurban4001South Africa
- CIBER‐BBNNetworking Centre on BioengineeringBiomaterials and Nanomedicineand Department of Organic ChemistryUniversity of BarcelonaBarcelona08028Spain
| |
Collapse
|
12
|
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
|
13
|
Omwenga EO, Awuor SO. The Bacterial Biofilms: Formation, Impacts, and Possible Management Targets in the Healthcare System. THE CANADIAN JOURNAL OF INFECTIOUS DISEASES & MEDICAL MICROBIOLOGY = JOURNAL CANADIEN DES MALADIES INFECTIEUSES ET DE LA MICROBIOLOGIE MEDICALE 2024; 2024:1542576. [PMID: 39717533 PMCID: PMC11666319 DOI: 10.1155/cjid/1542576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 11/04/2024] [Accepted: 11/18/2024] [Indexed: 12/25/2024]
Abstract
Introduction: The persistent increase in multidrug-resistant pathogens has catalyzed the creation of novel strategies to address antivirulence and anti-infective elements. Such methodologies aim to diminish the selective pressure exerted on bacterial populations, decreasing the likelihood of resistance emergence. This review explores the role of biofilm formation as a significant virulence factor and its impact on the development of antimicrobial resistance (AMR). Case Presentation: The ability of bacteria to form a superstructure-biofilm-has made resistance cases in the microbial world a big concern to public health and other sectors as it is a crucial virulence factor that causes difficulties in the management of infections, hence enhancing chronic infection occurrence. Biofilm formation dates to about 3.4 billion years when prokaryotes were discovered to be forming them and since then due to evolution and growth in science, they are more understood. Management and Outcome: The unique microenvironments within bacterial biofilms diminish antibiotic effectiveness and help bacteria evade the host immune system. Biofilm production is a widespread capability among diverse bacterial species. Biofilm formation is enhanced by quorum sensing (QS), reduction of nutrients, or harsh environments for the bacteria. Conclusion: The rise of severe, treatment-resistant biofilm infections poses major challenges in medicine and agriculture, yet much about how these biofilms form remains unknown.
Collapse
Affiliation(s)
- Eric Omori Omwenga
- Department of Medical Microbiology & Parasitology, School of Health Sciences, Kisii University, Kisii, Kenya
| | - Silas Onyango Awuor
- Department of Applied Health Sciences, School of Health Sciences, Kisii University, Kisii, Kenya
- Department of Medical Microbiology, Jaramogi Oginga Odinga Teaching and Referral Hospital, Kisumu, Kenya
| |
Collapse
|
14
|
Sandbakken ET, Høyer E, Witsø E, Søgaard CK, Díez-Sánchez A, Hoang L, Wik TS, Bergh K. Biofilm and the effect of sonication in a chronic Staphylococcus epidermidis orthopedic in vivo implant infection model. J Orthop Surg Res 2024; 19:820. [PMID: 39633500 PMCID: PMC11616123 DOI: 10.1186/s13018-024-05309-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 11/24/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND In diagnosing chronic orthopedic implant infections culture of sonicate represents a supplement to tissue cultures. However, the extent to which biofilm forms on implant surfaces and the degree of dislodgement of bacteria by sonication remains unclear. In this in vivo study using a low bacterial inoculum, we aimed to determine whether a variable effect of sonication could be observed in a standardized in vivo model. MATERIALS AND METHODS Seven Wistar rats underwent surgery with intramuscular implantation of two bone xenograft implants, each containing two steel plates. The grafts were inoculated with approximately 500 colony forming units (CFU) of Staphylococcus epidermidis ATCC 35984. After 20 days the rats were sacrificed, and the steel plates were removed from the bone grafts. Epifluorescence microscopy and scanning electron microscopy (SEM) were used to visualize biofilm formation and dislodgement on the plate surfaces. In addition to cultures of sonicate, a quantitative S. epidermidis specific PCR was developed for enumeration of bacteria. RESULTS A chronic, low-grade implant infection was successfully established, with all animals remaining in good health. All infected bone graft implants yielded abundant growth of S. epidermidis, with a median of 3.25 (1.6-4.6) × 10⁷ CFU per/graft. We were unable to distinguish infected plates from negative controls using epifluorescence microscopy. On infected plates small colonies of staphylococci were identified by SEM. The number of bacteria detected in the sonicate was low with 500 (100-2400) CFU/plate and 475 (140-1821) copies/plate by qPCR. The difference in area covered by fluorescent material before and after sonication was 10.1 (5.7-12.3) %, p = 0.018. CONCLUSION Despite the pronounced infection in the surrounding tissue, only few bacteria were detected on the surface of the steel implants. This is evident from the minimal findings by SEM before sonication, as well as the very low CFU counts and DNA copies in the sonicate. Sonication did not show variable effectiveness, indicating it is a valuable addition to, but not a replacement for biopsy cultures in cases of implant-associated infections with low-virulence microorganisms.
Collapse
Affiliation(s)
- Erik Thorvaldsen Sandbakken
- Department of Orthopedic Surgery, St. Olavs Hospital, Olav Kyrres Gate 13, 7030, Trondheim, Norway.
- Department of Neuromedicine and Movement Science, NTNU, Olav Kyrres Gate 13, 7030, Trondheim, Norway.
| | - Erling Høyer
- Departement of Medical Microbiology, St. Olavs Hospital, Erling Skjalgsons Gate 1, 7030, Trondheim, Norway
| | - Eivind Witsø
- Department of Orthopedic Surgery, St. Olavs Hospital, Olav Kyrres Gate 13, 7030, Trondheim, Norway
| | - Caroline Krogh Søgaard
- Department of Clinical and Molecular Medicine, NTNU, Erling Skjalgsons Gate 1, 7030, Trondheim, Norway
| | - Alberto Díez-Sánchez
- Cellular and Molecular Imaging Core Facility (CMIC), NTNU, Olav Kyrres Gate 10, 7030, Trondheim, Norway
| | - Linh Hoang
- Cellular and Molecular Imaging Core Facility (CMIC), NTNU, Olav Kyrres Gate 10, 7030, Trondheim, Norway
| | - Tina Strømdal Wik
- Department of Orthopedic Surgery, St. Olavs Hospital, Olav Kyrres Gate 13, 7030, Trondheim, Norway
- Department of Neuromedicine and Movement Science, NTNU, Olav Kyrres Gate 13, 7030, Trondheim, Norway
| | - Kåre Bergh
- Departement of Medical Microbiology, St. Olavs Hospital, Erling Skjalgsons Gate 1, 7030, Trondheim, Norway
- Department of Clinical and Molecular Medicine, NTNU, Erling Skjalgsons Gate 1, 7030, Trondheim, Norway
| |
Collapse
|
15
|
Goetz C, Sanschagrin L, Jubinville E, Jacques M, Jean J. Recent progress in antibiofilm strategies in the dairy industry. J Dairy Sci 2024:S0022-0302(24)01335-3. [PMID: 39603496 DOI: 10.3168/jds.2024-25554] [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: 08/06/2024] [Accepted: 11/01/2024] [Indexed: 11/29/2024]
Abstract
Biofilm formation allows microorganisms including bacteria to persist on abiotic or biotic surfaces, to resist treatments with biocides (disinfectants and antibiotics) and to evade the immune response in animal hosts much more than they do in the planktonic form. Bacteria able to form biofilm can be troublesome in the dairy industry, both by causing clinical symptoms in livestock and by colonizing milking devices and milk processing equipment, resulting in dairy products of lower quality and sometimes raising serious food safety issues. In fact, most of the bacterial species isolated frequently in the dairy chain have the ability to form biofilm. Common examples include Staphylococcus aureus and other staphylococci that frequently infect mammary glands, but also Bacillus spp., Listeria monocytogenes and Pseudomonas spp. which cause spoilage of dairy products and sometimes foodborne illnesses. The economic losses due to biofilm formation in the dairy industry are considerable, and scientists are constantly solicited to develop new antibiofilm strategies, especially using biocides of natural origin. Although the number of studies in this subject area has exploded in recent years, the in vivo efficacy of most novel approaches remains to be explored. Used alone or to increase the efficacy of disinfectants or antibiotics, they could allow the implementation of strategies having less impact on the environment. Their use is expected to lead to less reliance on antibiotics to treat intramammary infections in dairy farms and to the use of lower concentrations of chemical disinfectants in dairy processing plants.
Collapse
Affiliation(s)
- Coralie Goetz
- INRAE, L'Institut Agro Rennes-Angers, UMR 1253 STLO, Rennes Cedex, France
| | - Laurie Sanschagrin
- Département des sciences des aliments, Institut sur la Nutrition et les Aliments Fonctionnels, Université Laval, Québec, QC, Canada
| | - Eric Jubinville
- Département des sciences des aliments, Institut sur la Nutrition et les Aliments Fonctionnels, Université Laval, Québec, QC, Canada
| | - Mario Jacques
- Regroupement de recherche pour un lait de qualité optimale (Op+lait), Faculté de médecine vétérinaire, Université de Montréal, St Hyacinthe, QC, Canada
| | - Julie Jean
- Département des sciences des aliments, Institut sur la Nutrition et les Aliments Fonctionnels, Université Laval, Québec, QC, Canada.
| |
Collapse
|
16
|
Osiro KO, Hashemi N, Brango-Vanegas J, Oliveira SMD, Franco OL. Emerging peptide-based technology for biofilm control. Expert Opin Biol Ther 2024:1-5. [PMID: 39548688 DOI: 10.1080/14712598.2024.2430623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 11/04/2024] [Accepted: 11/13/2024] [Indexed: 11/18/2024]
Affiliation(s)
- Karen O Osiro
- Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, Brazil
| | - Nona Hashemi
- Joint School of Nanoscience and Nanoengineering, North Carolina A&T State University, Greensboro, NC, USA
| | - José Brango-Vanegas
- Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, Brazil
- S-Inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Brazil
| | - Samuel M D Oliveira
- Joint School of Nanoscience and Nanoengineering, North Carolina A&T State University, Greensboro, NC, USA
| | - Octavio L Franco
- Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, Brazil
- S-Inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Brazil
| |
Collapse
|
17
|
Straub J, Baertl S, Verheul M, Walter N, Wong RMY, Alt V, Rupp M. Antimicrobial resistance: Biofilms, small colony variants, and intracellular bacteria. Injury 2024; 55 Suppl 6:111638. [PMID: 39482024 DOI: 10.1016/j.injury.2024.111638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 05/25/2024] [Accepted: 05/27/2024] [Indexed: 11/03/2024]
Abstract
Soft tissue and bone infections continue to be a serious complication in orthopedic and trauma surgery. Both can lead to a high burden for the patients and the healthcare system. Musculoskeletal infections can be induced by intraoperative contamination, bacterial contamination of open wounds or hematogenous bacterial spread. During the recent decades, advances were achieved in the understanding of pathogenesis and antibiotic resistance. Despite some progress in the diagnosis and advancing of therapeutic concepts, groundbreaking successful improvement of treatment concepts is still missing. Current therapy concepts are based on the two pillars consisting of surgical debridement with joint or bone reconstruction as well as prolonged antibiotic therapy. An improved understanding of both host and pathogen-related factors leading to treatment failure is essential in musculoskeletal infections. Therefore, this review aims to give an overview of pathogen-related pathophysiology in musculoskeletal infections. It describes defense strategies of pathogens such as (1) biofilm, its development, characteristics, and treatment options. In addition, (2) characteristics of small colony variants and (3) intracellular bacteria are highlighted. Lastly (4) an outlook for potential and promising future therapeutic strategies is provided.
Collapse
Affiliation(s)
- Josina Straub
- Department of Trauma Surgery, University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| | - Susanne Baertl
- Department of Trauma Surgery, University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| | - Marielle Verheul
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
| | - Nike Walter
- Department of Trauma Surgery, University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| | - Ronald Man Yeung Wong
- Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Volker Alt
- Department of Trauma Surgery, University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| | - Markus Rupp
- Department of Trauma Surgery, University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany.
| |
Collapse
|
18
|
Hashim NT, Babiker R, Rahman MM, Chaitanya NCSK, Mohammed R, Dasnadi SP, Gismalla BG. Gum Arabic as a potential candidate in quorum quenching and treatment of periodontal diseases. FRONTIERS IN ORAL HEALTH 2024; 5:1459254. [PMID: 39439926 PMCID: PMC11493777 DOI: 10.3389/froh.2024.1459254] [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: 07/03/2024] [Accepted: 09/17/2024] [Indexed: 10/25/2024] Open
Abstract
Periodontal diseases are chronic inflammatory conditions influenced by bacterial biofilm formation and host immune responses, affecting millions worldwide. Traditional treatments like mechanical debridement and systemic antibiotics often face limitations, including biofilm resilience and antibiotic resistance. Gum Arabic (GA), a natural exudate from Acacia trees, presents a promising alternative with its anti-biofilm and anti-inflammatory properties. This review highlights the role of GA in periodontal therapy, particularly its ability to interfere with quorum sensing (QS) pathways, specifically the AI-2 signaling system used by key periodontal pathogens such as Porphyromonas gingivalis, Aggregatibacter actinomycetemcomitans, and Fusobacterium nucleatum. By disrupting QS, GA inhibits biofilm formation, reduces bacterial virulence, and promotes a balanced oral microbiome. GA's prebiotic properties also encourage the growth of beneficial bacteria, enhancing the host's immune response while preserving the systemic microbiome. Clinical studies demonstrate GA's effectiveness as an adjunct in periodontal therapy, with significant reductions in plaque accumulation, gingival inflammation, and bleeding. This highlights GA's potential as a natural therapeutic agent, offering an effective, antibiotic-sparing option in managing periodontal disease. However, further research is warranted to fully establish GA's role in comprehensive periodontal care and its long-term benefits.
Collapse
Affiliation(s)
- Nada Tawfig Hashim
- Department of Periodontics, RAK College of Dental Sciences, RAK Medical & Health Sciences University, Ras al-Khaimah, United Arab Emirates
| | - Rasha Babiker
- Department of Physiology, RAK College of Medical Sciences, RAK Medical & Health Sciences University, Ras-al-Khaimah, United Arab Emirates
| | - Mohammed Mustahsen Rahman
- Department of Periodontics, RAK College of Dental Sciences, RAK Medical & Health Sciences University, Ras al-Khaimah, United Arab Emirates
| | - Nallan C. S. K. Chaitanya
- Department of Oral Radiology, RAK College of Dental Sciences, RAK Medical & Health Sciences University, Ras al-Khaimah, United Arab Emirates
| | - Riham Mohammed
- Department of Oral Surgery, RAK College of Dental Sciences, RAK Medical & Health Sciences University, Ras al-Khaimah, United Arab Emirates
| | - Shahistha Parveen Dasnadi
- Department of Orthodontics, RAK College of Dental Sciences, RAK Medical & Health Sciences University, Ras al-Khaimah, United Arab Emirates
| | - Bakri Gobara Gismalla
- Department of Oral Rehabilitation, Faculty of Dentistry, University of Khartoum, Khartoum, Sudan
| |
Collapse
|
19
|
Rogers ME, de Pablos LM, Sunter JD. Gels and cells: the Leishmania biofilm as a space and place for parasite transmission. Trends Parasitol 2024; 40:876-885. [PMID: 39218719 DOI: 10.1016/j.pt.2024.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 09/04/2024]
Abstract
Leishmania make an abundant glycoprotein and proteophosphoglycan-rich gel, called the promastigote secretory gel, in the anterior midgut of their sand fly vector. This gel is a multi-faceted virulence factor which promotes the survival and transmission of the parasites between hosts. Here, we present the case that Leishmania parasites embedded in the promastigote secretory gel should be redefined as a biofilm as it shares striking similarities in biogenesis, form, and function with biofilms of other unicellular organisms. We believe that this reinterpretation will stimulate new hypotheses and avenues of research to improve our understanding of the developmental programme of Leishmania and the interaction these parasites and other kinetoplastids have with their insect hosts.
Collapse
Affiliation(s)
- Matthew E Rogers
- Department of Disease Control, London School of Hygiene and Tropical Medicine, London, UK
| | - Luis Miguel de Pablos
- Department of Parasitology, University of Granada, Granada, Spain; Institute of Biotechnology, University of Granada, Granada, Spain
| | | |
Collapse
|
20
|
Davignon G, Pietrosemoli N, Benaroudj N, Soupé-Gilbert ME, Cagliero J, Turc É, Picardeau M, Guentas L, Goarant C, Thibeaux R. Leptospira interrogans biofilm transcriptome highlights adaption to starvation and general stress while maintaining virulence. NPJ Biofilms Microbiomes 2024; 10:95. [PMID: 39349472 PMCID: PMC11442865 DOI: 10.1038/s41522-024-00570-0] [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: 05/03/2024] [Accepted: 09/15/2024] [Indexed: 10/02/2024] Open
Abstract
Life-threatening Leptospira interrogans navigate a dual existence: surviving in the environment and infecting mammalian hosts. Biofilm formation is presumably an important survival strategy to achieve this process. Understanding the relation between biofilm and virulence might improve our comprehension of leptospirosis epidemiology. Our study focused on elucidating Leptospira's adaptations and regulations involved in such complex microenvironments. To determine the transcriptional profile of Leptospira in biofilm, we compared the transcriptomes in late biofilms and in exponential planktonic cultures. While genes for motility, energy production, and metabolism were downregulated, those governing general stress response, defense against metal stress, and redox homeostasis showed a significant upsurge, hinting at a tailored defensive strategy against stress. Further, despite a reduced metabolic state, biofilm disruption swiftly restored metabolic activity. Crucially, bacteria in late biofilms or resulting from biofilm disruption retained virulence in an animal model. In summary, our study highlights Leptospira's adaptive equilibrium in biofilms: minimizing energy expenditure, potentially aiding in withstanding stresses while maintaining pathogenicity. These insights are important for explaining the survival strategies of Leptospira, revealing that a biofilm lifestyle may confer an advantage in maintaining virulence, an understanding essential for managing leptospirosis across both environmental and mammalian reservoirs.
Collapse
Affiliation(s)
- Grégoire Davignon
- Leptospirosis Research and Expertise Unit, Institut Pasteur de Nouvelle-Calédonie, Institut Pasteur International Network, Nouméa, New Caledonia
- Exact and Applied Sciences Institute (ISEA), University of New Caledonia, BP R4, 98851, Nouméa, New Caledonia
| | - Natalia Pietrosemoli
- Institut Pasteur, Université Paris Cité, Bioinformatics and Biostatistics Hub, F-75015, Paris, France
| | - Nadia Benaroudj
- Biology of Spirochetes, Institut Pasteur, Université Paris Cité, CNRS UMR 6047, F-75015, Paris, France
| | - Marie-Estelle Soupé-Gilbert
- Leptospirosis Research and Expertise Unit, Institut Pasteur de Nouvelle-Calédonie, Institut Pasteur International Network, Nouméa, New Caledonia
| | - Julie Cagliero
- Leptospirosis Research and Expertise Unit, Institut Pasteur de Nouvelle-Calédonie, Institut Pasteur International Network, Nouméa, New Caledonia
| | - Élodie Turc
- Institut Pasteur, Université Paris Cité, Plate-forme Technologique Biomics, F-75015, Paris, France
| | - Mathieu Picardeau
- Biology of Spirochetes, Institut Pasteur, Université Paris Cité, CNRS UMR 6047, F-75015, Paris, France
| | - Linda Guentas
- Exact and Applied Sciences Institute (ISEA), University of New Caledonia, BP R4, 98851, Nouméa, New Caledonia
| | - Cyrille Goarant
- Pacific Community SPC - Public Health Division - B.P. D5, Nouméa, New Caledonia
| | - Roman Thibeaux
- Leptospirosis Research and Expertise Unit, Institut Pasteur de Nouvelle-Calédonie, Institut Pasteur International Network, Nouméa, New Caledonia.
| |
Collapse
|
21
|
Kačániová M, Garzoli S, Ben Hsouna A, Bianchi A, Kluz MI, Elizondo-Luevano JH, Ban Z, Ben Saad R, Mnif W, Haščík P. The Potential of Thymus serpyllum Essential Oil as an Antibacterial Agent against Pseudomonas aeruginosa in the Preservation of Sous Vide Red Deer Meat. Foods 2024; 13:3107. [PMID: 39410141 PMCID: PMC11476099 DOI: 10.3390/foods13193107] [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: 08/29/2024] [Revised: 09/20/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
Foodborne infections caused by microbes are a serious health risk. Regarding this, customer preferences for "ready-to-eat" or minimally processed (MP) deer meat are one of the main risk factors. Given the health dangers associated with food, essential oil (EO) is a practical substitute used to decrease pathogenic germs and extend the shelf-life of MP meals. Nonetheless, further data regarding EO use in MP meals are required. In order to evaluate new, safer alternatives to chemicals for disease control and food preservation, this research was carried out in the following areas to assess the antibacterial and antibiofilm characteristics of Thymus serpyllum (TSEO) essential oil, which is extracted from dried flowering stalks. Furthermore, this study applied an essential oil of wild thyme and inoculated the sous vide deer meat with Pseudomonas aeruginosa for seven days at 4 °C in an effort to prolong its shelf-life. Against P. aeruginosa, the essential oil exhibited potent antibacterial action. The findings of the minimal biofilm inhibition concentration (MBIC) crystal violet test demonstrated the substantial antibiofilm activity of the TSEO. The TSEO modified the protein profiles of bacteria on glass and plastic surfaces, according to data from MALDI-TOF MS analysis. Moreover, it was discovered that P. aeruginosa was positively affected by the antibacterial properties of TSEO. The anti-Pseudomonas activity of the TSEO was marginally higher in vacuum-packed sous vide red deer meat samples than in control samples. The most frequently isolated species from sous vide deer meat, if we do not consider the applied bacteria Pseudomonas aeruginosa, were P. fragi, P. lundensis, and P. taetrolens. These results highlight the antibacterial and antibiofilm qualities of TSEO, demonstrating its potential for food preservation and extending the shelf-life of deer meat.
Collapse
Affiliation(s)
- Miroslava Kačániová
- Institute of Horticulture, Faculty of Horticulture and Landscape Engineering, Slovak University of Agriculture, Trieda Andreja Hlinku 2, 949 76 Nitra, Slovakia
- School of Medical & Health Sciences, University of Economics and Human Sciences in Warsaw, Okopowa 59, 01043 Warszawa, Poland;
| | - Stefania Garzoli
- Department of Chemistry and Technologies of Drug, Sapienza University, P. le Aldo Moro, 5, 00185 Rome, Italy;
| | - Anis Ben Hsouna
- Laboratory of Biotechnology and Plant Improvement, Centre of Biotechnology of Sfax, B.P “1177”, Sfax 3018, Tunisia; (A.B.H.); (R.B.S.)
- Department of Environmental Sciences and Nutrition, Higher Institute of Applied Sciences and Technology of Mahdia, University of Monastir, Monastir 5000, Tunisia
| | - Alessandro Bianchi
- Department of Agriculture, Food and Environment, University of Pisa, Via del Borghetto 80, 56124 Pisa, Italy;
| | - Maciej Ireneusz Kluz
- School of Medical & Health Sciences, University of Economics and Human Sciences in Warsaw, Okopowa 59, 01043 Warszawa, Poland;
| | - Joel Horacio Elizondo-Luevano
- Faculty of Agronomy, Universidad Autónoma de Nuevo León, Av. Francisco Villa S/N, Col. Ex Hacienda el Canadá, General Escobedo 66050, Nuevo León, Mexico;
| | - Zhaojun Ban
- Zhejiang Provincial Key Laboratory of Chemical and Biological Processing Technology of Farm Products, Zhejiang Provincial Collaborative Innovation Center of Agricultural Biological Resources Biochemical Manufacturing, School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou 310023, China;
| | - Rania Ben Saad
- Laboratory of Biotechnology and Plant Improvement, Centre of Biotechnology of Sfax, B.P “1177”, Sfax 3018, Tunisia; (A.B.H.); (R.B.S.)
| | - Wissem Mnif
- Department of Chemistry, College of Sciences of Bisha, University of Bisha, P.O. Box 199, Bisha 61922, Saudi Arabia;
| | - Peter Haščík
- Institute of Food Technology, Faculty of Biotechnology and Food Sciences, Slovak University of Agriculture, Trieda Andreja Hlinku 2, 949 76 Nitra, Slovakia;
| |
Collapse
|
22
|
Cui Y, Wang D, Zhang L, Qu X. Research progress on the regulatory mechanism of biofilm formation in probiotic lactic acid bacteria. Crit Rev Food Sci Nutr 2024:1-15. [PMID: 39244761 DOI: 10.1080/10408398.2024.2400593] [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: 09/10/2024]
Abstract
Probiotic lactic acid bacteria (LAB) must undergo three key stages of testing, including food processing, storage, and gastrointestinal tract environment, their beneficial effects could exert. The biofilm formation of probiotic LAB is helpful for improving their stress resistances, survival rates, and colonization abilities under adverse environmental conditions, laying an important foundation for their probiotic effects. In this review, the formation process, the composition and function of basic components of probiotic LAB biofilm have been summarized. This review focuses on the regulatory mechanism of probiotic LAB biofilm formation. In addition, the characteristics and related mechanisms of probiotics in biofilm state have been analyzed to guide the application of probiotic LAB biofilms in the field of health and food. The biofilm formation of LAB is an extremely complex process involving multiple regulatory factors. Besides quorum sensing (QS), other regulatory factors are not yet fully understood. The probiotic LAB in biofilm state exhibit superior survival rate, adhesion performance, and immunomodulation ability, attribute to various metabolic processes, including stress response, exopolysaccharide (EPS) metabolism, amino acid and protein metabolisms, etc. The understanding about regulatory mechanism of biofilm formation of different probiotic species and strains will accelerate the development and application of probiotics products.
Collapse
Affiliation(s)
- Yanhua Cui
- Department of Food Nutrition and Health, School of Medicine and Health, Harbin Institute of Technology, Harbin, China
| | - Dongqi Wang
- Department of Food Nutrition and Health, School of Medicine and Health, Harbin Institute of Technology, Harbin, China
| | - Lanwei Zhang
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Xiaojun Qu
- Institute of Microbiology, Heilongjiang Academy of Sciences, Harbin, China
| |
Collapse
|
23
|
Jotta VFM, García GJY, Fonseca PLC, de Mello Ferreira A, Azevedo V, Brenig B, Góes-Neto A, Badotti F. Taxonomic and functional characterization of biofilms from a photovoltaic panel reveals high genetic and metabolic complexity of the communities. J Appl Microbiol 2024; 135:lxae231. [PMID: 39257028 DOI: 10.1093/jambio/lxae231] [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: 07/05/2024] [Revised: 08/19/2024] [Accepted: 09/08/2024] [Indexed: 09/12/2024]
Abstract
AIMS Biofilms are complex microbial cell aggregates that attach to different surfaces in nature, industrial environments, or hospital settings. In photovoltaic panels (PVs), biofilms are related to significant energy conversion losses. In this study, our aim was to characterize the communities of microorganisms and the genes involved in biofilm formation. METHODS AND RESULTS In this study, biofilm samples collected from a PV system installed in southeastern Brazil were analyzed through shotgun metagenomics, and the microbial communities and genes involved in biofilm formation were investigated. A total of 2030 different genera were identified in the samples, many of which were classified as extremophiles or producers of exopolysaccharides. Bacteria prevailed in the samples (89%), mainly the genera Mucilaginibacter, Microbacterium, Pedobacter, Massilia, and Hymenobacter. The functional annotation revealed >12 000 genes related to biofilm formation and stress response. Genes involved in the iron transport and synthesis of c-di-GMP and c-AMP second messengers were abundant in the samples. The pathways related to these components play a crucial role in biofilm formation and could be promising targets for preventing biofilm formation in the PV. In addition, Raman spectroscopy analysis indicated the presence of hematite, goethite, and ferrite, consistent with the mineralogical composition of the regional soil and metal-resistant bacteria. CONCLUSIONS Taken together, our findings reveal that PV biofilms are a promising source of microorganisms of industrial interest and genes of central importance in regulating biofilm formation and persistence.
Collapse
Affiliation(s)
- Viviane Faria Morais Jotta
- Programa de Pós-Graduação em Tecnologia de Produtos e Processos, Centro Federal de Educação Tecnológica de Minas Gerais (CEFET-MG), 30510-000, Belo Horizonte, MG, Brazil
| | - Glen Jasper Yupanqui García
- Institute of Biological Sciences, Universidade Federal de Minas Gerais (UFMG), 31270-901, Belo Horizonte, MG, Brazil
| | - Paula Luize Camargos Fonseca
- Institute of Biological Sciences, Universidade Federal de Minas Gerais (UFMG), 31270-901, Belo Horizonte, MG, Brazil
| | - Angela de Mello Ferreira
- Department of Chemistry, Centro Federal de Educação Tecnológica de Minas Gerais (CEFET-MG), 30421-169, Belo Horizonte, MG, Brazil
| | - Vasco Azevedo
- Department of Genetics, Evolution and Ecology, Institute of Biological Science, Universidade Federal de Minas Gerais (UFMG), 31270-901, Belo Horizonte, MG, Brazil
| | - Bertram Brenig
- Georg-August-University Goettingen, Institute of Veterinary Medicine, Burckhardtweg 2, 37077 Göttingen, Germany
| | - Aristóteles Góes-Neto
- Institute of Biological Sciences, Universidade Federal de Minas Gerais (UFMG), 31270-901, Belo Horizonte, MG, Brazil
| | - Fernanda Badotti
- Programa de Pós-Graduação em Tecnologia de Produtos e Processos, Centro Federal de Educação Tecnológica de Minas Gerais (CEFET-MG), 30510-000, Belo Horizonte, MG, Brazil
- Department of Chemistry, Centro Federal de Educação Tecnológica de Minas Gerais (CEFET-MG), 30421-169, Belo Horizonte, MG, Brazil
| |
Collapse
|
24
|
Čuk N, Simončič B, Fink R, Tomšič B. Bacterial Adhesion to Natural and Synthetic Fibre-Forming Polymers: Influence of Material Properties. Polymers (Basel) 2024; 16:2409. [PMID: 39274042 PMCID: PMC11397841 DOI: 10.3390/polym16172409] [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: 08/01/2024] [Revised: 08/19/2024] [Accepted: 08/22/2024] [Indexed: 09/16/2024] Open
Abstract
Polymer-based textiles have a major impact on human well-being, as they provide the desired functional protection and aesthetic comfort when worn. However, natural and synthetic polymer-based textiles can also pose serious health risks, as they are surfaces that allow the adhesion of various bacteria, including pathogenic bacteria. To minimise these problems, antibacterial chemical treatments are generally applicable in the case of polymer-based textiles. However, to avoid the use of potentially toxic chemicals, sustainable approaches require the customised design of non-adhesive polymer-based textiles, considering their chemical, physicochemical, constructional, and textural properties. Before designing, several articles are required to gain sufficient knowledge of the described object. Despite the urgent need to combat bacteria (on polymer-based textiles), which pose a serious global health risk, only a few review articles have been published that address bacterial adhesion in the context of superhydrophobic and antibacterial textile materials, while only one review article holistically addresses the textile factors and their influence on this phenomenon. The aim of this review article is to expand the insufficient knowledge about bacterial adhesion to polymer-based textiles on the basis of theoretical findings and real examples through a high degree of structuring, simplification, holistic consideration, and visualization. Therefore, this review provides an insight into the mechanisms involved in bacterial adhesion and a comprehensive overview of the influence of different textile factors, such as chemical composition, hydrophilicity/hydrophobicity, surface charge, surface free energy, roughness, and porosity, on bacterial adhesion. To emphasise the importance of the synergistic effect of the combined textile factors, examples of the influence of hydrophilicity/hydrophobicity in combination with surface charge, surface roughness, and porosity are discussed. From the review, it can be concluded that the combination of hydrophilicity/hydrophobicity and the surface charge of textile fibres and bacteria is crucial for bacterial adhesion, with roughness and porosity being the most important factors among the constructive and textural properties of polymer-based textiles.
Collapse
Affiliation(s)
- Nina Čuk
- Faculty of Natural Sciences and Engineering, University of Ljubljana, Aškerčeva cesta 12, 1000 Ljubljana, Slovenia
| | - Barbara Simončič
- Faculty of Natural Sciences and Engineering, University of Ljubljana, Aškerčeva cesta 12, 1000 Ljubljana, Slovenia
| | - Rok Fink
- Faculty of Health Sciences, University of Ljubljana, Zdravstvena pot 5, 1000 Ljubljana, Slovenia
| | - Brigita Tomšič
- Faculty of Natural Sciences and Engineering, University of Ljubljana, Aškerčeva cesta 12, 1000 Ljubljana, Slovenia
| |
Collapse
|
25
|
Ronish LA, Biswas B, Bauer RM, Jacob ME, Piepenbrink KH. The role of extracellular structures in Clostridioides difficile biofilm formation. Anaerobe 2024; 88:102873. [PMID: 38844261 DOI: 10.1016/j.anaerobe.2024.102873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 04/27/2024] [Accepted: 06/03/2024] [Indexed: 07/08/2024]
Abstract
C. difficile infection (CDI) is a costly and increasing burden on the healthcare systems of many developed countries due to the high rates of nosocomial infections. Despite the availability of several antibiotics with high response rates, effective treatment is hampered by recurrent infections. One potential mechanism for recurrence is the existence of C. difficile biofilms in the gut which persist through the course of antibiotics. In this review, we describe current developments in understanding the molecular mechanisms by which C. difficile biofilms form and are stabilized through extracellular biomolecular interactions.
Collapse
Affiliation(s)
- Leslie A Ronish
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Baishakhi Biswas
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Robert M Bauer
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Mallory E Jacob
- Biochemistry Department, University of Geneva, Geneva, Switzerland
| | - Kurt H Piepenbrink
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA; Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA; Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA; Nebraska Food for Health Center, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA; Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA.
| |
Collapse
|
26
|
Sedighi O, Bednarke B, Sherriff H, Doiron AL. Nanoparticle-Based Strategies for Managing Biofilm Infections in Wounds: A Comprehensive Review. ACS OMEGA 2024; 9:27853-27871. [PMID: 38973924 PMCID: PMC11223148 DOI: 10.1021/acsomega.4c02343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 05/28/2024] [Accepted: 06/04/2024] [Indexed: 07/09/2024]
Abstract
Chronic wounds containing opportunistic bacterial pathogens are a growing problem, as they are the primary cause of morbidity and mortality in developing and developed nations. Bacteria can adhere to almost every surface, forming architecturally complex communities called biofilms that are tolerant to an individual's immune response and traditional treatments. Wound dressings are a primary source and potential treatment avenue for biofilm infections, and research has recently focused on using nanoparticles with antimicrobial activity for infection control. This Review categorizes nanoparticle-based approaches into four main types, each leveraging unique mechanisms against biofilms. Metallic nanoparticles, such as silver and copper, show promising data due to their ability to disrupt bacterial cell membranes and induce oxidative stress, although their effectiveness can vary based on particle size and composition. Phototherapy-based nanoparticles, utilizing either photodynamic or photothermal therapy, offer targeted microbial destruction by generating reactive oxygen species or localized heat, respectively. However, their efficacy depends on the presence of light and oxygen, potentially limiting their use in deeper or more shielded biofilms. Nanoparticles designed to disrupt extracellular polymeric substances directly target the biofilm structure, enhancing the penetration and efficacy of antimicrobial agents. Lastly, nanoparticles that induce biofilm dispersion represent a novel strategy, aiming to weaken the biofilm's defense and restore susceptibility to antimicrobials. While each method has its advantages, the selection of an appropriate nanoparticle-based treatment depends on the specific requirements of the wound environment and the type of biofilm involved. The integration of these nanoparticles into wound dressings not only promises enhanced treatment outcomes but also offers a reduction in the overall use of antibiotics, aligning with the urgent need for innovative solutions in the fight against antibiotic-tolerant infections. The overarching objective of employing these diverse nanoparticle strategies is to replace antibiotics or substantially reduce their required dosages, providing promising avenues for biofilm infection management.
Collapse
Affiliation(s)
- Omid Sedighi
- Department
of Electrical and Biomedical Engineering, University of Vermont, Burlington, Vermont 05405, United States
| | - Brooke Bednarke
- Department
of Electrical and Biomedical Engineering, University of Vermont, Burlington, Vermont 05405, United States
| | - Hannah Sherriff
- Department
of Electrical and Biomedical Engineering, University of Vermont, Burlington, Vermont 05405, United States
| | - Amber L. Doiron
- Department
of Electrical and Biomedical Engineering, University of Vermont, Burlington, Vermont 05405, United States
| |
Collapse
|
27
|
Las Heras K, Garcia-Orue I, Rancan F, Igartua M, Santos-Vizcaino E, Hernandez RM. Modulating the immune system towards a functional chronic wound healing: A biomaterials and Nanomedicine perspective. Adv Drug Deliv Rev 2024; 210:115342. [PMID: 38797316 DOI: 10.1016/j.addr.2024.115342] [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: 01/26/2024] [Revised: 05/16/2024] [Accepted: 05/18/2024] [Indexed: 05/29/2024]
Abstract
Chronic non-healing wounds persist as a substantial burden for healthcare systems, influenced by factors such as aging, diabetes, and obesity. In contrast to the traditionally pro-regenerative emphasis of therapies, the recognition of the immune system integral role in wound healing has significantly grown, instigating an approach shift towards immunological processes. Thus, this review explores the wound healing process, highlighting the engagement of the immune system, and delving into the behaviors of innate and adaptive immune cells in chronic wound scenarios. Moreover, the article investigates biomaterial-based strategies for the modulation of the immune system, elucidating how the adjustment of their physicochemical properties or their synergistic combination with other agents such as drugs, proteins or mesenchymal stromal cells can effectively modulate the behaviors of different immune cells. Finally this review explores various strategies based on synthetic and biological nanostructures, including extracellular vesicles, to finely tune the immune system as natural immunomodulators or therapeutic nanocarriers with promising biophysical properties.
Collapse
Affiliation(s)
- Kevin Las Heras
- NanoBioCel Research Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV-EHU), Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain
| | - Itxaso Garcia-Orue
- NanoBioCel Research Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV-EHU), Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN). Institute of Health Carlos III, Madrid, Spain
| | - Fiorenza Rancan
- Department of Dermatology, Venereology und Allergology,Clinical Research Center for Hair and Skin Science, Charité - Universitätsmedizin Berlin
| | - Manoli Igartua
- NanoBioCel Research Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV-EHU), Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN). Institute of Health Carlos III, Madrid, Spain
| | - Edorta Santos-Vizcaino
- NanoBioCel Research Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV-EHU), Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN). Institute of Health Carlos III, Madrid, Spain.
| | - Rosa Maria Hernandez
- NanoBioCel Research Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV-EHU), Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN). Institute of Health Carlos III, Madrid, Spain.
| |
Collapse
|
28
|
Alipour-Khezri E, Skurnik M, Zarrini G. Pseudomonas aeruginosa Bacteriophages and Their Clinical Applications. Viruses 2024; 16:1051. [PMID: 39066214 PMCID: PMC11281547 DOI: 10.3390/v16071051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024] Open
Abstract
Antimicrobial resistance poses a serious risk to contemporary healthcare since it reduces the number of bacterial illnesses that may be treated with antibiotics, particularly for patients with long-term conditions like cystic fibrosis (CF). People with a genetic predisposition to CF often have recurrent bacterial infections in their lungs due to a buildup of sticky mucus, necessitating long-term antibiotic treatment. Pseudomonas aeruginosa infections are a major cause of CF lung illness, and P. aeruginosa airway isolates are frequently resistant to many antibiotics. Bacteriophages (also known as phages), viruses that infect bacteria, are a viable substitute for antimicrobials to treat P. aeruginosa infections in individuals with CF. Here, we reviewed the utilization of P. aeruginosa bacteriophages both in vivo and in vitro, as well as in the treatment of illnesses and diseases, and the outcomes of the latter.
Collapse
Affiliation(s)
- Elaheh Alipour-Khezri
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz 51368, Iran;
| | - Mikael Skurnik
- Human Microbiome Research Program, and Department of Bacteriology and Immunology, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland
| | - Gholamreza Zarrini
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz 51368, Iran;
- Microbial Biotechnology Research Group, University of Tabriz, Tabriz 51368, Iran
| |
Collapse
|
29
|
Wang WM, Lu TH, Chen CY, Liao CM. Assessing microplastics-antibiotics coexistence induced ciprofloxacin-resistant Pseudomonas aeruginosa at a water region scale. WATER RESEARCH 2024; 257:121721. [PMID: 38728782 DOI: 10.1016/j.watres.2024.121721] [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: 01/25/2024] [Revised: 04/30/2024] [Accepted: 05/03/2024] [Indexed: 05/12/2024]
Abstract
Microplastics (MPs) waste is widespread globally in water systems. The opportunistic human pathogen Pseudomonas aeruginosa can cause serious acute and chronic infections that are notoriously difficult to treat. Ciprofloxacin (CIP) is broadly applied as an anti-P. aeruginosa drug. A growing evidence reveals that antibiotic-resistance genes-carrying Pseudomonas aeruginosa were detected on MPs forming plastisphere due to their adsorbability along with high occurrence of CIP in water environments. The MPs-niched CIP-resistant P. aeruginosa has been likely to emerge as an unignorable public health issue. Here, we offered a novel approach to assess the development of CIP-resistant P. aeruginosa under MPs-antibiotic coexistence at a water region scale. By combing the adsorption isotherm models used to estimate CIP condensation around MPs and a pharmacokinetic/pharmacodynamic-based microbial population dynamic model, we predicted the P. aeruginosa development on CIP-adsorbed MPs in waters. Our assessment revealed a high antibiotic resistance in the P. aeruginosa populations (∼50 %) with a wider range of waterborne total cell counts (∼10-2-104 cfu mL-1) among water regions in that the resistance proportion was primarily determined by CIP pollution level and relative abundance of various polymer type of MPs. We implicate that water region-specific MPs were highly likely to provide media for P. aeruginosa propagation. Our results highlight the importance of antibiotic-resistant pathogen colonization-emerging environmental medium interactions when addressing global threat from MPs pollution, in the context of MPs-antibiotics co-contamination assessment and for the continued provision of water system management.
Collapse
Affiliation(s)
- Wei-Min Wang
- Department of Bioenvironmental Systems Engineering, National Taiwan University, Taipei, Taiwan 106319, China
| | - Tien-Hsuan Lu
- Department of Science Education and Application, National Taichung University of Education, Taichung 403514, China
| | - Chi-Yun Chen
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL 32610, United States; Center for Environmental and Human Toxicology, University of Florida, FL 32608, United States
| | - Chung-Min Liao
- Department of Bioenvironmental Systems Engineering, National Taiwan University, Taipei, Taiwan 106319, China.
| |
Collapse
|
30
|
Azad MA, Patel R. Practical Guidance for Clinical Microbiology Laboratories: Microbiologic diagnosis of implant-associated infections. Clin Microbiol Rev 2024; 37:e0010423. [PMID: 38506553 PMCID: PMC11237642 DOI: 10.1128/cmr.00104-23] [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] [Indexed: 03/21/2024] Open
Abstract
SUMMARYImplant-associated infections (IAIs) pose serious threats to patients and can be associated with significant morbidity and mortality. These infections may be difficult to diagnose due, in part, to biofilm formation on device surfaces, and because even when microbes are found, their clinical significance may be unclear. Despite recent advances in laboratory testing, IAIs remain a diagnostic challenge. From a therapeutic standpoint, many IAIs currently require device removal and prolonged courses of antimicrobial therapy to effect a cure. Therefore, making an accurate diagnosis, defining both the presence of infection and the involved microorganisms, is paramount. The sensitivity of standard microbial culture for IAI diagnosis varies depending on the type of IAI, the specimen analyzed, and the culture technique(s) used. Although IAI-specific culture-based diagnostics have been described, the challenge of culture-negative IAIs remains. Given this, molecular assays, including both nucleic acid amplification tests and next-generation sequencing-based assays, have been used. In this review, an overview of these challenging infections is presented, as well as an approach to their diagnosis from a microbiologic perspective.
Collapse
Affiliation(s)
- Marisa Ann Azad
- Division of Infectious Diseases, Department of Medicine, The Ottawa Hospital, Ottawa, Canada
- Ottawa Hospital Research Institute, Ottawa, Canada
| | - Robin Patel
- Division of Public Health, Infectious Diseases, and Occupational Medicine, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
31
|
Ashford JR. Impaired oral health: a required companion of bacterial aspiration pneumonia. FRONTIERS IN REHABILITATION SCIENCES 2024; 5:1337920. [PMID: 38894716 PMCID: PMC11183832 DOI: 10.3389/fresc.2024.1337920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 05/20/2024] [Indexed: 06/21/2024]
Abstract
Laryngotracheal aspiration has a widely-held reputation as a primary cause of lower respiratory infections, such as pneumonia, and is a major concern of care providers of the seriously ill orelderly frail patient. Laryngeal mechanical inefficiency resulting in aspiration into the lower respiratory tract, by itself, is not the cause of pneumonia. It is but one of several factors that must be present simultaneously for pneumonia to develop. Aspiration of oral and gastric contentsoccurs often in healthy people of all ages and without significant pulmonary consequences. Inthe seriously ill or elderly frail patient, higher concentrations of pathogens in the contents of theaspirate are the primary catalyst for pulmonary infection development if in an immunocompromised lower respiratory system. The oral cavity is a complex and ever changing eco-environment striving to maintain homogeneity among the numerous microbial communities inhabiting its surfaces. Poor maintenance of these surfaces to prevent infection can result inpathogenic changes to these microbial communities and, with subsequent proliferation, can altermicrobial communities in the tracheal and bronchial passages. Higher bacterial pathogen concentrations mixing with oral secretions, or with foods, when aspirated into an immunecompromised lower respiratory complex, may result in bacterial aspiration pneumonia development, or other respiratory or systemic diseases. A large volume of clinical evidence makes it clear that oral cleaning regimens, when used in caring for ill or frail patients in hospitals and long-term care facilities, drastically reduce the incidence of respiratory infection and death. The purpose of this narrative review is to examine oral health as a required causative companionin bacterial aspiration pneumonia development, and the effectiveness of oral infection control inthe prevention of this disease.
Collapse
|
32
|
Quispe Haro JJ, Chen F, Los R, Shi S, Sun W, Chen Y, Idema T, Wegner SV. Optogenetic Control of Bacterial Cell-Cell Adhesion Dynamics: Unraveling the Influence on Biofilm Architecture and Functionality. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2310079. [PMID: 38613837 PMCID: PMC11187914 DOI: 10.1002/advs.202310079] [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: 12/21/2023] [Revised: 03/22/2024] [Indexed: 04/15/2024]
Abstract
The transition of bacteria from an individualistic to a biofilm lifestyle profoundly alters their biology. During biofilm development, the bacterial cell-cell adhesions are a major determinant of initial microcolonies, which serve as kernels for the subsequent microscopic and mesoscopic structure of the biofilm, and determine the resulting functionality. In this study, the significance of bacterial cell-cell adhesion dynamics on bacterial aggregation and biofilm maturation is elucidated. Using photoswitchable adhesins between bacteria, modifying the dynamics of bacterial cell-cell adhesions with periodic dark-light cycles is systematic. Dynamic cell-cell adhesions with liquid-like behavior improve bacterial aggregation and produce more compact microcolonies than static adhesions with solid-like behavior in both experiments and individual-based simulations. Consequently, dynamic cell-cell adhesions give rise to earlier quorum sensing activation, better intermixing of different bacterial populations, improved biofilm maturation, changes in the growth of cocultures, and higher yields in fermentation. The here presented approach of tuning bacterial cell-cell adhesion dynamics opens the door for regulating the structure and function of biofilms and cocultures with potential biotechnological applications.
Collapse
Affiliation(s)
- Juan José Quispe Haro
- Institute of Physiological Chemistry and PathobiochemistryUniversity of MünsterMünsterGermany
| | - Fei Chen
- Institute of Physiological Chemistry and PathobiochemistryUniversity of MünsterMünsterGermany
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaChina
| | - Rachel Los
- Department of BionanoscienceKavli Institute of NanoscienceDelft University of TechnologyDelftThe Netherlands
| | - Shuqi Shi
- National Engineering Research Center for BiotechnologyCollege of Biotechnology and Pharmaceutical EngineeringNanjing Tech UniversityNanjingChina
- State Key Laboratory of Materials‐Oriented Chemical EngineeringCollege of Biotechnology and Pharmaceutical EngineeringNanjing Tech UniversityNanjingChina
| | - Wenjun Sun
- National Engineering Research Center for BiotechnologyCollege of Biotechnology and Pharmaceutical EngineeringNanjing Tech UniversityNanjingChina
- State Key Laboratory of Materials‐Oriented Chemical EngineeringCollege of Biotechnology and Pharmaceutical EngineeringNanjing Tech UniversityNanjingChina
| | - Yong Chen
- National Engineering Research Center for BiotechnologyCollege of Biotechnology and Pharmaceutical EngineeringNanjing Tech UniversityNanjingChina
- State Key Laboratory of Materials‐Oriented Chemical EngineeringCollege of Biotechnology and Pharmaceutical EngineeringNanjing Tech UniversityNanjingChina
| | - Timon Idema
- Department of BionanoscienceKavli Institute of NanoscienceDelft University of TechnologyDelftThe Netherlands
| | - Seraphine V. Wegner
- Institute of Physiological Chemistry and PathobiochemistryUniversity of MünsterMünsterGermany
| |
Collapse
|
33
|
Hyun JE, Hwang CY. Antimicrobial Peptide Reduces Cytotoxicity and Inflammation in Canine Epidermal Keratinocyte Progenitor Cells Induced by Pseudomonas aeruginosa Infection. Vet Sci 2024; 11:235. [PMID: 38921982 PMCID: PMC11209461 DOI: 10.3390/vetsci11060235] [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: 04/16/2024] [Revised: 05/20/2024] [Accepted: 05/22/2024] [Indexed: 06/27/2024] Open
Abstract
The direct effects and antimicrobial activity of synthetic antimicrobial peptides (AMPs) obtained from dogs, including cBD, cBD103, and cCath, against P. aeruginosa wild-type strain PAO1 and canine keratinocytes were analyzed. Antibacterial effects on planktonic bacteria were assessed by determining the minimum bactericidal concentrations (MBCs) of AMPs and by a time-kill assay. Antibiofilm effects were assessed using the microtiter plate assay. We also evaluated the effects of AMPs on cell cytotoxicity and host immune response induced by stimulating canine epidermal keratinocyte progenitor (CPEK) cells with PAO1 and its LPS. cBD, cBD103, and cCath all exhibited dose-dependent antimicrobial and antibiofilm effects. In particular, 25 μg/mL cBD103 showed rapid bactericidal activity within 60 min and inhibited biofilm formation. In addition, pretreatment with cBD103 (25 µg/mL) and cCath (50 µg/mL) 1 h before stimulation significantly reduced the cytotoxicity of the CPEK cells by PAO1 and LPS-induced IL-6 and TNF-a expressions. cBD had little effect on the response to PAO1 and LPS in the cells. These results indicate the therapeutic potential of AMPs in P. aeruginosa skin infections. However, further studies on the mechanism of action of AMPs in keratinocytes and clinical trials are needed.
Collapse
Affiliation(s)
- Jae-Eun Hyun
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Cheol-Yong Hwang
- Laboratory of Veterinary Dermatology, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
34
|
Murphy C, Banasiewicz T, Duteille F, Ferrando PM, Jerez González JA, Koullias G, Long Z, Nasur R, Salazar Trujillo MA, Bassetto F, Dunk AM, Iafrati M, Jawień A, Matsumura H, O'Connor L, Sanchez V, Wu J. A proactive healing strategy for tackling biofilm-based surgical site complications: Wound Hygiene Surgical. J Wound Care 2024; 33:S1-S30. [PMID: 38787336 DOI: 10.12968/jowc.2024.33.sup5c.s1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Affiliation(s)
- Chris Murphy
- Vascular Nurse Specialist, Ottawa Hospital Limb Preservation Centre, Ottawa, Canada
| | - Tomasz Banasiewicz
- Head of Department of General Endocrine Surgery and Gastrointestinal Oncology, Poznań University of Medical Sciences, Poznań, Poland
| | | | - Pietro Maria Ferrando
- Consultant Plastic and Oncoplastic Surgeon, Plastic Surgery Department and Breast Unit, City of Health and Science, University Hospital of Turin, Italy
| | | | - George Koullias
- Associate Professor of Surgery, Division of Vascular & Endovascular Surgery, Stony Brook University Hospital & Stony Brook Southampton Hospital, USA
| | - Zhang Long
- Chief Surgeon, Associate Professor, Mentor of Master in Surgery, Executive Deputy Director of Wound Healing Center, Department of Interventional Radiology and Vascular Surgery, Peking University Third Hospital, Beijing, China
| | - Reem Nasur
- Consultant Obstetrician, Gynaecologist and Head of Women's Health, Blackpool Teaching Hospitals NHS Foundation Trust, UK
| | - Marco Antonio Salazar Trujillo
- Plastic and Reconstructive Surgeon, Consultant in Advanced Wound Management, Scientific Director of Plastic, Aesthetic and Laser Surgery, Renovarte, Colombia
| | - Franco Bassetto
- Full Professor of Plastic, Reconstructive and Aesthetic Surgery, Chief of the Clinic of Plastic and Reconstructive Surgery, Padova University Hospital, Padova, Italy
| | - Ann Marie Dunk
- RN MN(research) PhD(c) Ghent University, Belgium, Clinical Nurse Consultant, Tissue Viability Unit, Canberra Hospital, Australian Capital Territory, Australia
| | - Mark Iafrati
- Director of the Vanderbilt Wound Center and Professor of Vascular Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Arkadiusz Jawień
- Head of the Department of Vascular Surgery and Angiology, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Hajime Matsumura
- Professor, Chair of the Department of Plastic Surgery and Director of the General Informatics Division, Tokyo Medical University, Tokyo, Japan
| | - Louise O'Connor
- Independent Tissue Viability Nurse Consultant, Manchester, UK
| | - Violeta Sanchez
- Specialist Nurse in Complex Wounds and Pressure Ulcers, Son Llàtzer Hospital, Palma de Mallorca, Spain
| | - Jun Wu
- Professor, Director, Department of Burn and Plastic Surgery, First Affiliated Hospital, Shenzhen University, Shenzhen, China
| |
Collapse
|
35
|
La Corte SG, Stevens CA, Cárcamo-Oyarce G, Ribbeck K, Wingreen NS, Datta SS. Morphogenesis of bacterial colonies in polymeric environments. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.18.590088. [PMID: 38712130 PMCID: PMC11071276 DOI: 10.1101/2024.04.18.590088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Many bacteria live in polymeric fluids, such as mucus, environmental polysaccharides, and extracellular polymers in biofilms. However, lab studies typically focus on cells in polymer-free fluids. Here, we show that interactions with polymers shape a fundamental feature of bacterial life-how they proliferate in space in multicellular colonies. Using experiments, we find that when polymer is sufficiently concentrated, cells generically and reversibly form large serpentine "cables" as they proliferate. By combining experiments with biophysical theory and simulations, we demonstrate that this distinctive form of colony morphogenesis arises from an interplay between polymer-induced entropic attraction between neighboring cells and their hindered ability to diffusely separate from each other in a viscous polymer solution. Our work thus reveals a pivotal role of polymers in sculpting proliferating bacterial colonies, with implications for how they interact with hosts and with the natural environment, and uncovers quantitative principles governing colony morphogenesis in such complex environments.
Collapse
|
36
|
Seo S, Yang Q, Jeong S, Della Porta A, Kapoor H, Gibson DJ. A surfactant-based dressing can reduce the appearance of Pseudomonas aeruginosa pigments and uncover the dermal extracellular matrix in an ex vivo porcine skin wound model. Int Wound J 2024; 21:e14510. [PMID: 38148595 PMCID: PMC10958096 DOI: 10.1111/iwj.14510] [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: 10/18/2022] [Revised: 11/08/2023] [Accepted: 11/09/2023] [Indexed: 12/28/2023] Open
Abstract
From previous studies, we have shown that viable colony forming units of bacteria and bacterial biofilms are reduced after sequential treatment with a surfactant-based dressing. Here, we sought to test the impact on visible bacterial pigments and the ultrastructural impact following the sequential treatment of the same surfactant-based dressing. Mature Pseudomonas aeruginosa biofilms were grown on ex vivo porcine skin explants, and an imaging-based analysis was used to compare the skin with and without a concentrated surfactant. In explants naturally tinted by bacterial chromophores, wiping alone had no effect, while the use of a surfactant-based dressing reduced coloration. Similarly, daily wiping led to increased immunohistochemical staining for P. aeruginosa antigens, but not in the surfactant group. Confocal immunofluorescent imaging revealed limited bacterial penetration and coating of the dermis and loose pieces of sloughing material. Ultrastructural analysis confirmed that the biofilms were masking the extracellular matrix (ECM), but the surfactant could remove them, re-exposing the ECM. The masking of the ECM may provide another non-inflammatory explanation for delayed healing, as the ECM is no longer accessible for wound cell locomotion. The use of a poloxamer-based surfactant appears to be an effective way to remove bacterial chromophores and the biofilm coating the ECM fibres.
Collapse
Affiliation(s)
- Soojung Seo
- Department of OB/GYNInstitute for Wound Research, University of FloridaGainesvilleFloridaUSA
- Department of Biological SciencesUniversity of AlabamaTuscaloosaAlabamaUSA
| | - Qingping Yang
- Department of OB/GYNInstitute for Wound Research, University of FloridaGainesvilleFloridaUSA
| | - Sunyoung Jeong
- Department of OB/GYNInstitute for Wound Research, University of FloridaGainesvilleFloridaUSA
| | - Alessandra Della Porta
- Department of OB/GYNInstitute for Wound Research, University of FloridaGainesvilleFloridaUSA
| | - Harris Kapoor
- Department of OB/GYNInstitute for Wound Research, University of FloridaGainesvilleFloridaUSA
| | - Daniel J. Gibson
- Department of OB/GYNInstitute for Wound Research, University of FloridaGainesvilleFloridaUSA
- Capstone College of NursingUniversity of AlabamaTuscaloosaAlabamaUSA
| |
Collapse
|
37
|
MacConnell AE, Levack AE, Brown NM. Biofilm and How It Relates to Prosthetic Joint Infection. Orthop Clin North Am 2024; 55:161-169. [PMID: 38403363 DOI: 10.1016/j.ocl.2023.10.001] [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] [Indexed: 02/27/2024]
Abstract
Prosthetic joint infection following total joint arthroplasty is a devastating complication, resulting in increased morbidity and mortality for the patient. The formation of a biofilm on implanted hardware contributes to the difficulty in successful identification and eradication of the infection. Antibiotic therapy and surgical intervention are necessary for addressing this condition; we present a discussion on different treatment options, including those that are not yet routinely utilized in the clinical setting or are under investigation, to highlight the present and future of PJI management.
Collapse
Affiliation(s)
- Ashley E MacConnell
- Department of Orthopaedic Surgery and Rehabilitation, Loyola University Medical Center, 2160 South First Avenue, Suite 1700, Maywood, IL 60153, USA.
| | - Ashley E Levack
- Department of Orthopaedic Surgery and Rehabilitation, Loyola University Medical Center, 2160 South First Avenue, Suite 1700, Maywood, IL 60153, USA
| | - Nicholas M Brown
- Department of Orthopaedic Surgery and Rehabilitation, Loyola University Medical Center, 2160 South First Avenue, Suite 1700, Maywood, IL 60153, USA
| |
Collapse
|
38
|
Yang Y, Kong X, Niu B, Yang J, Chen Q. Differences in Biofilm Formation of Listeria monocytogenes and Their Effects on Virulence and Drug Resistance of Different Strains. Foods 2024; 13:1076. [PMID: 38611380 PMCID: PMC11011679 DOI: 10.3390/foods13071076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/19/2024] [Accepted: 03/21/2024] [Indexed: 04/14/2024] Open
Abstract
Listeria monocytogenes is recognized as one of the primary pathogens responsible for foodborne illnesses. The ability of L. monocytogenes to form biofilms notably increases its resistance to antibiotics such as ampicillin and tetracycline, making it exceedingly difficult to eradicate. Residual bacteria within the processing environment can contaminate food products, thereby posing a significant risk to public health. In this study, we used crystal violet staining to assess the biofilm-forming capacity of seven L. monocytogenes strains and identified ATCC 19112 as the strain with the most potent biofilm-forming. Subsequent fluorescence microscopy observations revealed that the biofilm-forming capacity was markedly enhanced after two days of culture. Then, we investigated into the factors contributing to biofilm formation and demonstrated that strains with more robust extracellular polymer secretion and self-agglutination capabilities exhibited a more pronounced ability to form biofilms. No significant correlation was found between surface hydrophobicity and biofilm formation capability. In addition, we found that after biofilm formation, the adhesion and invasion of cells were enhanced and drug resistance increased. Therefore, we hypothesized that the formation of biofilm makes L. monocytogenes more virulent and more difficult to remove by antibiotics. Lastly, utilizing RT-PCR, we detected the expression levels of genes associated with biofilm formation, including those involved in quorum sensing (QS), flagellar synthesis, and extracellular polymer production. These genes were significantly upregulated after biofilm formation. These findings underscore the critical relationship between extracellular polymers, self-agglutination abilities, and biofilm formation. In conclusion, the establishment of biofilms not only enhances L. monocytogenes' capacity for cell invasion and adhesion but also significantly increases its resistance to drugs, presenting a substantial threat to food safety.
Collapse
Affiliation(s)
- Yujuan Yang
- School of Life Sciences, Shanghai University, Shanghai 200444, China; (Y.Y.); (B.N.)
| | - Xiangxiang Kong
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China;
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Bing Niu
- School of Life Sciences, Shanghai University, Shanghai 200444, China; (Y.Y.); (B.N.)
| | - Jielin Yang
- Technical Centre for Animal, Plant and Food Inspection and Quarantine of Shanghai Customs, Shanghai 200135, China
| | - Qin Chen
- School of Life Sciences, Shanghai University, Shanghai 200444, China; (Y.Y.); (B.N.)
| |
Collapse
|
39
|
Oladosu VI, Park S, Sauer K. Flip the switch: the role of FleQ in modulating the transition between the free-living and sessile mode of growth in Pseudomonas aeruginosa. J Bacteriol 2024; 206:e0036523. [PMID: 38436566 PMCID: PMC10955856 DOI: 10.1128/jb.00365-23] [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] [Indexed: 03/05/2024] Open
Abstract
Pseudomonas aeruginosa is a Gram-negative, opportunistic pathogen causing chronic infections that are associated with the sessile/biofilm mode of growth rather than the free-living/planktonic mode of growth. The transcriptional regulator FleQ contributes to both modes of growth by functioning both as an activator and repressor and inversely regulating flagella genes associated with the planktonic mode of growth and genes contributing to the biofilm mode of growth. Here, we review findings that enhance our understanding of the molecular mechanism by which FleQ enables the transition between the two modes of growth. We also explore recent advances in the mechanism of action of FleQ to both activate and repress gene expression from a single promoter. Emphasis will be on the role of sigma factors, cyclic di-GMP, and the transcriptional regulator AmrZ in inversely regulating flagella and biofilm-associated genes and converting FleQ from a repressor to an activator.
Collapse
Affiliation(s)
- Victoria I. Oladosu
- Department of Biological Sciences, Binghamton University, Binghamton, New York, USA
| | - Soyoung Park
- Binghamton Biofilm Research Center, Binghamton University, Binghamton, New York, USA
| | - Karin Sauer
- Department of Biological Sciences, Binghamton University, Binghamton, New York, USA
| |
Collapse
|
40
|
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
|
41
|
Sousa AM, Ferreira D, Rodrigues LR, Pereira MO. Aptamer-based therapy for fighting biofilm-associated infections. J Control Release 2024; 367:522-539. [PMID: 38295992 DOI: 10.1016/j.jconrel.2024.01.061] [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: 09/30/2023] [Revised: 01/06/2024] [Accepted: 01/27/2024] [Indexed: 02/06/2024]
Abstract
Biofilms are key players in the pathogenesis of most of chronic infections associated with host tissue or fluids and indwelling medical devices. These chronic infections are hard to be treated due to the increased biofilms tolerance towards antibiotics in comparison to planktonic (or free living) cells. Despite the advanced understanding of their formation and physiology, biofilms continue to be a challenge and there is no standardized therapeutic approach in clinical practice to eradicate them. Aptamers offer distinctive properties, including excellent affinity, selectivity, stability, making them valuable tools for therapeutic purposes. This review explores the flexibility and designability of aptamers as antibiofilm drugs but, importantly, as targeting tools for diverse drug and delivery systems. It highlights specific examples of application of aptamers in biofilms of diverse species according to different modes of action including inhibition of motility and adhesion, blocking of quorum sensing molecules, and dispersal of biofilm-cells to planktonic state. Moreover, it discusses the limitations and challenges that impaired an increased success of the use of aptamers on biofilm management, as well as the opportunities related to aptamers modifications that can significantly expand their applicability on the biofilm field.
Collapse
Affiliation(s)
- Ana Margarida Sousa
- CEB - Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal; LABBELS - Associate Laboratory, Braga, Guimarães, Portugal.
| | - Débora Ferreira
- CEB - Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal; LABBELS - Associate Laboratory, Braga, Guimarães, Portugal
| | - Lígia Raquel Rodrigues
- CEB - Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal; LABBELS - Associate Laboratory, Braga, Guimarães, Portugal
| | - Maria Olívia Pereira
- CEB - Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal; LABBELS - Associate Laboratory, Braga, Guimarães, Portugal.
| |
Collapse
|
42
|
Cheng JH, Du R, Sun DW. Regulating bacterial biofilms in food and biomedicine: unraveling mechanisms and Innovating strategies. Crit Rev Food Sci Nutr 2024; 65:1894-1910. [PMID: 38384205 DOI: 10.1080/10408398.2024.2312539] [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] [Indexed: 02/23/2024]
Abstract
Bacterial biofilm has brought a lot of intractable problems in food and biomedicine areas. Conventional biofilm control mainly focuses on inactivation and removal of biofilm. However, with robust construction and enhanced resistance, the established biofilm is extremely difficult to eradicate. According to the mechanism of biofilm development, biofilm formation can be modulated by intervening in the key factors and regulatory systems. Therefore, regulation of biofilm formation has been proposed as an alternative way for effective biofilm control. This review aims to provide insights into the regulation of biofilm formation in food and biomedicine. The underlying mechanisms for early-stage biofilm establishment are summarized based on the key factors and correlated regulatory networks. Recent developments and applications of novel regulatory strategies such as anti/pro-biofilm agents, nanomaterials, functionalized surface materials and physical strategies are also discussed. The current review indicates that these innovative methods have contributed to effective biofilm control in a smart, safe and eco-friendly way. However, standard methodology for regulating biofilm formation in practical use is still missing. As biofilm formation in real-world systems could be far more complicated, further studies and interdisciplinary collaboration are still needed for simulation and experiments in the industry and other open systems.
Collapse
Affiliation(s)
- Jun-Hu Cheng
- School of Food Science and Engineering, South China University of Technology, Guangzhou, China
- Academy of Contemporary Food Engineering, South China University of Technology, Guangzhou Higher Education Mega Centre, Guangzhou, China
- Engineering and Technological Research Centre of Guangdong Province on Intelligent Sensing and Process Control of Cold Chain Foods, & Guangdong Province Engineering Laboratory for Intelligent Cold Chain Logistics Equipment for Agricultural Products, Guangzhou Higher Education Mega Centre, Guangzhou, China
| | - Rong Du
- School of Food Science and Engineering, South China University of Technology, Guangzhou, China
- Academy of Contemporary Food Engineering, South China University of Technology, Guangzhou Higher Education Mega Centre, Guangzhou, China
- Engineering and Technological Research Centre of Guangdong Province on Intelligent Sensing and Process Control of Cold Chain Foods, & Guangdong Province Engineering Laboratory for Intelligent Cold Chain Logistics Equipment for Agricultural Products, Guangzhou Higher Education Mega Centre, Guangzhou, China
| | - Da-Wen Sun
- School of Food Science and Engineering, South China University of Technology, Guangzhou, China
- Academy of Contemporary Food Engineering, South China University of Technology, Guangzhou Higher Education Mega Centre, Guangzhou, China
- Engineering and Technological Research Centre of Guangdong Province on Intelligent Sensing and Process Control of Cold Chain Foods, & Guangdong Province Engineering Laboratory for Intelligent Cold Chain Logistics Equipment for Agricultural Products, Guangzhou Higher Education Mega Centre, Guangzhou, China
- Food Refrigeration and Computerized Food Technology (FRCFT), Agriculture and Food Science Centre, University College Dublin, National University of Ireland, Dublin 4, Ireland
| |
Collapse
|
43
|
Chen X, Cui H, Li H, Wang J, Fu P, Yin J, Tang S, Ke Y. Functionalization of graphene oxide with amphiphilic block copolymer to enhance antibacterial activity. Colloids Surf B Biointerfaces 2024; 234:113690. [PMID: 38086276 DOI: 10.1016/j.colsurfb.2023.113690] [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: 09/14/2023] [Revised: 11/20/2023] [Accepted: 12/01/2023] [Indexed: 02/09/2024]
Abstract
Functionalization of GO with an amphiphilic block copolymer is designed with an aim to enhance its biocompatibility, however, long copolymer chains can screen the blade effect of GO to sacrifice its antimicrobial activities. To solve this problem, low molecular weight of poly(ethylene glycol) (PEG), poly(3-hydroxybutyrate-co-3-hydroxyvalerate) (PHBV) and their block copolymer were respectively introduced onto GO via an isophorone diisocyanate modified GO as a intermediate, followed by a solvent evaporation of an oil-in-water emulsion treatment (SE treatment) to induce block copolymer into polymer micelle via phase separation to refresh the sharp edges of GO. Block copolymer modified GO possessed similar dispersibility and stability to PEG modified GO, and even higher loading capacity of the hydrophobic drug than PHBV modified GO, illustrating its superior properties to homopolymer. PEG, PHBV and their block copolymer modified GO were nontoxic towards ATDC5 cells while cultured for 3 days and compatible with erythrocytes within 8 h. SE treatment enhanced greatly the loading capacity of the hydrophobic drug and the accumulative release reached 91.3% within 24 h. The inhibition zone of the block copolymer modified GO was 14.1 mm and 14.8 mm against E. coli and S. aureus, comparable to that of PEG modified GO. The bacterial reduction rate of the copolymer micelle modified GO was 87.1% and 82.7% towards E. coli and S. aureus, much greater than that of PEG, PHBV and their block copolymer modified GO at a concentration of 1 mg/mL. The antibiofilm capacity of the copolymer micelle modified GO were equal to that of PEG modified, demonstrating its great promise in tissue engineering application for repair of infected tissue defects.
Collapse
Affiliation(s)
- Xi Chen
- Department of Biomedical Engineering, Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Hao Cui
- Department of Biomedical Engineering, Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Hui Li
- Department of Biomedical Engineering, Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Jiayin Wang
- Department of Biomedical Engineering, Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Pengcheng Fu
- Department of Biomedical Engineering, Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Jun Yin
- Department of Biomedical Engineering, Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - ShunQing Tang
- Department of Biomedical Engineering, Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, College of Life Science and Technology, Jinan University, Guangzhou 510632, China.
| | - Yu Ke
- Department of Biomedical Engineering, Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, College of Life Science and Technology, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
44
|
Vohra M, Kour A, Kalia NP, Kumar M, Sharma S, Jaglan S, Kamath N, Sharma S. A comprehensive review of genomics, transcriptomics, proteomics, and metabolomic insights into the differentiation of Pseudomonas aeruginosa from the planktonic to biofilm state: A multi-omics approach. Int J Biol Macromol 2024; 257:128563. [PMID: 38070800 DOI: 10.1016/j.ijbiomac.2023.128563] [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: 07/20/2023] [Revised: 11/06/2023] [Accepted: 11/30/2023] [Indexed: 01/26/2024]
Abstract
Biofilm formation by Pseudomonas aeruginosa is primarily responsible for chronic wound and lung infections in humans. These infections are persistent owing to the biofilm's high tolerance to antimicrobials and constantly changing environmental factors. Understanding the mechanism governing biofilm formation can help to develop therapeutics explicitly directed against the molecular markers responsible for this process. After numerous years of research, many genes responsible for both in vitro and in vivo biofilm development remain unidentified. However, there is no "all in one" complete in vivo or in vitro biofilm model. Recent findings imply that the shift from planktonic bacteria to biofilms is a complicated and interrelated differentiation process. Research on the applications of omics technologies in P. aeruginosa biofilm development is ongoing, and these approaches hold great promise for expanding our knowledge of the mechanisms of biofilm formation. This review discusses the different factors that affect biofilm formation and compares P. aeruginosa biofilm formation using the omics approaches targeting essential biological macromolecules, such as DNA, RNA, Protein, and metabolome. Furthermore, we have outlined the application of currently available omics tools, such as genomics, proteomics, metabolomics, transcriptomics, and integrated multi-omics methodologies, to understand the differential gene expression (biofilm vs. planktonic bacteria) of P. aeruginosa biofilms.
Collapse
Affiliation(s)
- Mustafa Vohra
- Department of Medical Laboratory Science, Lovely Professional University, Punjab 144411, India; Department of Microbiology, Shri Vinoba Bhave Civil Hospital, Silvassa 396230, India
| | - Avleen Kour
- Department of Medical Laboratory Science, Lovely Professional University, Punjab 144411, India
| | - Nitin Pal Kalia
- Department of Biological Sciences (Pharmacology & Toxicology), National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana 500037, India
| | - Manoj Kumar
- Maternal and Child Health Program, Research Department, Sidra Medicine, Doha 122104, Qatar
| | - Sarika Sharma
- Department of Sponsored Research, Division of Research & Development, Lovely Professional University, India
| | - Sundeep Jaglan
- Fermentation & Microbial Biotechnology Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180016, J&K, India
| | - Narayan Kamath
- Department of Microbiology, Shri Vinoba Bhave Civil Hospital, Silvassa 396230, India; Department of Microbiology, NAMO Medical Education and Research Institute, Silvassa 396230, India
| | - Sandeep Sharma
- Department of Medical Laboratory Science, Lovely Professional University, Punjab 144411, India.
| |
Collapse
|
45
|
Guan C, Huang Y, Zhou Y, Han Y, Liu S, Liu S, Kong W, Wang T, Zhang Y. FlhF affects the subcellular clustering of WspR through HsbR in Pseudomonas aeruginosa. Appl Environ Microbiol 2024; 90:e0154823. [PMID: 38112425 PMCID: PMC10807432 DOI: 10.1128/aem.01548-23] [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: 09/05/2023] [Accepted: 11/04/2023] [Indexed: 12/21/2023] Open
Abstract
In bacteria, the second messenger cyclic di-GMP (c-di-GMP) is synthesized and degraded by multiple diguanylate cyclases (DGCs) and phosphodiesterases. A high level of c-di-GMP induces biofilm formation and represses motility. WspR, a hybrid response regulator DGC, produces c-di-GMP when it is phosphorylated. FlhF, a signal recognition particle-type GTPase, is initially localized to the cell poles and is indispensable for polar flagellar localization in Pseudomonas aeruginosa. In this study, we report that deletion of flhF affected biofilm formation and the c-di-GMP level in P. aeruginosa. Phenotypic analysis of a flhF knockout mutant revealed increased biofilm formation, wrinkled colonies on Congo red agar, and an elevated c-di-GMP level compared to the wild-type strain, PAO1. Yeast and bacterial two-hybrid systems showed that FlhF binds to the response regulator HsbR, and HsbR binds to WspR. Deletion of hsbR or wspR in the ΔflhF background abolished the phenotype of ΔflhF. In addition, confocal microscopy demonstrated that WspR-GFP was distributed throughout the cytoplasm and formed a visible cluster at one cell pole in PAO1 and ΔhsbR, but it was mainly distributed as visible clusters at the lateral side of the periplasm and with visible clusters at both cell poles in ΔflhF. These findings suggest that FlhF influences the subcellular cluster and localization of WspR and negatively modulates WspR DGC activity in a manner dependent on HsbR. Together, our findings demonstrate a novel mechanism for FlhF modulating the lifestyle transition between motility and biofilm via HsbR to regulate the DGC activity of WspR.IMPORTANCECyclic di-GMP (c-di-GMP) is a second messenger that controls flagellum biosynthesis, adhesion, virulence, motility, exopolysaccharide production, and biofilm formation in bacteria. Recent research has shown that distinct diguanylate cyclases (DGCs) or phosphodiesterases (PDEs) produce highly specific outputs. Some DGCs and PDEs contribute to the total global c-di-GMP concentration, but others only affect local c-di-GMP in a microenvironment. However, the underlying mechanisms are unclear. Here, we report that FlhF affects the localization and DGC activity of WspR via HsbR and is implicated in local c-di-GMP signaling in Pseudomonas aeruginosa. This study establishes the link between the c-di-GMP signaling system and the flagellar localization and provides insight for understanding the complex regulatory network of c-di-GMP signaling.
Collapse
Affiliation(s)
- Congcong Guan
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi’an, Shaanxi, China
| | - Yi Huang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi’an, Shaanxi, China
| | - Yun Zhou
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi’an, Shaanxi, China
| | - Yuqian Han
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi’an, Shaanxi, China
| | - Shuhui Liu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi’an, Shaanxi, China
| | - Shimin Liu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi’an, Shaanxi, China
| | - Weina Kong
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi’an, Shaanxi, China
| | - Tietao Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi’an, Shaanxi, China
| | - Yani Zhang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi’an, Shaanxi, China
| |
Collapse
|
46
|
Liu Q, He R, Zhang C, Zhang R, Yuan J. Bacterial surface swimming states revealed by TIRF microscopy. SOFT MATTER 2024; 20:661-671. [PMID: 38164039 DOI: 10.1039/d3sm01317k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
Motility near solid surfaces plays a key role in the life cycle of bacteria and is essential for biofilm formation, biofilm dispersal, and virulence. The alignment of the cell body with the surface during surface swimming impacts bacterial surface sensing. Here, we developed a high-throughput method for characterizing the orientation of the cell body relative to the surface using total internal reflection fluorescence (TIRF) microscopy. The angle between the cell body and the surface was determined by maximizing image cross-correlations between the TIRF image of the cell and a reference library. Utilizing this technique, we surprisingly identified six distinct surface swimming states of Pseudomonas aeruginosa according to the body alignment and the flagellar position. Furthermore, we observed that the near-surface swimming speed is greater in the pull state than in the push state, attributed to hydrodynamic effects near the liquid-solid interface. Hydrodynamic force analysis of the swimming states provided rich insights into the mechanics of bacterial surface swimming. Our technique is readily applicable to the study of surface motility across a wide spectrum of bacterial species.
Collapse
Affiliation(s)
- Qiuqian Liu
- Hefei National Research Center for Physical Sciences at the Microscale and Department of Physics, University of Science and Technology of China, Hefei, Anhui 230026, China.
| | - Rui He
- Hefei National Research Center for Physical Sciences at the Microscale and Department of Physics, University of Science and Technology of China, Hefei, Anhui 230026, China.
| | - Chi Zhang
- Hefei National Research Center for Physical Sciences at the Microscale and Department of Physics, University of Science and Technology of China, Hefei, Anhui 230026, China.
| | - Rongjing Zhang
- Hefei National Research Center for Physical Sciences at the Microscale and Department of Physics, University of Science and Technology of China, Hefei, Anhui 230026, China.
| | - Junhua Yuan
- Hefei National Research Center for Physical Sciences at the Microscale and Department of Physics, University of Science and Technology of China, Hefei, Anhui 230026, China.
| |
Collapse
|
47
|
Pool-Yam L, Ramón-Sierra J, Oliva AI, Zamora-Bustillos R, Ortiz-Vázquez E. Effect of conA-unbound proteins from Melipona beecheii honey on the formation of Pseudomonas aeruginosa ATCC 27853 biofilm. Arch Microbiol 2024; 206:54. [PMID: 38180520 DOI: 10.1007/s00203-023-03783-7] [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: 10/12/2023] [Revised: 11/20/2023] [Accepted: 12/01/2023] [Indexed: 01/06/2024]
Abstract
Pseudomonas aeruginosa is an opportunistic bacterium that can form a biofilm with the ability to colonize different surfaces and for increasing resistance to antibiotics. An alternative to solve this problem may be the use of non-glucose/mannose glycosylated proteins from Melipona beecheii honey, which are capable of inhibiting the growth of this pathogen. In this work, the antibiofilm activity of the conA-unbound protein fraction (F1) from M. beecheii was evaluated. The crude protein extract (CPE) and the F1 fraction inhibited the P. aeruginosa biofilm growth above 80% at 4 and 1.3 µg/mL, respectively. These proteins affected the structure of the biofilm, as well as fleQ and fleR gene expressions involved in the formation and regulation of the P. aeruginosa biofilm. The results demonstrated that the F1 fraction proteins of M. beecheii honey inhibit and affect the formation of the P. aeruginosa biofilm.
Collapse
Affiliation(s)
- Luis Pool-Yam
- División de Estudios de Posgrado E Investigación, Instituto Tecnológico de Conkal, Avenida Tecnológico S/N Conkal, C.P. 97345, Conkal, Yucatán, México
| | - Jesús Ramón-Sierra
- División de Estudios de Posgrado E Investigación, Instituto Tecnológico de Mérida, Av. Tecnológico Km. 4.5 S/N, C.P. 97118, Mérida, Yucatán, México
| | - A I Oliva
- Departamento de Física Aplicada, CINVESTAV-IPN, Unidad Mérida, Carretera Antigua a Progreso Km. 6, Cordemex, C.P. 97310, Mérida, Yucatán, México
| | - Roberto Zamora-Bustillos
- División de Estudios de Posgrado E Investigación, Instituto Tecnológico de Conkal, Avenida Tecnológico S/N Conkal, C.P. 97345, Conkal, Yucatán, México.
| | - Elizabeth Ortiz-Vázquez
- División de Estudios de Posgrado E Investigación, Instituto Tecnológico de Mérida, Av. Tecnológico Km. 4.5 S/N, C.P. 97118, Mérida, Yucatán, México.
| |
Collapse
|
48
|
Guo Q, Zhan Y, Zhang W, Wang J, Yan Y, Wang W, Lin M. Development and Regulation of the Extreme Biofilm Formation of Deinococcus radiodurans R1 under Extreme Environmental Conditions. Int J Mol Sci 2023; 25:421. [PMID: 38203592 PMCID: PMC10778927 DOI: 10.3390/ijms25010421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/22/2023] [Accepted: 12/24/2023] [Indexed: 01/12/2024] Open
Abstract
To grow in various harsh environments, extremophiles have developed extraordinary strategies such as biofilm formation, which is an extremely complex and progressive process. However, the genetic elements and exact mechanisms underlying extreme biofilm formation remain enigmatic. Here, we characterized the biofilm-forming ability of Deinococcus radiodurans in vitro under extreme environmental conditions and found that extremely high concentrations of NaCl or sorbitol could induce biofilm formation. Meantime, the survival ability of biofilm cells was superior to that of planktonic cells in different extreme conditions, such as hydrogen peroxide stress, sorbitol stress, and high UV radiation. Transcriptome profiles of D. radiodurans in four different biofilm development stages further revealed that only 13 matched genes, which are involved in environmental information processing, carbohydrate metabolism, or stress responses, share sequence homology with genes related to the biofilm formation of Escherichia coli, Pseudomonas aeruginosa, and Staphylococcus aureus. Overall, 64% of the differentially expressed genes are functionally unknown, indicating the specificity of the regulatory network of D. radiodurans. The mutation of the drRRA gene encoding a response regulator strongly impaired biofilm formation ability, implying that DrRRA is an essential component of the biofilm formation of D. radiodurans. Furthermore, transcripts from both the wild type and the drRRA mutant were compared, showing that the expression of drBON1 (Deinococcus radioduransBON domain-containing protein 1) significantly decreased in the drRRA mutant during biofilm development. Further analysis revealed that the drBON1 mutant lacked the ability to form biofilm and DrRRA, and as a facilitator of biofilm formation, could directly stimulate the transcription of the biofilm-related gene drBON1. Overall, our work highlights a molecular mechanism mediated by the response regulator DrRRA for controlling extreme biofilm formation and thus provides guidance for future studies to investigate novel mechanisms that are used by D. radiodurans to adapt to extreme environments.
Collapse
Affiliation(s)
- Qiannan Guo
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (Q.G.); (Y.Z.); (W.Z.); (J.W.); (Y.Y.); (W.W.)
- Key Laboratory of Agricultural Microbiome (MARA), Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Yuhua Zhan
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (Q.G.); (Y.Z.); (W.Z.); (J.W.); (Y.Y.); (W.W.)
- Key Laboratory of Agricultural Microbiome (MARA), Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Wei Zhang
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (Q.G.); (Y.Z.); (W.Z.); (J.W.); (Y.Y.); (W.W.)
- Key Laboratory of Agricultural Microbiome (MARA), Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Jin Wang
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (Q.G.); (Y.Z.); (W.Z.); (J.W.); (Y.Y.); (W.W.)
- Key Laboratory of Agricultural Microbiome (MARA), Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Yongliang Yan
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (Q.G.); (Y.Z.); (W.Z.); (J.W.); (Y.Y.); (W.W.)
- Key Laboratory of Agricultural Microbiome (MARA), Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Wenxiu Wang
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (Q.G.); (Y.Z.); (W.Z.); (J.W.); (Y.Y.); (W.W.)
- Key Laboratory of Agricultural Microbiome (MARA), Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Min Lin
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (Q.G.); (Y.Z.); (W.Z.); (J.W.); (Y.Y.); (W.W.)
- Key Laboratory of Agricultural Microbiome (MARA), Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| |
Collapse
|
49
|
Huq M, Wahid SUH, Istivan T. Biofilm Formation in Campylobacter concisus: The Role of the luxS Gene. Microorganisms 2023; 12:46. [PMID: 38257873 PMCID: PMC10820981 DOI: 10.3390/microorganisms12010046] [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: 11/30/2023] [Revised: 12/19/2023] [Accepted: 12/22/2023] [Indexed: 01/24/2024] Open
Abstract
Campylobacter concisus is a bacterium that inhabits human oral cavities and is an emerging intestinal tract pathogen known to be a biofilm producer and one of the bacterial species found in dental plaque. In this study, biofilms of oral and intestinal C. concisus isolates were phenotypically characterized. The role of the luxS gene, which is linked to the regulation of biofilm formation in other pathogens, was assessed in relation to the pathogenic potential of this bacterium. Biofilm formation capacity was assessed using phenotypic assays. Oral strains were shown to be the highest producers. A luxS mutant was created by inserting a kanamycin cassette within the luxS gene of the highest biofilm-forming isolate. The loss of the polar flagellum was observed with scanning and transmission electron microscopy (SEM and TEM). Furthermore, the luxS mutant exhibited a significant reduction (p < 0.05) in biofilm formation, motility, and its expression of flaB, in addition to the capability to invade intestinal epithelial cells, compared to the parental strain. The study concluded that C. concisus oral isolates are significantly higher biofilm producers than the intestinal isolates and that LuxS plays a role in biofilm formation, invasion, and motility in this bacterium.
Collapse
Affiliation(s)
- Mohsina Huq
- School of Science, STEM College, RMIT University, Bundoora, Melbourne, VIC 3083, Australia
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| | | | - Taghrid Istivan
- School of Science, STEM College, RMIT University, Bundoora, Melbourne, VIC 3083, Australia
| |
Collapse
|
50
|
Kalia M, Amari D, Davies DG, Sauer K. cis-DA-dependent dispersion by Pseudomonas aeruginosa biofilm and identification of cis-DA-sensory protein DspS. mBio 2023; 14:e0257023. [PMID: 38014955 PMCID: PMC10746223 DOI: 10.1128/mbio.02570-23] [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: 09/20/2023] [Accepted: 10/11/2023] [Indexed: 11/29/2023] Open
Abstract
IMPORTANCE Dispersion is an essential stage of the biofilm life cycle resulting in the release of bacteria from a biofilm into the surrounding environment. Dispersion contributes to bacterial survival by relieving overcrowding within a biofilm and allowing dissemination of cells into new habitats for colonization. Thus, dispersion can contribute to biofilm survival as well as disease progression and transmission. Cells dispersed from a biofilm rapidly lose their recalcitrant antimicrobial-tolerant biofilm phenotype and transition to a state that is susceptible to antibiotics. However, much of what is known about this biofilm developmental stage has been inferred from exogenously induced dispersion. Our findings provide the first evidence that native dispersion is coincident with reduced cyclic dimeric guanosine monophosphate levels, while also relying on at least some of the same factors that are central to the environmentally induced dispersion response, namely, BdlA, DipA, RbdA, and AmrZ. Additionally, we demonstrate for the first time that cis-DA signaling to induce dispersion is attributed to the two-component sensor/response regulator DspS, a homolog of the DSF sensor RpfC. Our findings also provide a path toward manipulating the native dispersion response as a novel and highly promising therapeutic intervention.
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
| | - Diana Amari
- Department of Biological Sciences, Binghamton University, Binghamton, New York, USA
- Binghamton Biofilm Research Center, Binghamton University, Binghamton, New York, USA
| | - David G. Davies
- 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
|