1
|
Shen Y, Xiao Y, Xie E, Puig-Bargués J, Yao Y, Kuang N, Li Y. Biofouling control strategy through denatured extracellular proteins: An empirical evidence from reclaimed water distribution systems. WATER RESEARCH 2025; 280:123538. [PMID: 40156976 DOI: 10.1016/j.watres.2025.123538] [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: 02/03/2025] [Revised: 03/18/2025] [Accepted: 03/21/2025] [Indexed: 04/01/2025]
Abstract
Biofouling remains a significant challenge in water treatment fields, leading to a decline in the hydraulic performance, increased operational costs, and potential health risks. Previous biofouling control strategies primarily focused on the removal of particulates and microorganisms, often neglecting the role of extracellular proteins. Using a reclaimed water distribution system as an example, this study proposes a strategy to inhibit biofouling formation by utilizing urea, a reported protein denaturant with fertilizer functionality. Results indicated that urea significantly slowed the accumulation of biofouling, leading to a 16.4-49.4 % decrease in biofouling weight, an 18.6-55.3 % decrease in extracellular protein content, and a 25.9-45.3 % reduction in extracellular polymer substance (EPS) content. Urea mitigated biofouling through two mechanisms: (1) disrupting protein structures, which convert tightly bound EPS to loosely bound EPS, and (2) downregulating biofilm-forming signaling proteins, thereby inhibiting biofouling formation. In the process, proteins, polysaccharides, and microorganisms exhibited clear mutual promotion relationships. Additionally, urea weakened microbial symbiotic interactions by affecting protein signaling molecules, inhibiting microbial growth and polysaccharide metabolism. The research confirms that denaturing extracellular proteins to mitigate biofouling is a feasible and efficient approach. The findings aim to provide valuable insights for the development of sustainable and effective biofouling cleaning strategies.
Collapse
Affiliation(s)
- Yan Shen
- State Key Laboratory of Efficient Utilization of Agricultural Water Resources, China Agricultural University, Beijing 100083, PR China; Engineering Research Center for Agricultural Water-Saving and Water Resources, Ministry of Education, Beijing 100083, PR China
| | - Yang Xiao
- State Key Laboratory of Efficient Utilization of Agricultural Water Resources, China Agricultural University, Beijing 100083, PR China; Engineering Research Center for Agricultural Water-Saving and Water Resources, Ministry of Education, Beijing 100083, PR China
| | - En Xie
- State Key Laboratory of Efficient Utilization of Agricultural Water Resources, China Agricultural University, Beijing 100083, PR China; Engineering Research Center for Agricultural Water-Saving and Water Resources, Ministry of Education, Beijing 100083, PR China
| | - Jaume Puig-Bargués
- Department of Chemical and Agricultural Engineering and Technology, University of Girona, Girona 17003, Spain
| | - Yuqian Yao
- State Key Laboratory of Efficient Utilization of Agricultural Water Resources, China Agricultural University, Beijing 100083, PR China; Engineering Research Center for Agricultural Water-Saving and Water Resources, Ministry of Education, Beijing 100083, PR China
| | - Naikun Kuang
- State Key Laboratory of Efficient Utilization of Agricultural Water Resources, China Agricultural University, Beijing 100083, PR China; Engineering Research Center for Agricultural Water-Saving and Water Resources, Ministry of Education, Beijing 100083, PR China
| | - Yunkai Li
- State Key Laboratory of Efficient Utilization of Agricultural Water Resources, China Agricultural University, Beijing 100083, PR China; Engineering Research Center for Agricultural Water-Saving and Water Resources, Ministry of Education, Beijing 100083, PR China.
| |
Collapse
|
2
|
Wareham-Mathiassen S, Nateqi M, Badrinarayanan SA, Glenting VP, Dragheim MB, Agner AR, Rasmussen TS, Bay L, Jelsbak L, Bengtsson H, Bjarnsholt T. Evaluating antimicrobial efficacy in medical devices: The critical role of simulating in use test conditions. BIOMATERIALS ADVANCES 2025; 172:214241. [PMID: 40010022 DOI: 10.1016/j.bioadv.2025.214241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 02/28/2025]
Abstract
Biofilm infections represent the greatest risk associated with medical devices and implants, constituting 65 %-70 % of all device associated infections. Efforts to develop antimicrobial technologies for biomedical applications aim to reduce infection rates, antibiotic use, and the induction of antimicrobial resistance. However, standard laboratory test conditions often overestimate efficacy, highlighting the need for experimental designs that simulate real-world settings. To this end, we evaluated four commercially available antimicrobial materials containing silver (AG1, AG2, AG3) or zinc (ZN1) to assess their ability to mitigate microbial proliferation in for longer duration or multi-use medical devices. The materials' homogeneity and surface topography were characterized through Scanning Electron Microscopy (SEM) coupled with Energy Dispersive Spectroscopy (EDS) and Atomic Force Microscopy (AFM). Antimicrobial efficacy was tested using a modified ISO 22196 protocol under clinically relevant conditions and a dry contact test developed to mimic in-use conditions for many extracorporeal medical devices. Results revealed homogeneous elemental distributions in AG1, AG2, and ZN1, and heterogeneous clusters for AG3. Surface roughness was highest for AG2 (170.1 nm), followed by TPE control (155.3 nm), ZN1 (83.51 nm) and silicone control (66.74 nm). All test materials demonstrated antimicrobial efficacies against S. aureus and E. coli, but not against C. albicans. In the dry contact assay, only AG2 proved effective against E. coli, and P. aeruginosa, underlining the role of humidity in antimicrobial action. Results were further corroborated by measurement of ion release by the materials at various temperatures, revealing greater release at higher temperatures. These outcomes emphasize the importance of testing antimicrobial materials under in use conditions to minimize discrepancies between laboratory results and clinical outcomes. Our findings provide a valuable framework for testing and integrating these materials into next-generation multi-use medical devices.
Collapse
Affiliation(s)
- Sofia Wareham-Mathiassen
- Department of Immunology and Microbiology, Copenhagen University, Copenhagen, Denmark; Department of Technology Exploration, Devices & Delivery Solutions, Novo Nordisk A/S, Bagsværd, Denmark.
| | - Mohammed Nateqi
- Department of Immunology and Microbiology, Copenhagen University, Copenhagen, Denmark; Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads bldg. 221, DK-2800 Kgs Lyngby, Denmark; Department of Engineering, Devices & Delivery Solutions, Novo Nordisk A/S, Bagsværd, Denmark
| | - Sai Achyuth Badrinarayanan
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads bldg. 221, DK-2800 Kgs Lyngby, Denmark
| | - Vera Pinto Glenting
- Department of Microbiology, Devices & Delivery Solutions, Novo Nordisk A/S, Bagsværd, Denmark
| | | | | | - Tina Secher Rasmussen
- Leachables & Elemental Impurities, Department of Chemistry, Manufacturing, and Controls, Novo Nordisk A/S, Bagsværd, Denmark
| | - Lene Bay
- Costerton Biofilm Center, Department of Immunology and Microbiology, Faculty of Medical and Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lars Jelsbak
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads bldg. 221, DK-2800 Kgs Lyngby, Denmark
| | - Henrik Bengtsson
- Bioinnovation Hub, Devices & Delivery Solutions, Novo Nordisk A/S, Bagsværd, Denmark
| | - Thomas Bjarnsholt
- Department of Immunology and Microbiology, Copenhagen University, Copenhagen, Denmark; Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
3
|
Wang H, Sun X, Wang Y, Shi W, Wu L, Miao L. Marine steel protection based on biomineralization for sustainable development of coastal cities. BIORESOURCE TECHNOLOGY 2025; 428:132404. [PMID: 40139470 DOI: 10.1016/j.biortech.2025.132404] [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: 01/01/2025] [Revised: 03/13/2025] [Accepted: 03/14/2025] [Indexed: 03/29/2025]
Abstract
Corrosion research, spanning over 150 years, remains critically important, particularly for addressing marine microbially induced corrosion on steel, which causes significant economic losses and safety risks. This study proposes a biomineralization method using marine urease-producing bacteria to protect steel. Urease-producing bacteria were enriched to promote biomineralization, and a seawater corrosion experiment was conducted to evaluate its efficacy. Results showed that biomineralization significantly reduced corrosion rates, especially with yeast extract enrichment, and decreased the abundance of sulfate-reducing bacteria and sulfur-oxidizing bacteria in biofilms. Functional gene analysis identified Thioalkalivibrio as a key indicator of sulfate reduction. The findings demonstrated that the formed biomineralized film acted as a protective layer to isolate the steel from the corrosive seawater, which contributed to the advancement of novel techniques for corrosion inhibition of marine steel to achieve long-term sustainability for ships and engineering structures.
Collapse
Affiliation(s)
- Hengxing Wang
- Institute of Geotechnical Engineering, Southeast University, Nanjing, Jiangsu, China
| | - Xiaohao Sun
- Institute of Geotechnical Engineering, Southeast University, Nanjing, Jiangsu, China; Department of Civil and Environmental Engineering, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong Special Administrative Region.
| | - Yong Wang
- Department of Civil and Environmental Engineering, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong Special Administrative Region
| | - Wenbo Shi
- School of Intelligent Transportation, Xuchang University, Xuchang, Henan, China
| | - Linyu Wu
- School of Civil Engineering and Architecture, Wuhan Polytechnic University, Wuhan, Hubei, China
| | - Linchang Miao
- Institute of Geotechnical Engineering, Southeast University, Nanjing, Jiangsu, China.
| |
Collapse
|
4
|
Farrukh M, Munawar A, Nawaz Z, Hussain N, Hafeez AB, Szweda P. Antibiotic resistance and preventive strategies in foodborne pathogenic bacteria: a comprehensive review. Food Sci Biotechnol 2025; 34:2101-2129. [PMID: 40351726 PMCID: PMC12064539 DOI: 10.1007/s10068-024-01767-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 11/08/2024] [Accepted: 11/18/2024] [Indexed: 05/14/2025] Open
Abstract
Antibiotic resistance in foodborne bacteria poses a substantial global health challenge. Reports indicate that antibiotic overuse in middle-class and low-income countries is a significant factor in the ever-increasing resistance. Resistance mechanisms have developed through enzymatic hydrolysis, reduced membrane permeability, efflux pumps, and target site mutations. Preventive measures like proper hygiene and safe food preparation, vaccination, antibiotic stewardship and surveillance, implementing infection prevention and control (IPC) measures, good agricultural practices, and investigating novel approaches like CRISPR, NGS, nanotechnology, and bacteriophages may be employed to address this challenge. Naturally occurring preservatives (e.g., nisin) are alternatives to antibiotics for food preservation. Prebiotics, probiotics, nanobiotics, phage treatment, and antimicrobial peptides are also substitutes for antibiotics. Furthermore, plant-derived compounds, such as essential oils and plant extracts, are promising substitutes for antibiotics in animal production. This review focuses on the mechanisms of underlying antibiotic resistance in foodborne pathogens, necessary preventive measures, and the challenges associated. Graphical abstract Created using BioRender https://www.biorender.com/.
Collapse
Affiliation(s)
- Masooma Farrukh
- Center for Applied Molecular Biology (CAMB), University of the Punjab, Lahore, Pakistan
| | - Ayesha Munawar
- Center for Applied Molecular Biology (CAMB), University of the Punjab, Lahore, Pakistan
| | - Zeenat Nawaz
- Center for Applied Molecular Biology (CAMB), University of the Punjab, Lahore, Pakistan
| | - Nazim Hussain
- Center for Applied Molecular Biology (CAMB), University of the Punjab, Lahore, Pakistan
| | - Ahmer Bin Hafeez
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry, Gdańsk University of Technology, Ul. G. Narutowicza 11/12, 80-233 Gdańsk, Poland
| | - Piotr Szweda
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry, Gdańsk University of Technology, Ul. G. Narutowicza 11/12, 80-233 Gdańsk, Poland
| |
Collapse
|
5
|
He X, Zhang W, Liu J, Liu J, Chen Y, Luan C, Zhang J, Bao G, Lin X, Muh F, Lin T, Lu F. The global regulatory role of RsbUVW in virulence and biofilm formation in MRSA. Microb Pathog 2025; 203:107508. [PMID: 40158706 DOI: 10.1016/j.micpath.2025.107508] [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: 12/26/2024] [Revised: 03/22/2025] [Accepted: 03/23/2025] [Indexed: 04/02/2025]
Abstract
The widespread prevalence of methicillin-resistant Staphylococcus aureus (MRSA) has caused serious challenges to clinical treatment. This study was designed to explore effective targets for MRSA prevention and control. The key virulence regulator was screened through the correlation analysis between virulence and various regulatory factors in the main clinical epidemic MRSA. The potential key factors were inactivated to further evaluate the inhibitory effect on the virulence of MRSA standard strain S. aureus ATCC43300 and its influence on drug resistance and biofilm formation. Enterobacterial repetitive intergenic consensus-PCR was used to analyze the clinical epidemic genotypes of MRSA. The virulence of MRSA was evaluated mainly by measuring its adhesion and invasion ability to A549 cells, the lethality to Galleria mellonella larvae, and the transcription level of related genes. The biofilm formation was assessed by crystal violet staining on polystyrene microplates. The results showed that most virulence factors of clinical representative MRSA strain were significantly positively correlated with RsbUVW system. After knocking out the rsbV gene, a key component of the rsbUVW system, the virulence of S. aureus ATCC43300 was significantly reduced (P < 0.05), as indicated by a significant decrease in lethality against G. mellonella larvae and invasion against A549 cells, and a significant decrease in the expression of immune escape related virulence factors polysaccharide intercellular adhesin (PIA) and staphyloxanthin. The biomass and stability of protein-dependent biofilm by S. aureus ATCC43300 were significantly increased. This study will provide useful information for the effective prevention and control of MRSA.
Collapse
Affiliation(s)
- Xinlong He
- Department of Pathogenic Biology, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China; Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou, 225001, China; The Key Laboratory of the Jiangsu Higher Education Institutions for Nucleic Acid & Cell Fate Regulation (Yangzhou University), Yangzhou, 225001, China; Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225001, China
| | - Wenwen Zhang
- Department of Pathogenic Biology, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China; Department of Clinical Laboratory, Changning Maternity and Infant Health Hospital, Affiliated Hospital of East China Normal University, Shanghai, 200050, China
| | - Jie Liu
- Department of Pathogenic Biology, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China
| | - Jiali Liu
- Department of Pathogenic Biology, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China
| | - Yinsong Chen
- Department of Lung, Third People's Hospital of Yangzhou, Yangzhou, China
| | - Changjiao Luan
- Department of Lung, Third People's Hospital of Yangzhou, Yangzhou, China
| | - Jun Zhang
- Department of Clinical Laboratory, First Affiliated Hospital of Yangzhou University, Yangzhou, 225000, China
| | - Guangyu Bao
- Department of Clinical Laboratory, First Affiliated Hospital of Yangzhou University, Yangzhou, 225000, China
| | - Xiangfang Lin
- Department of Clinical Laboratory, First Affiliated Hospital of Yangzhou University, Yangzhou, 225000, China
| | - Fauzi Muh
- Department of Epidemiology & Tropical Diseases, Faculty of Public Health, Universitas Diponegoro, Tembalang, Semarang, 50275, Indonesia
| | - Tao Lin
- Department of Clinical Laboratory, First Affiliated Hospital of Yangzhou University, Yangzhou, 225000, China.
| | - Feng Lu
- Department of Pathogenic Biology, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China.
| |
Collapse
|
6
|
Ribeiro PDS, Stasko J, Shircliff A, Fernandes LG, Putz EJ, Andreasen C, Azevedo V, Ristow P, Nally JE. Investigations into the growth and formation of biofilm by Leptospira biflexa at temperatures encountered during infection. Biofilm 2025; 9:100243. [PMID: 39758814 PMCID: PMC11697785 DOI: 10.1016/j.bioflm.2024.100243] [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: 10/07/2024] [Revised: 12/05/2024] [Accepted: 12/09/2024] [Indexed: 01/07/2025] Open
Abstract
The genus Leptospira comprises unique atypical spirochete bacteria that includes the etiological agent of leptospirosis, a globally important zoonosis. Biofilms are microecosystems composed of microorganisms embedded in a self-produced matrix that offers protection against hostile factors. Leptospires form biofilms in vitro, in situ in rice fields and unsanitary urban areas, and in vivo while colonizing rodent kidneys. The complex three-dimensional biofilm matrix includes secreted polymeric substances such as proteins, extracellular DNA (eDNA), and saccharides. The genus Leptospira comprises pathogenic and saprophytic species with the saprophytic L. biflexa being commonly used as a model organism for the genus. In this study, the growth and formation of biofilms by L. biflexa was investigated not just at 29 °C, but at 37 °C/5 % CO2, a temperature similar to that encountered during host infection. Planktonic free-living L. biflexa grow in HAN media at both 29 °C and 37 °C/5 % CO2, but cells grown at 37 °C/5 % CO2 are longer (18.52 μm ± 3.39) compared to those at 29 °C (13.93 μm ± 2.84). Biofilms formed at 37 °C/5 % CO2 had more biomass compared to 29 °C, as determined by crystal violet staining. Confocal microscopy determined that the protein content within the biofilm matrix was more prominent than double-stranded DNA, and featured a distinct layer attached to the solid substrate. Additionally, the model enabled effective protein extraction for proteomic comparison across different biofilm phenotypes. Results highlight an important role for proteins in biofilm matrix structure by leptospires and the identification of their specific protein components holds promise for strategies to mitigate biofilm formation.
Collapse
Affiliation(s)
- Priscyla dos Santos Ribeiro
- Federal University of Minas Gerais, Belo Horizonte, Brazil
- Federal University of Bahia, National Institute of Science and Technology in Interdisciplinary and Transdisciplinary Studies in Ecology and Evolution, Salvador, Brazil
| | - Judith Stasko
- Infectious Bacterial Diseases Research Unit, USDA Agriculture Research Service, National Animal Disease Center, Ames, IA, USA
| | - Adrienne Shircliff
- Infectious Bacterial Diseases Research Unit, USDA Agriculture Research Service, National Animal Disease Center, Ames, IA, USA
| | - Luis Guilherme Fernandes
- Infectious Bacterial Diseases Research Unit, USDA Agriculture Research Service, National Animal Disease Center, Ames, IA, USA
| | - Ellie J. Putz
- Infectious Bacterial Diseases Research Unit, USDA Agriculture Research Service, National Animal Disease Center, Ames, IA, USA
| | - Claire Andreasen
- Department of Veterinary Pathology, College of Veterinary Medicine, Ames, IA, USA
| | - Vasco Azevedo
- Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Paula Ristow
- Federal University of Bahia, National Institute of Science and Technology in Interdisciplinary and Transdisciplinary Studies in Ecology and Evolution, Salvador, Brazil
| | - Jarlath E. Nally
- Infectious Bacterial Diseases Research Unit, USDA Agriculture Research Service, National Animal Disease Center, Ames, IA, USA
| |
Collapse
|
7
|
Yang Y, Yan J, Olson R, Jiang X. Comprehensive genomic and evolutionary analysis of biofilm matrix clusters and proteins in the Vibrio genus. mSystems 2025; 10:e0006025. [PMID: 40207939 DOI: 10.1128/msystems.00060-25] [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: 01/16/2025] [Accepted: 03/12/2025] [Indexed: 04/11/2025] Open
Abstract
Vibrio cholerae pathogens cause cholera, an acute diarrheal disease resulting in significant morbidity and mortality worldwide. Biofilms in vibrios enhance their survival in natural ecosystems and facilitate transmission during cholera outbreaks. Critical components of the biofilm matrix include the Vibrio polysaccharides produced by the vps-1 and vps-2 gene clusters and the biofilm matrix proteins encoded in the rbm gene cluster, together comprising the biofilm matrix cluster. However, the biofilm matrix clusters and their evolutionary patterns in other Vibrio species remain underexplored. In this study, we systematically investigated the distribution, diversity, and evolution of biofilm matrix clusters and proteins across the Vibrio genus. Our findings reveal that these gene clusters are sporadically distributed throughout the genus, even appearing in species phylogenetically distant from Vibrio cholerae. Evolutionary analysis of the major biofilm matrix proteins RbmC and Bap1 shows that they are structurally and sequentially related, having undergone structural domain and modular alterations. Additionally, a novel loop-less Bap1 variant was identified, predominantly represented in two phylogenetically distant V. cholerae subspecies clades that share specific gene groups associated with the presence or absence of the protein. Furthermore, our analysis revealed that rbmB, a gene involved in biofilm dispersal, shares a recent common ancestor with Vibriophage tail proteins, suggesting that phages may mimic host functions to evade biofilm-associated defenses. Our study offers a foundational understanding of the diversity and evolution of biofilm matrix clusters in vibrios, laying the groundwork for future biofilm engineering through genetic modification. IMPORTANCE Biofilms help vibrios survive in nature and spread cholera. However, the genes that control biofilm formation in vibrios other than Vibrio cholerae are not well understood. In this study, we analyzed the biofilm matrix gene clusters and proteins across diverse Vibrio species to explore their patterns and evolution. We discovered that these genes are spread across different Vibrio species, including those not closely related to V. cholerae. We also found various forms of key biofilm proteins with different structures. Additionally, we identified genes involved in biofilm dispersal that are related to vibriophage genes, highlighting the role of phages in biofilm development. This study not only provides a foundational understanding of biofilm diversity and evolution in vibrios but also leads to new strategies for engineering biofilms through genetic modification, which is crucial for managing cholera outbreaks and improving the environmental resilience of these bacteria.
Collapse
Affiliation(s)
- Yiyan Yang
- Intramural Research Program, National Library of Medicine, National Institutes of Health, Bethesda, Maryland, USA
| | - Jing Yan
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, Connecticut, USA
- Quantitative Biology Institute, Yale University, New Haven, Connecticut, USA
| | - Rich Olson
- Department of Molecular Biology and Biochemistry, Molecular Biophysics Program, Wesleyan University, Middletown, Connecticut, USA
| | - Xiaofang Jiang
- Intramural Research Program, National Library of Medicine, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
8
|
Zhou ZY, Bai SJ, He J, Xiong QX, Zhong ZD, Lu CW, Kuang LF, Jian ZR, Gu JL, Liu MZ, Li PF, Wang EL, Wang GX, Ling F, Yu Q, Liu T. Pathogenicity, ultrastructure and genomics analysis of Nocardia Seriolae isolated from largemouth bass (Micropterus salmoides). Microb Pathog 2025:107715. [PMID: 40383242 DOI: 10.1016/j.micpath.2025.107715] [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: 03/05/2025] [Revised: 05/09/2025] [Accepted: 05/16/2025] [Indexed: 05/20/2025]
Abstract
Fish granulomatous diseases induced by Nocardia seriolae have caused significant economic losses in global aquaculture. Understanding the structural and genetic basis of N. seriolae' s survival and immune evasion strategies is crucial. However, while some aspects of its pathogenicity have been explored, a comprehensive characterization of its ultrastructural features and the associated genetic determinants, especially in direct correlation, has remained incompletely elucidated. In this study, we utilized specialized staining methods to further characterize the pathology of chronic granulomatous inflammation induced by N. seriolae in largemouth bass (Micropterus salmoides). Using scanning electron microscope (SEM) and transmission electron microscope (TEM), we revealed further insights into the morphological (surface structure, cell wall) and intracellular features (mesosomes, ribosomes, lipids) of N. seriolae, alongside diverse division patterns. Through genomic analysis, the gene clusters related to cell wall synthesis and cell division were characterized, including peptidoglycan, arabinogalactan, mycolic acids (MAs) synthesis, and the division and cell wall (DCW) gene cluster, highlighting the conservation and potential functions of these genes in these processes. In conclusion, these findings provide crucial foundational knowledge regarding the ultrastructure and genetic determinants potentially involved in the pathogenicity of N. seriolae. By integrating detailed ultrastructural observations with genomic insights, this study enhances our understanding of its complex biology and aims to strengthen the foundation for the development of prevention, control, and detection methods for fish granulomatous diseases.
Collapse
Affiliation(s)
- Zheng-Yang Zhou
- Hainan Institute of Northwest A&F University, Sanya, Hainan Province 572024, P.R. China; College of Animal Science and Technology, Northwest A&F University, Xinong Road 22nd, Yangling, Shaanxi, 712100, P.R. China; Engineering Research Center of the Innovation and Development of Green Fishery Drugs, Universities of Shaanxi Province, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Shang-Jie Bai
- Hainan Institute of Northwest A&F University, Sanya, Hainan Province 572024, P.R. China; College of Animal Science and Technology, Northwest A&F University, Xinong Road 22nd, Yangling, Shaanxi, 712100, P.R. China; Engineering Research Center of the Innovation and Development of Green Fishery Drugs, Universities of Shaanxi Province, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Jie He
- Chengdu Lilai Biotechnology Co., Ltd., Chengdu, Sichuan 610000, P.R. China
| | - Quan-Xin Xiong
- Chengdu Lilai Biotechnology Co., Ltd., Chengdu, Sichuan 610000, P.R. China
| | - Zhen-Dong Zhong
- Chengdu Lilai Biotechnology Co., Ltd., Chengdu, Sichuan 610000, P.R. China
| | - Chen-Wang Lu
- Hainan Institute of Northwest A&F University, Sanya, Hainan Province 572024, P.R. China; College of Animal Science and Technology, Northwest A&F University, Xinong Road 22nd, Yangling, Shaanxi, 712100, P.R. China; Engineering Research Center of the Innovation and Development of Green Fishery Drugs, Universities of Shaanxi Province, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Lin-Feng Kuang
- Hainan Institute of Northwest A&F University, Sanya, Hainan Province 572024, P.R. China; College of Animal Science and Technology, Northwest A&F University, Xinong Road 22nd, Yangling, Shaanxi, 712100, P.R. China; Engineering Research Center of the Innovation and Development of Green Fishery Drugs, Universities of Shaanxi Province, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Zheng-Ran Jian
- College of Animal Science and Technology, Northwest A&F University, Xinong Road 22nd, Yangling, Shaanxi, 712100, P.R. China
| | - Jin-Lai Gu
- College of Animal Science and Technology, Northwest A&F University, Xinong Road 22nd, Yangling, Shaanxi, 712100, P.R. China
| | - Ming-Zhu Liu
- Guangxi Key Laboratory of Aquatic Biotechnology and Modern Ecological Aquaculture, Guangxi Academy of Sciences, Nanning 530007, P.R. China
| | - Peng-Fei Li
- Guangxi Key Laboratory of Aquatic Biotechnology and Modern Ecological Aquaculture, Guangxi Academy of Sciences, Nanning 530007, P.R. China
| | - Er-Long Wang
- Hainan Institute of Northwest A&F University, Sanya, Hainan Province 572024, P.R. China; College of Animal Science and Technology, Northwest A&F University, Xinong Road 22nd, Yangling, Shaanxi, 712100, P.R. China; Engineering Research Center of the Innovation and Development of Green Fishery Drugs, Universities of Shaanxi Province, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Gao-Xue Wang
- Hainan Institute of Northwest A&F University, Sanya, Hainan Province 572024, P.R. China; College of Animal Science and Technology, Northwest A&F University, Xinong Road 22nd, Yangling, Shaanxi, 712100, P.R. China; Engineering Research Center of the Innovation and Development of Green Fishery Drugs, Universities of Shaanxi Province, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Fei Ling
- Hainan Institute of Northwest A&F University, Sanya, Hainan Province 572024, P.R. China; College of Animal Science and Technology, Northwest A&F University, Xinong Road 22nd, Yangling, Shaanxi, 712100, P.R. China; Engineering Research Center of the Innovation and Development of Green Fishery Drugs, Universities of Shaanxi Province, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Qing Yu
- Guangxi Key Laboratory of Aquatic Biotechnology and Modern Ecological Aquaculture, Guangxi Academy of Sciences, Nanning 530007, P.R. China.
| | - Tao Liu
- Hainan Institute of Northwest A&F University, Sanya, Hainan Province 572024, P.R. China; College of Animal Science and Technology, Northwest A&F University, Xinong Road 22nd, Yangling, Shaanxi, 712100, P.R. China; Engineering Research Center of the Innovation and Development of Green Fishery Drugs, Universities of Shaanxi Province, Northwest A&F University, Yangling, Shaanxi 712100, China.
| |
Collapse
|
9
|
Lipke PN. Not gently down the stream: flow induces amyloid bonding in environmental and pathological fungal biofilms. mBio 2025:e0020325. [PMID: 40377304 DOI: 10.1128/mbio.00203-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2025] Open
Abstract
Surface-bound biofilms are the predominant microbial life form in the environment and host organisms. Many biofilms survive and thrive under physical stress from liquid flow in streams, fuel lines, blood, and airways. Strategies for biofilm persistence include shear-dependent adhesion (called catch bonding). In some cases, biofilms are physically strengthened by the formation of cross-β bonds between proteins: the same process that generates amyloids. Cross-β bonds have low dissociation rates. In biofilms, they bind cells to substrates, each other, and the biofilm matrix. Most fungal adhesins include amino acid sequences that can form amyloids. Shear flow activates these adhesins by unfolding pseudo-stable protein domains. The unfolding exposes sequence segments that can form cross-β bonds. These segments interact to form high-avidity adhesin patches on the cell surface. Thus, cross-β bonding is a consequence of flow-induced exposure of the cross-β core sequences. Liquid flow leads to both biofilm establishment through catch bonding and biofilm strengthening through amyloid-like bonds. This shear-dependent induction of biofilm establishment and persistence is a model for many microbial systems.IMPORTANCEThe microbes in biofilms persist in many environments, including industrial and pathological settings. These surface-associated communities show high resistance to antibiotics and microbicides. Biofilms also resist scouring by liquid flow. Amyloid-like cross-β bonds allow the establishment, strengthening, and persistence of many biofilms. This discovery opens a window on the novel use of anti-amyloid strategies to control microbes in biofilms.
Collapse
Affiliation(s)
- Peter N Lipke
- Biology Department, Brooklyn College of the City University of New York, Brooklyn, New York, USA
| |
Collapse
|
10
|
Yan X, Lin Z, Shen H, Chen Y, Chen L. Photo-responsive antibacterial metal organic frameworks. J Mater Chem B 2025. [PMID: 40370037 DOI: 10.1039/d5tb00105f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2025]
Abstract
The misuse and overuse of antibiotics have caused the emergence of antibiotic-resistant bacteria, making bacterial infections more challenging. The increasing prevalence of multidrug-resistant pathogens has driven researchers to explore novel therapeutic strategies. Phototherapy strategies that utilize photo-responsive biomaterials for their antibacterial properties have gained widespread attention due to their capability of precisely controlling bacterial inactivation with minimal side effects. Despite their potential, photodynamic therapies suffer from phototoxicity and low efficiency of photosensitizers, while photothermal therapy risks overheating, which may harm healthy tissues, thus restricting its broader application. Metal organic frameworks (MOFs) have unique physicochemical properties, which provide a promising way to deal with these challenges. MOFs can function as reservoirs, loading and releasing antibacterial agents, such as antibiotics or metal ions, upon light illumination by virtue of their metastable coordination bonds. Their porous structures enable controlled drug release and encapsulation of photosensitizers. Furthermore, MOFs' tunable composition and pore structure allow for the light-triggered generation of heat and reactive oxygen species, enhancing their antibacterial effectiveness. By doping MOFs with functional materials, it is possible to achieve multi-mode antibacterial effects. In this review, we will outline recent advancements of photo-responsive antibacterial MOFs, categorize their underlying mechanisms of action and highlight their prospects in addressing bacterial resistance.
Collapse
Affiliation(s)
- Xiaojie Yan
- School of Life Sciences, Shanghai University, Shanghai 200444, China.
| | - Zhengzheng Lin
- School of Life Sciences, Shanghai University, Shanghai 200444, China.
| | - He Shen
- School of Life Sciences, Shanghai University, Shanghai 200444, China.
| | - Yu Chen
- School of Life Sciences, Shanghai University, Shanghai 200444, China.
| | - Liang Chen
- School of Life Sciences, Shanghai University, Shanghai 200444, China.
- State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200438, P. R. China
| |
Collapse
|
11
|
Zhang X, Zhai Y, Zhu J, Zhu Z, Wen Y, Gao Q, Wang L, Lin J, Qian Y, Chen L, Du H. Regulation of type 3 fimbria expression by RstA affects biofilm formation and virulence in Klebsiella pneumoniae ATCC43816. Microbiol Spectr 2025:e0307624. [PMID: 40372035 DOI: 10.1128/spectrum.03076-24] [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: 12/07/2024] [Accepted: 04/02/2025] [Indexed: 05/16/2025] Open
Abstract
Klebsiella pneumoniae causes both community-acquired and healthcare-associated infections, presenting a major therapeutic challenge to global public health. RstBA is a common two-component regulatory system that controls downstream gene expression in certain Enterobacteriaceae species. However, the role of RstBA in K. pneumoniae infection remains unknown. To determine its function, a wild-type K. pneumoniae strain (ATCC43816) and rstA mutant and complementation strains were constructed. Phenotypic experiments and in vivo animal infection assays demonstrated that deletion of rstA decreased virulence and biofilm formation. RNA sequencing analysis of ATCC43816 and rstA mutant strains was performed to study the regulatory mechanisms, revealing differential expression of genes involved in arginine and proline metabolism, phenylalanine metabolism, and quorum sensing. In addition, the mrkI and the mrkABCDF gene cluster, which regulates and encodes type 3 fimbriae, exhibited lower expression in the absence of rstA, possibly related to decreased virulence and biofilm formation. Quantitative real-time reverse transcription PCR, promoter activity assays, and electrophoretic mobility shift assays were conducted to identify the transcriptional regulation of mrkI and mrkABCDF by rstA. Our findings show that rstA regulates type 3 fimbriae expression by regulating mrkI indirectly and regulating mrkA directly by binding to its promoter. This study provides new insights into the functional importance of RstA in regulating biofilm formation and virulence in K. pneumoniae.IMPORTANCEKlebsiella pneumoniae is an opportunistic pathogen that has become a significant cause of community-acquired and nosocomial infections. The rise of hypervirulent and multi-drug-resistant K. pneumoniae poses a significant threat to public health. The two-component regulatory system is a typical signal-sensing and stress-response system widely distributed in bacteria, playing a critical regulatory role in bacterial infection. Through in vivo and in vitro experiments, we demonstrate that rstA regulates the expression of type 3 fimbriae by regulating mrkI indirectly and mrkA directly, thereby playing an essential role in the virulence and biofilm formation of K. pneumoniae. Understanding the regulatory mechanism of RstA in K. pneumoniae provides a proof-of-concept for identifying new genetic targets for controlling K. pneumoniae infection, which may aid in the development of therapeutic drugs.
Collapse
Affiliation(s)
- Xiaoyun Zhang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Clinical Laboratory, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Yaxuan Zhai
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jie Zhu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Zhichen Zhu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yicheng Wen
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Qizhao Gao
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Liang Wang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jiayao Lin
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yan Qian
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Liang Chen
- Department of Pharmacy Practice, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, New York, USA
| | - Hong Du
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
12
|
Oliva RL, Khadka UB, Camenzind T, Dyckmans J, Joergensen RG. Constituent of extracellular polymeric substances (EPS) produced by a range of soil bacteria and fungi. BMC Microbiol 2025; 25:298. [PMID: 40375143 DOI: 10.1186/s12866-025-04034-z] [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: 02/19/2025] [Accepted: 05/08/2025] [Indexed: 05/18/2025] Open
Abstract
Extracellular polymeric substances (EPS) produced by soil bacteria and fungi are crucial for microbial growth and provide many functions for the soil and its microbes. EPS composition may depend on microbial community composition and the soil physical and chemical environment, nevertheless, not much is known about the EPS constituents' specific roles nor how they interact to alter biofilm's functions. We hypothesized that EPS production would be enhanced with the presence of a surface and with a more labile carbon source. Also, that even though carbohydrates and proteins are the main constituents of EPS, we could still find quantifiable amounts of mannosamine and galactosamine (two amino sugars previously shown to be part of microbial biofilms). Ten soil bacterial and ten soil fungal species were cultured with glycerol or starch and with or without a quartz matrix. After a 4-day cultivation, EPS were extracted, and seven constituents were determined: carbohydrates, DNA, proteins, muramic acid, mannosamine, galactosamine, and glucosamine. We found EPS composition was strongly modified by microbial type, whereas differences in EPS production were driven mostly by environmental conditions. The EPS-carbohydrate/protein ratio was higher for cultures grown in starch media than in glycerol and increased in the presence of quartz. EPS-carbohydrate concentration reflected environmental changes of substrate quality and surface presence. Contrastingly, changes in the other EPS constituent composition are likely due to intrinsic microbial characteristics. Our findings open the pathway to study microbial biofilms in more complex environments (such as soils) and shed light to the importance of extracellular structures to microbial processes.
Collapse
Affiliation(s)
- Rebeca Leme Oliva
- Soil Biology and Plant Nutrition, University of Kassel, Nordbahnhofstr. 1a, Witzenhausen, 37213, Germany.
| | - Umesh B Khadka
- Soil Biology and Plant Nutrition, University of Kassel, Nordbahnhofstr. 1a, Witzenhausen, 37213, Germany
| | - Tessa Camenzind
- Institute of Biology, Freie Universität Berlin, Altensteinstr. 6, Berlin, 14195, Germany
| | - Jens Dyckmans
- Centre for Stable Isotope Research and Analysis, University of Göttingen, Büsgenweg 2, Göttingen, 37077, Germany
| | - Rainer Georg Joergensen
- Soil Biology and Plant Nutrition, University of Kassel, Nordbahnhofstr. 1a, Witzenhausen, 37213, Germany
| |
Collapse
|
13
|
Esin JJ, Visick KL, Kroken AR. Calcium signaling controls early stage biofilm formation and dispersal in Vibrio fischeri. J Bacteriol 2025:e0007725. [PMID: 40366159 DOI: 10.1128/jb.00077-25] [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: 03/10/2025] [Accepted: 04/17/2025] [Indexed: 05/15/2025] Open
Abstract
Bacterial dispersal from a biofilm is presently the least-studied step of the biofilm life cycle. The symbiotic bacterial species Vibrio fischeri is a model organism for studying biofilms relevant to a eukaryotic host; however, methodology is lacking to readily study the dispersal of this microbe from biofilms formed in the lab. Here, we adapted a time-lapse assay to visualize biofilm dispersal by V. fischeri. We observed biofilm formation and dispersal for multiple V. fischeri isolates, which displayed a variety of biofilm architecture phenotypes and dispersal dynamics. We then investigated V. fischeri strain ES114 using genetic tools and mutants available for this strain. ES114 exhibited calcium-dependent biofilm formation followed by a rapid (less than 10 min) coordinated dispersal event that occurred approximately 5 h from the experimental start. Biofilm dispersal was largely independent of the dispersal-promoting protease encoded by lapG. Although we found no role under our conditions for either biofilm formation or dispersal for several other factors including polysaccharides and autoinducers, we determined that biofilm formation was enhanced, and dispersal was delayed, with increased concentrations of calcium. Furthermore, biofilm formation depended on the calcium-responsive diguanylate cyclase (DGC) CasA, and dispersal could be modulated by overexpressing CasA. Our work has thus developed a new tool for the V. fischeri field and uncovered a key role for calcium signaling and c-di-GMP in early biofilm formation and dispersal in V. fischeri. IMPORTANCE Biofilm formation and dispersal are critical steps in both symbiotic and pathogenic colonization. Relative to biofilm formation, the process of dispersal in the model symbiont Vibrio fischeri, and other bacteria, is understudied. Here, we adapted an imaging assay to study early biofilm formation and the dispersal process in V. fischeri. We demonstrated that our assay can quantify biofilm formation and dispersal over time, can reveal phenotypic differences in diverse natural wild-type isolates, and is sensitive enough to investigate the impact of environmental factors. Our data confirm that calcium is a potent biofilm formation signal and identify the diguanylate cyclase CasA as a key regulator. This work leads the way for more in-depth research about unknown mechanisms of biofilm dispersal.
Collapse
Affiliation(s)
- Jeremy J Esin
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, Illinois, USA
| | - Karen L Visick
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, Illinois, USA
| | - Abby R Kroken
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, Illinois, USA
| |
Collapse
|
14
|
van Baalen C, Ketzetzi S, Tintor A, Gabay I, Isa L. Gating and tunable confinement of active colloids within patterned environments. SOFT MATTER 2025; 21:3850-3858. [PMID: 40261054 PMCID: PMC12013467 DOI: 10.1039/d4sm01512f] [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: 12/20/2024] [Accepted: 04/14/2025] [Indexed: 04/24/2025]
Abstract
Active colloidal particles typically exhibit a pronounced affinity for accumulating and being captured at boundaries. Here, we engineer long-range repulsive interactions between colloids that self-propel under an electric field and patterned obstacles. As a result of these interactions, particles turn away from obstacles and avoid accumulation. We show that by tuning the applied field frequency, we precisely and rapidly control the effective size of the obstacles and therefore modulate the particle approach distance. This feature allows us to achieve gating and tunable confinement of our active particles whereby they can access regions between obstacles depending on the applied field. Our work provides a versatile means to directly control confinement and organization, paving the way towards applications such as sorting particles based on motility or localizing active particles on demand.
Collapse
Affiliation(s)
- Carolina van Baalen
- Laboratory for Soft Materials and Interfaces, Department of Materials, ETH Zürich, Vladimir-Prelog-Weg 5, 8093 Zürich, Switzerland.
| | - Stefania Ketzetzi
- Laboratory for Soft Materials and Interfaces, Department of Materials, ETH Zürich, Vladimir-Prelog-Weg 5, 8093 Zürich, Switzerland.
| | - Anushka Tintor
- Laboratory for Soft Materials and Interfaces, Department of Materials, ETH Zürich, Vladimir-Prelog-Weg 5, 8093 Zürich, Switzerland.
| | - Israel Gabay
- Faculty of Mechanical Engineering, Technion - Israel Institute of Technology, Haifa 3200003, Israel.
| | - Lucio Isa
- Laboratory for Soft Materials and Interfaces, Department of Materials, ETH Zürich, Vladimir-Prelog-Weg 5, 8093 Zürich, Switzerland.
| |
Collapse
|
15
|
Panigrahi S, Konatam S, Tandi A, Roy DN. A comprehensive review of emerging 3D-printing materials against bacterial biofilm growth on the surface of healthcare settings. Biomed Mater 2025; 20:032007. [PMID: 40306307 DOI: 10.1088/1748-605x/add2bb] [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: 01/19/2025] [Accepted: 04/30/2025] [Indexed: 05/02/2025]
Abstract
A significant burden on the healthcare system, microbial contamination of biomedical surfaces can result in hospital-acquired illnesses. Bacteria, viruses, and fungi may live on surfaces for days or months and spread to patients and medical personnel. This article describes the 3D printing technologies, such as fused deposition modeling, bioprinting, binder jetting/inkjet, poly-jet, electron beam manufacturing, stereolithography, selective laser sintering, and laminated object manufacturing used for manufacturing the healthcare setting's surface to reduce bacterial contamination with exploring anti-biofilm activity against different bacterial species responsible for infections, based on the critical evaluation of published reports. This strategy has immense potential to become an upcoming approach for advancing the coating concept on the material's surface in healthcare settings. Our literature evaluation identifies beneficial 3D printing materials and associated technologies against microorganisms' growth, mainly bacteria involved in implant-based infection, emphasizing the development of anti-biofilm 3D-printed surfaces. Additionally, the authors have identified a few key areas where research and development are critically required to advance 3D-printing technology in healthcare settings.
Collapse
Affiliation(s)
- Shristi Panigrahi
- Department of Biotechnology, National Institute of Technology-Raipur, Raipur, Chhattisgarh, India
| | - Shraavani Konatam
- Department of Biotechnology, National Institute of Technology-Raipur, Raipur, Chhattisgarh, India
| | - Antara Tandi
- Department of Biotechnology, National Institute of Technology-Raipur, Raipur, Chhattisgarh, India
| | - Dijendra Nath Roy
- Department of Biotechnology, National Institute of Technology-Raipur, Raipur, Chhattisgarh, India
| |
Collapse
|
16
|
Lan J, Zou J, Xin H, Sun J, Han T, Sun M, Niu M. Nanomedicines as disruptors or inhibitors of biofilms: Opportunities in addressing antimicrobial resistance. J Control Release 2025; 381:113589. [PMID: 40032007 DOI: 10.1016/j.jconrel.2025.113589] [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: 12/18/2024] [Revised: 02/02/2025] [Accepted: 02/26/2025] [Indexed: 03/05/2025]
Abstract
The problem of antimicrobial resistance (AMR) has caused global concern due to its great threat to human health. Evidences are emerging for a critical role of biofilms, one of the natural protective mechanisms developed by bacteria during growth, in resisting commonly used clinical antibiotics. Advances in nanomedicines with tunable physicochemical properties and unique anti-biofilm mechanisms provide opportunities for solving AMR risks more effectively. In this review, we summarize the five "A" stages (adhesion, amplification, alienation, aging and allocation) of biofilm formation and mechanisms through which they protect the internal bacteria. Aimed at the characteristics of biofilms, we emphasize the design "THAT" principles (targeting, hacking, adhering and transport) of nanomedicines in their interactions with biofilms and internal bacteria. Furthermore, recent progresses in multimodal antibacterial nanomedicines, including biofilms disruption and bactericidal activity, and the types of currently available antibiofilm nanomedicines contained organic and inorganic nanomedicines are outlined and highlighted their potential applications in the development of preclinical research. Last but not least, we offer a perspective for the effectiveness of nanomedicines designed to address AMR and challenges associated with their clinical translation.
Collapse
Affiliation(s)
- Jiaming Lan
- Department of Interventional Radiology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, China
| | - Jingyu Zou
- Department of Neurosurgery, The First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning, China
| | - He Xin
- Department of Interventional Radiology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, China
| | - Jin Sun
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, China
| | - Tao Han
- Department of Oncology, The First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning, China.
| | - Mengchi Sun
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, China; School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, China.
| | - Meng Niu
- Department of Interventional Radiology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, China.
| |
Collapse
|
17
|
Li C, Hou J, Kong M, Yao Y, Adyel TM, Wu J, You G, Yu Y, Liu S, Yang Z, Miao L. Increasing drying changes the relationship between biodiversity and ecosystem multifunctionality. NPJ Biofilms Microbiomes 2025; 11:72. [PMID: 40328787 PMCID: PMC12056148 DOI: 10.1038/s41522-025-00711-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 04/22/2025] [Indexed: 05/08/2025] Open
Abstract
Increased drying of rivers under global climate change is leading to biodiversity loss. However, it is not clear whether biodiversity loss affects river functions. In this study, we investigated the changes in biofilm community diversity and functions in an artificial stream after different drying durations. A critical drying duration of around 60 days was found in the microbial composition and functions. Therefore, different drying durations can be divided into short-term drying (~0-20 days) and long-term drying (~60-130 days) to analyse the effect of biodiversity in terms of ecosystem functions. In summary, the dominant relationship of biodiversity on community stability got uncoupled after long-term drying. Community assembly became dominant in maintaining multifunctionality with increasing drying duration rather than biodiversity as traditionally perceived. This study reveals the importance of community assembly, extending theoretical knowledge of the relationship between biodiversity and ecosystem multifunctionality.
Collapse
Affiliation(s)
- Chaoran Li
- Key Laboratory of Integrated Regulation and Resources Development on Shallow Lakes, Ministry of Education, College of Environment, Hohai University, Nanjing, 210098, People's Republic of China
| | - Jun Hou
- Key Laboratory of Integrated Regulation and Resources Development on Shallow Lakes, Ministry of Education, College of Environment, Hohai University, Nanjing, 210098, People's Republic of China
| | - Ming Kong
- Nanjing Institute of Environmental Sciences, Ministry of Ecology and Environment, Nanjing, 210042, People's Republic of China
| | - Yu Yao
- School of Environment, Nanjing Normal University, Nanjing, 210023, China
| | - Tanveer M Adyel
- Centre for Nature Positive Solutions, School of Science, RMIT University, Melbourne, VIC, 3000, Australia
| | - Jun Wu
- Key Laboratory of Integrated Regulation and Resources Development on Shallow Lakes, Ministry of Education, College of Environment, Hohai University, Nanjing, 210098, People's Republic of China
| | - Guoxiang You
- Key Laboratory of Integrated Regulation and Resources Development on Shallow Lakes, Ministry of Education, College of Environment, Hohai University, Nanjing, 210098, People's Republic of China
| | - Yue Yu
- Department of Civil, Environmental, and Geomatic Engineering, ETH Zürich, Zürich, 8092, Switzerland
| | - Songqi Liu
- School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou, 215009, People's Republic of China
| | - Zijun Yang
- Key Laboratory of Integrated Regulation and Resources Development on Shallow Lakes, Ministry of Education, College of Environment, Hohai University, Nanjing, 210098, People's Republic of China
| | - Lingzhan Miao
- Key Laboratory of Integrated Regulation and Resources Development on Shallow Lakes, Ministry of Education, College of Environment, Hohai University, Nanjing, 210098, People's Republic of China.
| |
Collapse
|
18
|
Verma N, Bajiya M, Dolhey R, Surabhi, Yadav AS, Chaudhary C, Meena D, Arya H, Bhatt TK, Yadav JK, Shukla JN, Swaroop S, Pandey J. Mechanistic Insights into the Antibiofilm Activity of Simvastatin and Lovastatin against Bacillus subtilis. Mol Pharm 2025; 22:2703-2722. [PMID: 40100146 DOI: 10.1021/acs.molpharmaceut.5c00191] [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: 03/20/2025]
Abstract
Statins have been reported for diverse pleiotropic activities, including antimicrobial and antibiofilm. However, due to the limited understanding of their mode of action, none of the statins have gained approval for antimicrobial or antibiofilm applications. In a recent drug repurposing study, we observed that two statins (i.e., Simvastatin and Lovastatin) interact stably with TasA(28-261), the principal extracellular matrix protein of Bacillus subtilis, and also induce inhibition of biofilm formation. Nevertheless, the underlying mechanism remained elusive. In the present study, we examined the impact of these statins on the physiological activity of TasA(28-261), specifically its interaction with TapA(33-253) and aggregation into the amyloid-like structure using purified recombinant TasA(28-261) and TapA(33-253) in amyloid detection-specific in vitro assays (i.e., CR binding and ThT staining assays). Results revealed that both statins interfered with amyloid formation by the TasA(28-261)-TapA(33-253) complex, while neither statin inhibited amyloid formation by lysozyme, a model amyloid-forming protein. Moreover, neither statin significantly altered the expressions of terminal regulatory genes (viz, sinR, sinI) and terminal effector genes (viz, tasA, tapA, and bslA) involved in biofilm formation by B. subtilis. While the intricate interplay between Simvastatin and Lovastatin with the diverse molecular constituents of B. subtilis biofilm remains to be elucidated conclusively, the findings obtained during the present study suggest that the underlying mechanism for Simvastatin- and Lovastatin-mediated inhibition of B. subtilis biofilm formation is manifested by interfering with the aggregation and amyloid formation by TasA(28-261)-TapA(33-253). These results represent one of the first experimental evidence for the underlying mechanism of antibiofilm activity of statins and offer valuable directions for future research to harness statins as antibiofilm therapeutics.
Collapse
Affiliation(s)
- Nidhi Verma
- Department of Biotechnology, Central University of Rajasthan, Kishangarh, Ajmer 305801, Rajasthan, India
| | - Mamta Bajiya
- Department of Biotechnology, Central University of Rajasthan, Kishangarh, Ajmer 305801, Rajasthan, India
| | - Ragini Dolhey
- Department of Biotechnology, Central University of Rajasthan, Kishangarh, Ajmer 305801, Rajasthan, India
| | - Surabhi
- Department of Biotechnology, Central University of Rajasthan, Kishangarh, Ajmer 305801, Rajasthan, India
| | - Abhishek Singh Yadav
- Department of Biotechnology, Central University of Rajasthan, Kishangarh, Ajmer 305801, Rajasthan, India
| | - Chhavi Chaudhary
- Department of Biotechnology, Central University of Rajasthan, Kishangarh, Ajmer 305801, Rajasthan, India
| | - Dhankesh Meena
- Department of Biochemistry, Central University of Rajasthan, Kishangarh, Ajmer 305801, Rajasthan, India
| | - Hemant Arya
- Department of Biotechnology, Central University of Rajasthan, Kishangarh, Ajmer 305801, Rajasthan, India
| | - Tarun K Bhatt
- Department of Biotechnology, Central University of Rajasthan, Kishangarh, Ajmer 305801, Rajasthan, India
| | - Jay Kant Yadav
- Department of Biotechnology, Central University of Rajasthan, Kishangarh, Ajmer 305801, Rajasthan, India
| | - Jayendra Nath Shukla
- Department of Biotechnology, Central University of Rajasthan, Kishangarh, Ajmer 305801, Rajasthan, India
| | - Shiv Swaroop
- Department of Biochemistry, Central University of Rajasthan, Kishangarh, Ajmer 305801, Rajasthan, India
| | - Janmejay Pandey
- Department of Biotechnology, Central University of Rajasthan, Kishangarh, Ajmer 305801, Rajasthan, India
| |
Collapse
|
19
|
Cota-Gastélum LA, Reyes-López MÁ, Escamilla-Montes R, Luna-González A, Calderón-Vázquez CL, Diarte-Plata G. In vitro controlled release of the probiotic strain Bacillus licheniformis PPL2016 microencapsulated: Simulating the digestive system by age class and sex in the blue swimming crab Callinectes arcuatus. Braz J Microbiol 2025:10.1007/s42770-025-01674-1. [PMID: 40319424 DOI: 10.1007/s42770-025-01674-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 04/11/2025] [Indexed: 05/07/2025] Open
Abstract
This work aims to encapsulate Bacillus licheniformis PPL2016 (12 × 106 CFU/mL), a marine probiotic characterized at a biochemical and molecular level, in sodium alginate (2%) microparticles and to evaluate its controlled and directed release in a simulated digestive system (DS) of the swimming crab Callinectes arcuatus, considering the following age classes and sexes: Adult Female, Juvenile Female, Adult Male, and Juvenile. The encapsulation process was carried out using the ionic gelation technique. The microcapsules were characterized physiochemically by their size, morphology, number of encapsulated bacteria after the encapsulation process, as well as bacterial survival after 45 days of storage (4 °C). The in vitro release and survival studies of bacteria inside the organs that make up the DS of C. arcuatus were carried out using a protocol developed in our laboratory by applying extracts of dissected organs from the DS (stomach, hepatopancreas and intestine) of the swimming crab. A χ2 test (α = 0.05) was performed at linearization (Log10) of the percentages of the controlled releases of microencapsulated B. licheniformis PPL2016 at different times (0 h, 4 h, 8 h, 12 h), corresponding to the extracts of the organs which simulated the digestive system of C. arcuatus. After biochemical characterization B. licheniformis PPL2016 was considered probiotic bacteria. Microparticles with an average size of 602 to 639 µm were obtained after using the ionic gelation method. Bacterial survival and encapsulation efficacy showed high cell viability and performance above 77.94%. Stability studies showed that storage at a temperature of 4 °C, kept almost 100% of viable bacteria for 15 days; however, cell viability decreased to a survival of 90% after 30 days of storage at this temperature. Regardless of reduced cell viability after 30 days, there are enough viable bacterial cells. Release and survival studies showed that alginate particles had a protective effect on bacteria, these results suggest that microparticles can be produced by a low-cost method. In juvenile males, the percentage of release of probiotic bacteria was greater in TIV in the enzyme extract of the intestine (12 h) with 95 ± 0.45%. Juvenile males had the lowest in vitro release at the stomach stage (0 h) and thus marks the significance for their low release of microcapsules at the beginning of the in vitro release (χ2 = 6.7509; χ2Calculated Pool = 13.5188; χ2Calculated Critical (0.05, 21) = 11.5919; p < 0.05), with the highest significance in the intestine (12 h) (χ2 = 1.2602; χ2Calculated Pool = 13.5188; χ2Calculated Critical (0.05, 21) = 11.5919; p < 0.05). Significant differences in vitro bacterial release were recorded for age classes and sexes of C. arcuatus.
Collapse
Affiliation(s)
- Luis Abraham Cota-Gastélum
- Aquaculture Department, Instituto Politécnico Nacional, Centro Interdisciplinario de Investigación Para El Desarrollo Integral Regional-Sinaloa Unit, Blvd Juan de Dios Bátiz Paredes 250, Col. San Joachín, 81101, Guasave, Sinaloa, Mexico
| | - Miguel Ángel Reyes-López
- Centro de Biotecnología Genómica. Laboratory of Conservation Medicine, Instituto Politécnico Nacional, Blvd. Del Maestro SN, Narciso Mendoza, 88710, Reynosa, Tamaulipas, Mexico
| | - Ruth Escamilla-Montes
- Aquaculture Department, Instituto Politécnico Nacional, Centro Interdisciplinario de Investigación Para El Desarrollo Integral Regional-Sinaloa Unit, Blvd Juan de Dios Bátiz Paredes 250, Col. San Joachín, 81101, Guasave, Sinaloa, Mexico
| | - Antonio Luna-González
- Aquaculture Department, Instituto Politécnico Nacional, Centro Interdisciplinario de Investigación Para El Desarrollo Integral Regional-Sinaloa Unit, Blvd Juan de Dios Bátiz Paredes 250, Col. San Joachín, 81101, Guasave, Sinaloa, Mexico
| | - Carlos Ligne Calderón-Vázquez
- Aquaculture Department, Instituto Politécnico Nacional, Centro Interdisciplinario de Investigación Para El Desarrollo Integral Regional-Sinaloa Unit, Blvd Juan de Dios Bátiz Paredes 250, Col. San Joachín, 81101, Guasave, Sinaloa, Mexico
| | - Genaro Diarte-Plata
- Aquaculture Department, Instituto Politécnico Nacional, Centro Interdisciplinario de Investigación Para El Desarrollo Integral Regional-Sinaloa Unit, Blvd Juan de Dios Bátiz Paredes 250, Col. San Joachín, 81101, Guasave, Sinaloa, Mexico.
| |
Collapse
|
20
|
Zhang X, Feng X, Ma L, Lei J, Li G, Zhang W, Liang H, Tong B, Wu D, Yang C, Tan L. A sonosensitive diphenylalanine-based broad-spectrum antimicrobial peptide. Nat Biomed Eng 2025:10.1038/s41551-025-01377-w. [PMID: 40316686 DOI: 10.1038/s41551-025-01377-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 03/14/2025] [Indexed: 05/04/2025]
Abstract
The antimicrobial effect of antimicrobial peptides is typically slow; they can be rapidly biodegraded and often have non-selective toxicity and elaborate sequences. Here we report a short peptide that is activated by ultrasound, that shows high broad-spectrum antibacterial efficiency (>99%) against clinically isolated methicillin-resistant bacteria (specifically, Staphylococcus aureus, Escherichia coli, Staphylococcus epidermidis, Enterobacter cancerogenus and Pseudomonas aeruginosa) with 15 min of ultrasound irradiation, and that has negligible toxicity and low self-antibacterial activity. We selected the peptide, FFRKSKEK (a segment from the human host-defence LL-37 peptide), from a library of peptides with piezoelectric diphenylalanine (FF) sequences, low toxicity, hydrophobicity and net positive charge. We show via all-atom molecular dynamics simulations that ultrasound amplifies the membrane-penetrating ability of peptides with FF sequences and that its piezoelectric polarization generates reactive-oxygen species and disturbs bacterial electron-transport chains. In a goat model of hard-to-treat intervertebral infection, the sonosensitive peptide led to better outcomes than vancomycin. Antimicrobial peptides activated by ultrasound may offer a clinically relevant strategy for combating antibiotic-resistant infections.
Collapse
Affiliation(s)
- Xiaoguang Zhang
- Department of Orthopaedics, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaobo Feng
- Department of Orthopaedics, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liang Ma
- Department of Orthopaedics, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jie Lei
- Department of Orthopaedics, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gaocai Li
- Department of Orthopaedics, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weifeng Zhang
- Department of Orthopaedics, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huaizhen Liang
- Department of Orthopaedics, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bide Tong
- Department of Orthopaedics, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Di Wu
- Department of Orthopaedics, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cao Yang
- Department of Orthopaedics, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Lei Tan
- Department of Orthopaedics, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
21
|
Azevedo A, Teixeira-Santos R, Gomes LC, Duarte SOD, Monteiro GA, Mergulhão FJ. Engineering Escherichia coli Biofilms for Curcumin Production. Molecules 2025; 30:2031. [PMID: 40363836 PMCID: PMC12073880 DOI: 10.3390/molecules30092031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 03/21/2025] [Accepted: 04/28/2025] [Indexed: 05/15/2025] Open
Abstract
Biofilms are emerging platforms for the production of valuable compounds. The present study is the first to assess the capacity of Escherichia coli biofilms to produce curcumin through the expression of a biosynthetic pathway involving three genes: 4-coumarate-CoA ligase (4CL), diketide-CoA synthase (DCS), and curcumin synthase (CURS). The effects of chemical induction with isopropyl β-d-1-thiogalactopyranoside (IPTG) and ferulic acid (FA), and the incubation temperature on biofilm formation and curcumin production were evaluated. Biofilms were formed in 12-well microtiter plates over three days and then induced with 1 mM IPTG and FA at 2 or 8 mM. After induction, the samples were incubated for two days at 26 or 30 °C. Total and culturable planktonic and biofilm cells, as well as biofilm thickness and volumetric and specific curcumin production, were assessed on days 3, 4, and 5. The results demonstrated that biofilms produced up to 10-fold higher curcumin levels (0.9-2.2 fg·cell-1) than their planktonic counterparts (0.1-0.3 fg·cell-1). The highest specific curcumin production (2.2 fg·cell-1) was achieved using 8 mM FA. However, no significant differences in curcumin production were observed between the induced samples incubated at the tested temperatures. These results validated the potential of biofilm systems for expressing a complete exogenous biosynthetic pathway using metabolic engineering, particularly for curcumin production.
Collapse
Affiliation(s)
- Ana Azevedo
- LEPABE—Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal; (A.A.); (R.T.-S.); (L.C.G.)
- ALICE—Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Rita Teixeira-Santos
- LEPABE—Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal; (A.A.); (R.T.-S.); (L.C.G.)
- ALICE—Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Luciana C. Gomes
- LEPABE—Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal; (A.A.); (R.T.-S.); (L.C.G.)
- ALICE—Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Sofia O. D. Duarte
- iBB—Institute for Bioengineering and Biosciences, Department of Bioengineering, Instituto Superior Técnico, University of Lisbon, 1049-001 Lisboa, Portugal; (S.O.D.D.); (G.A.M.)
- Associate Laboratory i4HB—Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Gabriel A. Monteiro
- iBB—Institute for Bioengineering and Biosciences, Department of Bioengineering, Instituto Superior Técnico, University of Lisbon, 1049-001 Lisboa, Portugal; (S.O.D.D.); (G.A.M.)
- Associate Laboratory i4HB—Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Filipe J. Mergulhão
- LEPABE—Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal; (A.A.); (R.T.-S.); (L.C.G.)
- ALICE—Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| |
Collapse
|
22
|
Yaseen Z, Aslam S, Rahmat Ullah S, Rahman A, Khan Niazi MB, Andleeb S. Nature-inspired designing of KLR- and KLS-rich antimicrobial peptides: unleashing the antibiofilm potential of RbP12 against MDR S. aureus and P. aeruginosa. J Antimicrob Chemother 2025; 80:1331-1341. [PMID: 40126541 DOI: 10.1093/jac/dkaf079] [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: 07/22/2024] [Accepted: 02/17/2025] [Indexed: 03/25/2025] Open
Abstract
OBJECTIVES Biofilm formation is a mechanism exhibited by bacteria, making them 10-1000 times more resistant than planktonic cells. The aim was to collect the most suitable characteristics from already available antibiofilm peptides and design novel antibiofilm peptide sequences along with these characteristics altogether in one sequence. METHODS Antibiofilm peptides were collected from AMP database (APD3), and sequence analysis was performed to derive the most suitable features. An artificial design approach, modified database filtering technology, was chosen to design novel peptide sequences, and their activity was predicted by machine-learning prediction models. Antibacterial and antibiofilm potential of the selected peptide sequence (arginine-based peptide 12; RbP12) was assessed against Staphylococcus aureus P10 and Pseudomonas aeruginosa PA64. RESULTS A total of 34 peptides were designed, of which 22 were arginine based and 12 were serine based. All the designed peptides were predicted to have antibiofilm properties. RbP12 was found to inhibit the growth of S. aureus P10 completely at an MIC of 85 mg/L, while the percentage inhibition of P. aeruginosa PA64 was calculated to be 32.1%. Significant inhibition of biofilms by RbP12 was observed in the case of both S. aureus P10 and P. aeruginosa PA64. An MTT assay showed no significant cytotoxicity by RbP12 with 96% cell viability. CONCLUSIONS RbP12 was found to have higher antibacterial and antibiofilm activity against S. aureus P10 compared with P. aeruginosa PA64. With 96% cell viability, usage of RbP12 on human skin is totally safe. Based on these results, the aim is to develop self-assembled peptide hydrogels for wound healing in future work.
Collapse
Affiliation(s)
- Zeeshan Yaseen
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12, Islamabad 44000, Pakistan
| | - Saiqa Aslam
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12, Islamabad 44000, Pakistan
| | - Sidra Rahmat Ullah
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12, Islamabad 44000, Pakistan
| | - Abdur Rahman
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12, Islamabad 44000, Pakistan
| | - Muhammad Bilal Khan Niazi
- Department of Chemical Engineering, King Fahd University of Petroleum and Minerals (KFUPM), Dhahran 31261, Saudi Arabia
| | - Saadia Andleeb
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12, Islamabad 44000, Pakistan
| |
Collapse
|
23
|
Shah T, Zhu C, Shah C, Upadhyaya I, Upadhyay A. Trans-cinnamaldehyde nanoemulsion reduces Salmonella Enteritidis biofilm on steel and plastic surfaces and downregulates expression of biofilm associated genes. Poult Sci 2025; 104:105086. [PMID: 40168703 PMCID: PMC11997393 DOI: 10.1016/j.psj.2025.105086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 04/03/2025] Open
Abstract
Salmonella Enteritidis is a major poultry-associated foodborne pathogen that can form sanitizer-tolerant biofilms on various surfaces. The biofilm-forming capability of S. Enteritidis facilitates its survival on farm and food processing equipment. Conventional sanitization methods are not completely effective in killing S. Enteritidis biofilms. This study investigated the efficacy of a Generally Recognized as Safe phytochemical Trans-cinnamaldehyde (TC), and in its nanoemulsion form (TCNE), for inhibiting S. Enteritidis biofilm formation and inactivating mature biofilms developed on polystyrene and stainless-steel surfaces. Moreover, the effect of TC on Salmonella genes critical for biofilm formation was studied. TCNE was prepared using a high energy sonication method with Tween 80. For biofilm inhibition assay, S. Enteritidis was allowed to form biofilms either in the presence or absence of sub-inhibitory concentration (SIC; 0.01 %) of TCNE at 25°C and the biofilm formed was quantified at 24-h intervals for 48 h. For the inactivation assay, S. Enteritidis biofilms developed at 25°C for 48 h were exposed to TCNE (0.5, 1 %) for 1, 5, and 15 min, and surviving S. Enteritidis in the biofilm were enumerated. SIC of TCNE inhibited S. Enteritidis biofilm by 45 % on polystyrene and 75 % on steel surface after 48 h at 25°C compared to control (P < 0.05). All TCNE treatments rapidly inactivated S. Enteritidis mature biofilm on polystyrene and steel surfaces (P < 0.05). The lower concentration of TCNE (0.5 %) reduced S. Enteritidis counts by 1.5 log CFU/ml as early as 1 min of exposure on both polystyrene and stainless-steel surfaces. After 15 min of exposure, TCNE at concentration of 0.5 or 1 % reduced S. Enteritidis count significantly by 4.5 log CFU or 6 log CFU/ml on polystyrene or stainless-steel surfaces. TC downregulated the expression of S. Enteritidis genes (hilA, hilC, flhD, csgA, csgD, sdiA) responsible for biofilm formation (P < 0.05). Results suggest that TCNE has potential as a natural disinfectant for controlling S. Enteritidis biofilms on common farm and food processing surfaces, such as plastic and steel.
Collapse
Affiliation(s)
- Trushenkumar Shah
- Department of Animal Science, University of Connecticut, Storrs, Connecticut, USA
| | - Chen Zhu
- Department of Animal Science, University of Connecticut, Storrs, Connecticut, USA
| | - Chetna Shah
- Department of Animal Science, University of Connecticut, Storrs, Connecticut, USA
| | - Indu Upadhyaya
- Department of Extension, University of Connecticut, Storrs, Connecticut, USA
| | - Abhinav Upadhyay
- Department of Animal Science, University of Connecticut, Storrs, Connecticut, USA.
| |
Collapse
|
24
|
Aboudalle A, Barthomeuf M, Castel X, Le Gendre L, Pissavin C. Antibacterial activity of photocatalytic titanium dioxide (TiO 2) thin films for Listeria monocytogenes biofilms disinfection. Photochem Photobiol 2025. [PMID: 40275714 DOI: 10.1111/php.14105] [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: 02/12/2025] [Revised: 04/04/2025] [Accepted: 04/08/2025] [Indexed: 04/26/2025]
Abstract
The presence of microbial biofilms on equipment surfaces is a recurrent problem in the food industry. To reduce the risk of biofilm development, a preventive method based on photoactive antibacterial surfaces is proposed. In the present study, crystalline rutile form titanium dioxide (TiO2) thin layers are deposited on stainless steel substrates by RF sputtering under reactive plasma. Such layers are assessed for their bactericidal activity on two strains of Listeria monocytogenes. After 1 h of irradiation under UV-A at 365 nm, a decrease of 2 log of the number of adherent Listeria cells is observed. Analysis with scanning electron microscopy suggests damages to the bacterial walls. Moreover, the peroxidation of the membrane lipids of L. monocytogenes by the radical species formed by photocatalysis is confirmed since malondialdehyde was detected after irradiation. Furthermore, the present work investigates the role of the redox species generated by photocatalysis. Indeed, experiments carried out in the presence of scavenger molecules (DMSO, EDTA-2Na, superoxide dismutase) show that holes are the main redox species involved in the antibacterial activity of the deposited layers. These results allow a better understanding of the role of the redox species generated by the photocatalytic activity of the rutile TiO2 thin layers.
Collapse
Affiliation(s)
- Arwa Aboudalle
- Biological Engineering Department, Univ Rennes, IUT Saint-Brieuc, Saint-Brieuc, France
| | - Marion Barthomeuf
- Biological Engineering Department, Univ Rennes, IUT Saint-Brieuc, Saint-Brieuc, France
| | - Xavier Castel
- Univ Rennes, CNRS, IETR - UMR 6164, Saint-Brieuc, France
| | | | - Christine Pissavin
- Biological Engineering Department, Univ Rennes, IUT Saint-Brieuc, Saint-Brieuc, France
| |
Collapse
|
25
|
Bai R, Yang L, Jia M, Chen R, Zhang H, Pan Y, He P, Miao X, Fan Q, Hu W. Cascade Production of In Situ Oxygen and Singlet Oxygen from Self-Assembled Nanophotosensitizer for Anti-Hypoxic Photodynamic Therapy. NANO LETTERS 2025. [PMID: 40249842 DOI: 10.1021/acs.nanolett.5c01324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/20/2025]
Abstract
Photosensitizers (PSs) capable of in situ oxygen (O2) production are attractive for overcoming hypoxia in photodynamic therapy (PDT). However, these PSs generally require multiple components and complex fabrication procedures, preventing their clinical translation. Herein, we develop a single-component nanophotosensitizer via simple self-assembly that enables cascade production of in situ O2 and singlet oxygen (1O2) for superior antibacterial PDT (aPDT). Perylene tetracarboxylic acid (PTA) molecules self-assemble into nanophotosensitizers (PTA NPs). Mechanism studies reveal dual functionality of PTA NPs due to their antiparallel-displaced π-π stacking. Aggregated PTA molecules undergo intermolecular electron transfer to yield substantial photogenerated holes, while unimolecular PTA undergoes intersystem crossing to produce triplet PS (3PS*). These holes effectively oxidize water into O2 in situ, which then participates in downstream photosensitization with 3PS* to yield 1O2. This cascade reaction affords PTA NPs with continuous O2 supply and efficient 1O2 production, enabling a 63.07% higher antibacterial rate compared with the clinical antibiotic vancomycin.
Collapse
Affiliation(s)
- Ruida Bai
- State Key Laboratory of Flexible Electronics (LoFE) and Institute of Flexible Electronics (IFE), Northwestern Polytechnical University, Xi'an 710072, P. R. China
| | - Linfang Yang
- State Key Laboratory of Flexible Electronics (LoFE) and Institute of Flexible Electronics (IFE), Northwestern Polytechnical University, Xi'an 710072, P. R. China
| | - Mingxuan Jia
- State Key Laboratory of Flexible Electronics (LoFE) and Institute of Flexible Electronics (IFE), Northwestern Polytechnical University, Xi'an 710072, P. R. China
| | - Ruizhe Chen
- State Key Laboratory of Flexible Electronics (LoFE) and Institute of Flexible Electronics (IFE), Northwestern Polytechnical University, Xi'an 710072, P. R. China
| | - Haolin Zhang
- State Key Laboratory of Flexible Electronics (LoFE) and Institute of Flexible Electronics (IFE), Northwestern Polytechnical University, Xi'an 710072, P. R. China
| | - Yonghui Pan
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing 210023, P. R. China
| | - Ping He
- State Key Laboratory of Flexible Electronics (LoFE) and Institute of Flexible Electronics (IFE), Northwestern Polytechnical University, Xi'an 710072, P. R. China
| | - Xiaofei Miao
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing 210023, P. R. China
| | - Quli Fan
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing 210023, P. R. China
| | - Wenbo Hu
- State Key Laboratory of Flexible Electronics (LoFE) and Institute of Flexible Electronics (IFE), Northwestern Polytechnical University, Xi'an 710072, P. R. China
| |
Collapse
|
26
|
Matavacas J, von Wachenfeldt C. Protein Homeostasis Impairment Alters Phenotypic Heterogeneity of Biofilm Communities. Mol Microbiol 2025. [PMID: 40243034 DOI: 10.1111/mmi.15366] [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: 12/17/2024] [Revised: 03/24/2025] [Accepted: 03/28/2025] [Indexed: 04/18/2025]
Abstract
Biofilms are highly organized, cooperating communities of microorganisms encased in a self-produced extracellular matrix, providing resilience against external stress such as antimicrobial agents and host defenses. A hallmark of biofilms is their phenotypic heterogeneity, which enhances the overall growth and survival of the community. In this study, we demonstrate that removing the dnaK and tig genes encoding the core molecular chaperones DnaK (Hsp70 homolog) and Trigger factor disrupted protein homeostasis in Bacillus subtilis and resulted in the formation of an extremely mucoid biofilm with aberrant architecture, compromised structural integrity, and altered phenotypic heterogeneity. These changes include a large reduction in the motile subpopulation and an overrepresentation of matrix producers and endospores. Overproduction of poly-γ-glutamic acid contributed crucially to the mucoid phenotype and aberrant biofilm architecture. Homeostasis impairment, triggered by elevated temperatures, in wild-type cells led to mucoid and aberrant biofilm phenotypes similar to those observed in strains lacking both dnaK and tig. Our findings show that disruption of protein homeostasis, whether due to the absence of molecular chaperones or because of environmental factors, severely changes biofilm features.
Collapse
Affiliation(s)
- Judith Matavacas
- The Microbiology Group, Department of Biology, Lund University, Lund, Sweden
| | | |
Collapse
|
27
|
Blöcher L, Schwabe J, Glatter T, Søgaard-Andersen L. Identification of EcpK, a bacterial tyrosine pseudokinase important for exopolysaccharide biosynthesis in Myxococcus xanthus. J Bacteriol 2025; 207:e0049924. [PMID: 40067014 PMCID: PMC12004946 DOI: 10.1128/jb.00499-24] [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: 11/26/2024] [Accepted: 02/16/2025] [Indexed: 04/18/2025] Open
Abstract
Bacteria synthesize chemically diverse capsular and secreted polysaccharides that function in many physiological processes and are widely used in industrial applications. In the ubiquitous Wzx/Wzy-dependent biosynthetic pathways for these polysaccharides, the polysaccharide co-polymerase (PCP) facilitates the polymerization of repeat units in the periplasm, and in Gram-negative bacteria, also polysaccharide translocation across the outer membrane. These PCPs belong to the PCP-2 family, are integral inner membrane proteins with extended periplasmic domains, and functionally depend on alternating between different oligomeric states. The oligomeric state is determined by a cognate cytoplasmic bacterial tyrosine kinase (BYK), which is either part of the PCP or a stand-alone protein. Interestingly, BYK-like proteins, which lack key catalytic residues and/or the phosphorylated Tyr residues, have been described. In Myxococcus xanthus, the exopolysaccharide (EPS) is synthesized and exported via the Wzx/Wzy-dependent EPS pathway in which EpsV serves as the PCP. Here, we confirm that EpsV lacks the BYK domain. Using phylogenomics, experiments, and computational structural biology, we identify EcpK as important for EPS biosynthesis and show that it structurally resembles canonical BYKs but lacks residues important for catalysis and Tyr phosphorylation. Using proteomic analyses, two-hybrid assays, and structural modeling, we demonstrate that EcpK directly interacts with EpsV. Based on these findings, we suggest that EcpK is a BY pseudokinase and functions as a scaffold, which by direct protein-protein interactions, rather than by Tyr phosphorylation, facilitates EpsV function. EcpK and EpsV homologs are present in other bacteria, suggesting broad conservation of this mechanism and establishing a phosphorylation-independent PCP-2 subfamily.IMPORTANCEBacteria produce a variety of polysaccharides with important biological functions. In Wzx/Wzy-dependent pathways for the biosynthesis of secreted and capsular polysaccharides in Gram-negative bacteria, the polysaccharide co-polymerase (PCP) is a key protein that facilitates repeat unit polymerization and polysaccharide translocation across the outer membrane. PCP function depends on assembly/disassembly cycles that are determined by the phosphorylation/dephosphorylation cycles of an associated bacterial tyrosine kinase (BYK). Here, we identify the BY pseudokinase EcpK as essential for exopolysaccharide biosynthesis in Myxococcus xanthus. Based on experiments and computational structural biology, we suggest that EcpK is a scaffold protein, guiding the assembly/disassembly cycles of the partner PCP via binding/unbinding cycles independently of Tyr phosphorylation/dephosphorylation cycles. We suggest that this novel mechanism is broadly conserved.
Collapse
Affiliation(s)
- Luca Blöcher
- Department of Ecophysiology, Max Planck Institute for Terrestrial Microbiology, Marburg, Germany
| | - Johannes Schwabe
- Department of Ecophysiology, Max Planck Institute for Terrestrial Microbiology, Marburg, Germany
| | - Timo Glatter
- Core Facility for Mass Spectrometry and Proteomics Max Planck Institute for Terrestrial Microbiology, Marburg, Germany
| | - Lotte Søgaard-Andersen
- Department of Ecophysiology, Max Planck Institute for Terrestrial Microbiology, Marburg, Germany
| |
Collapse
|
28
|
Hao Z, Zhou H, Gao D, Qiu L, Xing C. Rational Design of Quinoidal Conjugated Polymers for Photothermal Antibacterial Therapy. Macromol Rapid Commun 2025; 46:e2401031. [PMID: 39838606 DOI: 10.1002/marc.202401031] [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: 12/15/2024] [Revised: 01/12/2025] [Indexed: 01/23/2025]
Abstract
The increasing prevalence of antibiotic resistance, driven by the overuse and misuse of conventional antibiotics, has become a critical public health concern. Photothermal antibacterial therapy (PTAT) utilizes heat generated by photothermal agents under light exposure to inhibit bacterial growth without inducing resistance, attracting more and more attention. Quinoid conjugated polymers, especially para-azaquinodimethane (AQM) polymer, are a class of organic semiconductors known for efficient π-electron delocalization, near-infrared absorption, and narrow bandgap, showing great potential in the application of photothermal reagents. However, current AQM polymers face challenges related to their solubility, photostability, and biocompability. In this study, tetraglycol is introduced onto the AQM core for improving the drawbacks of the resulting polymers. Two AQM polymers with different electron donor (thiophene and 2,2'-bithiophene) are synthesized and evaluated for their various properties. PAQMT exhibited superior performance, including higher extinction coefficients, improved light absorption, and greater stability under repeated NIR irradiation. PAQMT is further developed into nanoparticles via encapsulation, resulting in excellent colloidal stability, effective bacterial inhibition under 808 nm NIR light. This work provides new strategy in improving the solubility, photostability, and photothermal properties of AQM polymers, offers opportunities for promoting the application of quinoidal conjugated polymers in PTAT.
Collapse
Affiliation(s)
- Zhide Hao
- School of Materials Science and Engineering, Hebei University of Technology, Tianjin, 300401, P. R. China
| | - Hailin Zhou
- School of Chemical Engineering and Technology, Hebei University of Technology, Tianjin, 300401, P. R. China
| | - Dong Gao
- Key Laboratory of Hebei Province for Molecular Biophysics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin, 300401, P. R. China
| | - Liang Qiu
- Key Laboratory of Hebei Province for Molecular Biophysics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin, 300401, P. R. China
| | - Chengfen Xing
- School of Materials Science and Engineering, Hebei University of Technology, Tianjin, 300401, P. R. China
- School of Chemical Engineering and Technology, Hebei University of Technology, Tianjin, 300401, P. R. China
- Key Laboratory of Hebei Province for Molecular Biophysics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin, 300401, P. R. China
| |
Collapse
|
29
|
Zhang K, Wang B, Sun F, Yang Z. Semiconducting Perylene Diimide J-aggregates Cross-linked Hydrogel Enables High-Efficiency Photothermal Controlled Release of Nitric Oxide for Antibiofilm Therapy. Adv Healthc Mater 2025; 14:e2404754. [PMID: 39924765 DOI: 10.1002/adhm.202404754] [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: 11/27/2024] [Revised: 01/18/2025] [Indexed: 02/11/2025]
Abstract
Antibiofilm treatment, particularly drug-containing wound healing dressings, does not typically penetrate the robust protective extracellular polymeric substance of biofilm and eradicate the bacteria. Here, a rational design of nitric oxide (NO) donor N,N'-di-sec-butyl-N,N'-dinitroso-1,4-phenylenediamine (BNN6)-based injectable hydrogel, is reported in which the NO release can be triggered by a photothermal effect owing to semiconducting perylene diimide (PDI) J-aggregation fibers. The synthetic PDI derivatives self-assembling into 0D nanoparticles and then aggregating to 1D J fiber is accompanied by absorbance red-shifting from 700 to 790 nm and then to 852 nm. After encapsulating BNN6, a "sandwich roll" (SR) like structure is evenly crosslinked into an injectable hydrogel (SRH) exhibiting a high photothermal convenience efficiency of 72%, which enables the SRH to achieve highly efficient photocontrol NO release. The SRH shows excellent injectability, shape adaptability, and effective antibacterial efficacy over 99% to the E.coli and S. aureus. and remarkable in vivo antibiofilm efficiency of 99.58% by laser irradiation. Furthermore, the synergistic treatment displays the ability to eliminate inflammation, facilitate angiogenesis, and promote collagen deposition, thereby significantly stimulating the healing process of wounds. The semiconducting J-aggregation injectable hydrogel can be a versatile strategy for the treatment of biofilm.
Collapse
Affiliation(s)
- Kangxin Zhang
- Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Key Laboratory of Flexible Electronics, Fujian Normal University and Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou, 350117, China
| | - Bo Wang
- Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Key Laboratory of Flexible Electronics, Fujian Normal University and Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou, 350117, China
| | - Fengwei Sun
- Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Key Laboratory of Flexible Electronics, Fujian Normal University and Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou, 350117, China
| | - Zhen Yang
- Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Key Laboratory of Flexible Electronics, Fujian Normal University and Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou, 350117, China
| |
Collapse
|
30
|
Wan R, Li W, Yang K, Li L, Wang S, Lei L, Tang H, Gu H. Immunomodulatory and bone regenerative properties of copper/procyanidins-modified titanium surfaces. BIOMATERIALS ADVANCES 2025; 169:214199. [PMID: 39894624 DOI: 10.1016/j.bioadv.2025.214199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 01/09/2025] [Accepted: 01/22/2025] [Indexed: 02/04/2025]
Abstract
The inflammatory response triggered by the interaction between implants and macrophages is essential for bone regeneration around these implants. This study presents the application of dopamine hydrochloride to develop a copper and procyanidins coating on titanium surfaces to investigate its effects on bacterial inhibition, macrophage polarization, and osteogenic differentiation. The results demonstrated that this copper/procyanidins coating significantly suppressed the growth of Escherichia coli and Staphylococcus aureus. Notably, the initial release of Cu2+ ions promoted macrophage polarization toward a pro-inflammatory phenotype while stimulating the secretion of anti-inflammatory factors. Subsequently, the reduced Cu2+ release combined with procyanidins facilitated the transition from M1 to M2 macrophages-an essential process for bacterial phagocytosis and bone regeneration. Furthermore, this coating enhanced the secretion of osteogenic factors by bone marrow mesenchymal stem cells, enhancing their osteogenic differentiation and integration with bone tissue. These findings highlight the potential of copper/procyanidins coating in developing implant surfaces with immune-modulating and sustained antibacterial properties.
Collapse
Affiliation(s)
- Rongxin Wan
- Central Laboratory, the Second Hospital of Tianjin Medical University, Tianjin 300211, China.
| | - Wenbo Li
- Department of Orthopaedics, the Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Kuo Yang
- Central Laboratory, the Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Lijun Li
- Department of Orthopaedics, the Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Shaojing Wang
- Central Laboratory, the Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Li Lei
- Central Laboratory, the Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Huiqin Tang
- Central Laboratory, the Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Hanqing Gu
- Central Laboratory, the Second Hospital of Tianjin Medical University, Tianjin 300211, China
| |
Collapse
|
31
|
Smith E, Matthews A, Westra ER, Custodio R. Disruption of Pseudomonas aeruginosa quorum sensing influences biofilm formation without affecting antibiotic tolerance. MICROBIOLOGY (READING, ENGLAND) 2025; 171:001557. [PMID: 40279159 PMCID: PMC12032407 DOI: 10.1099/mic.0.001557] [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: 09/23/2024] [Accepted: 04/07/2025] [Indexed: 04/26/2025]
Abstract
The opportunistic bacterial pathogen Pseudomonas aeruginosa is a leading cause of antimicrobial resistance-related deaths, and novel antimicrobial therapies are urgently required. P. aeruginosa infections are difficult to treat due to the bacterium's propensity to form biofilms, whereby cells aggregate to form a cooperative, protective structure. Autolysis, the self-killing of bacterial cells, and the bacterial cell-to-cell communication system, quorum sensing (QS), play essential roles in biofilm formation. Strains of P. aeruginosa that have lost the lasI/R QS system commonly develop in patients, and previous studies have characterized distinctive autolysis phenotypes in these strains. Yet, the underlying causes and implications of these autolysis phenotypes remain unknown. This study confirmed these autolysis phenotypes in the PA14 QS mutant strains, ΔlasI and ΔlasR, and investigated the consequences of QS loss and associated autolysis on biofilm formation and antibiotic susceptibility. QS mutants exhibited delayed biofilm formation but ultimately surpassed the wild-type (WT) in biofilm mass. However, the larger biofilm mass of the QS mutants was not reflected in higher live-cell numbers, indicating an altered biofilm structure. Nevertheless, QS mutant biofilms were not more susceptible to antibiotics than the WT. Artificial supplementation of ΔlasI with a QS signal molecule (autoinducer) restored the strain's QS system without the associated costs of QS, enabling ΔlasI to achieve higher pre-treatment and post-treatment live-cell numbers. Overall, the lack of QS functioning was not detrimental to biofilm antibiotic tolerance, though the artificial disruption of QS may reduce the advantages of QS mutants within in vivo mixed-strain populations. Much remains to be understood regarding the regulation and induction of the autolysis phenotypes observed in these strains, and future research to fully elucidate the control and consequences of autolysis may offer potential for novel antimicrobial therapies.
Collapse
Affiliation(s)
- Elvina Smith
- Environment and Sustainability Institute, Biosciences, University of Exeter, Penryn Campus, Penryn TR10 9FE, UK
| | - Andrew Matthews
- Environment and Sustainability Institute, Biosciences, University of Exeter, Penryn Campus, Penryn TR10 9FE, UK
| | - Edze R. Westra
- Environment and Sustainability Institute, Biosciences, University of Exeter, Penryn Campus, Penryn TR10 9FE, UK
| | - Rafael Custodio
- Environment and Sustainability Institute, Biosciences, University of Exeter, Penryn Campus, Penryn TR10 9FE, UK
- Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| |
Collapse
|
32
|
Kim S, Jin YH, Mah JH. Inhibitory effects of garlic, cinnamon, and rosemary on viability, heat resistance, and biofilm formation of Bacillus cereus spores in the broth of a fermented soybean paste stew, Cheonggukjang jjigae. Food Res Int 2025; 206:116078. [PMID: 40058924 DOI: 10.1016/j.foodres.2025.116078] [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: 09/25/2024] [Revised: 01/22/2025] [Accepted: 02/22/2025] [Indexed: 05/13/2025]
Abstract
Foods prepared through heating, including broths, have the potential and risk of survival of Bacillus cereus, which has the ability to form spores and biofilms. This study evaluated the efficacy of various natural products (particularly spices) in mitigating B. cereus contamination in Cheonggukjang jjigae (CJ) broth. The following characteristics of B. cereus were examined: viability of vegetative cells (including other pathogenic bacteria) and planktonic spores, heat resistance of planktonic spores and spores in intact biofilms, and biofilm formation and persistence. In an antimicrobial test to evaluate the inhibitory effects of spice and cruciferous vegetable extracts on B. cereus CH3 vegetative cells, cinnamon, garlic, and rosemary extracts were selected as they have shown significant inhibitory effects, with inhibition zones of 20-29 mm in diameter at the highest concentration tested (160 mg/mL, unless otherwise stated). These spice extracts also exhibited antimicrobial activity against other foodborne pathogens, including Staphylococcus aureus, Listeria monocytogenes, Salmonella Typhimurium, and Escherichia coli O157:H7. Garlic extract showed the greatest inhibitory effect on the viability and heat resistance of planktonic spores of B. cereus CH3, and cinnamon and rosemary extracts exhibited similar effects. Garlic extract reduced B. cereus CH3 spore counts in phosphate buffer solution (PBS) and CJ broth by 20.22 % and 14.08 %, respectively, compared to control (treated with the same ethanol amount instead of the extract), and effectively weakened spore heat resistance, reducing the D100°C-values of planktonic spores of B. cereus CH3 in PBS and CJ broth by 32.89 % and 23.08 %, respectively, compared to control. As for the characteristics related to biofilm, garlic extract showed the highest inhibitory effect on biofilm formation and persistence and heat resistance of spores in intact biofilms, followed by rosemary and cinnamon extracts. All three spice extracts completely inhibited biofilm formation even at the lowest concentration (20 mg/mL) at the early stage of biofilm formation. They completely eradicated biofilm persistence formed in brain heart infusion (BHI) and CJ broth at the highest concentration. A high garlic extract concentration (80 mg/mL) also reduced the D100°C-values of spores in biofilms formed in BHI and CJ broth by 16.34 % and 9.00 %, respectively, compared to control. Taken together, garlic extract was most effective in mitigating B. cereus contamination in a concentration-dependent manner in in vitro-menstrua and CJ broth. This study may provide one of the promising strategies to reduce the risk of B. cereus in soybean stews such as CJ.
Collapse
Affiliation(s)
- Sohyeon Kim
- Department of Food and Biotechnology, Korea University, Sejong 30019, Republic of Korea
| | - Young Hun Jin
- Department of Food and Biotechnology, Korea University, Sejong 30019, Republic of Korea
| | - Jae-Hyung Mah
- Department of Food and Biotechnology, Korea University, Sejong 30019, Republic of Korea.
| |
Collapse
|
33
|
Roujansky A, Diop S, Pasqueron J, Aparicio M, Cook F, Kallel H, Mounier R. Pathophysiology and Prevention of Ventriculostomy-Related Infections: A Review. Neurosurgery 2025; 96:744-750. [PMID: 39264162 DOI: 10.1227/neu.0000000000003181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 08/07/2024] [Indexed: 09/13/2024] Open
Abstract
This qualitative review aims to summarize current knowledge on ventriculostomy-related infection (VRI) pathophysiology and its prevention. VRI generally occurs at day 10, mainly because of Gram-positive cocci , after a cerebrospinal fluid leak. Skin microbiota and biofilm seem to play a major role in VRI pathogenesis. Colonization of external ventricular drain by biofilm is universal and occurs quickly after catheter insertion. However, pathogens from the skin are more often associated with VRI than commensal bacteria. A review of proposed preventive measures shows that none has proven to be fully efficient. Periprocedural and prolonged systemic prophylactic antimicrobials have not shown to prevent VRIs and may promote the emergence of more resistant or pathogenic strains. Antimicrobial and silver-impregnated external ventricular drains, although promising, have not demonstrated preventive effects and may modify bacterial ecology. These results are consistent with the proposed pathophysiology. Finally, we will present a few propositions for future research that may help in improving our knowledge and thus better prevent VRIs. Until then, given the available data, limiting the duration of ventricular drainage may be the most attainable option to prevent VRIs.
Collapse
Affiliation(s)
- Ariane Roujansky
- Réanimation polyvalente, Centre Hospitalier de Cayenne, Cayenne , French Guiana
- Tropical Biome et immunopathologie CNRS UMR-9017, Inserm U 1019, Université de Guyane, Cayenne , French Guiana
| | - Sylvain Diop
- Département d'Anesthésie et réanimation, Hôpital Marie Lannelongue, Le Plessis-Robinson , France
| | - Jean Pasqueron
- Service d'anesthésie-réanimation chirurgicale, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpitaux Universitaires Henri Mondor, Créteil , France
| | - Maxime Aparicio
- Department of Anaesthesiology and Critical Care, Georges Pompidou European Hospital, Paris , France
| | - Fabrice Cook
- Service d'anesthésie-réanimation, Centre Hospitalier du Sud Francilien, Corbeil-Essonnes , France
| | - Hatem Kallel
- Réanimation polyvalente, Centre Hospitalier de Cayenne, Cayenne , French Guiana
- Tropical Biome et immunopathologie CNRS UMR-9017, Inserm U 1019, Université de Guyane, Cayenne , French Guiana
| | - Roman Mounier
- Department of Anaesthesiology and Critical Care, Georges Pompidou European Hospital, Paris , France
- Université Paris Cité, Paris , France
- INSERM U955, équipe 15, institut Mondor de la recherche biomédicale, Université Paris-Est-Créteil, Créteil , France
| |
Collapse
|
34
|
Zhang D, Kukkar D, Bhatt P, Kim KH, Kaur K, Wang J. Novel nanomaterials-based combating strategies against drug-resistant bacteria. Colloids Surf B Biointerfaces 2025; 248:114478. [PMID: 39778220 DOI: 10.1016/j.colsurfb.2024.114478] [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: 11/09/2024] [Revised: 12/22/2024] [Accepted: 12/24/2024] [Indexed: 01/11/2025]
Abstract
Numerous types of contemporary antibiotic treatment regimens have become ineffective with the increasing incidence of drug tolerance. As a result, it is pertinent to seek novel and innovative solutions such as antibacterial nanomaterials (NMs) for the prohibition and treatment of hazardous microbial infections. Unlike traditional antibiotics (e.g., penicillin and tetracycline), the unique physicochemical characteristics (e.g., size dependency) of NMs endow them with bacteriostatic and bactericidal potential. However, it is yet difficult to mechanistically predict or decipher the networks of molecular interaction (e.g., between NMs and the biological systems) and the subsequent immune responses. In light of such research gap, this review outlines various mechanisms accountable for the inception of drug tolerance in bacteria. It also delineates the primary factors governing the NMs-induced molecular mechanisms against microbes, specifically drug-resistant bacteria along with the various NM-based mechanisms of antibacterial activity. The review also explores future directions and prospects for NMs in combating drug-resistant bacteria, while addressing challenges to their commercial viability within the healthcare industry.
Collapse
Affiliation(s)
- Daohong Zhang
- Yantai Key Laboratory of Nanoscience and Technology for Prepared Food, Yantai Engineering Research Center of Green Food Processing and Quality Control, College of Food Engineering, Ludong University, Yantai, Shandong 264025, China
| | - Deepak Kukkar
- Department of Biotechnology, Chandigarh University, Gharuan, Mohali 140413, India; University Center for Research and Development, Chandigarh University, Gharuan, Mohali 140413, India.
| | - Poornima Bhatt
- Department of Biotechnology, Chandigarh University, Gharuan, Mohali 140413, India; University Center for Research and Development, Chandigarh University, Gharuan, Mohali 140413, India
| | - Ki-Hyun Kim
- Department of Civil and Environmental Engineering, Hanyang University, 222 Wangsimni-Ro, Seoul 04763, South Korea.
| | - Kamalpreet Kaur
- Department of Chemistry, Mata Gujri College, Fatehgarh Sahib, Punjab 140406, India
| | - Jianlong Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, China
| |
Collapse
|
35
|
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
|
36
|
Xu W, Lin Z, Cortez-Jugo C, Qiao GG, Caruso F. Antimicrobial Phenolic Materials: From Assembly to Function. Angew Chem Int Ed Engl 2025; 64:e202423654. [PMID: 39905990 DOI: 10.1002/anie.202423654] [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: 12/04/2024] [Indexed: 02/06/2025]
Abstract
Infectious diseases pose considerable challenges to public health, particularly with the rise of multidrug-resistant pathogens that globally cause high mortality rates. These pathogens can persist on surfaces and spread in public and healthcare settings. Advances have been made in developing antimicrobial materials to reduce the transmission of pathogens, including materials composed of naturally sourced polyphenols and their derivatives, which exhibit antimicrobial potency, broad-spectrum activity, and a lower likelihood of promoting resistance. This review provides an overview of recent advances in the fabrication of antimicrobial phenolic biomaterials, where natural phenolic compounds act as active antimicrobial agents or encapsulate other antimicrobial agents (e.g., metal ions, antimicrobial peptides, natural biopolymers). Various forms of phenolic biomaterials synthesized through these two strategies, including antimicrobial particles, capsules, hydrogels, and coatings, are summarized, with a focus on their application in wound healing, bone repair and regeneration, oral health, and antimicrobial coatings for medical devices. The potential of these advanced phenolic biomaterials provides a promising therapeutic approach for combating antimicrobial-resistant infections and reducing microbial transmission.
Collapse
Affiliation(s)
- Wanjun Xu
- Department of Chemical Engineering, The University of Melbourne Parkville, Victoria, 3010, Australia
| | - Zhixing Lin
- Department of Chemical Engineering, The University of Melbourne Parkville, Victoria, 3010, Australia
| | - Christina Cortez-Jugo
- Department of Chemical Engineering, The University of Melbourne Parkville, Victoria, 3010, Australia
| | - Greg G Qiao
- Department of Chemical Engineering, The University of Melbourne Parkville, Victoria, 3010, Australia
| | - Frank Caruso
- Department of Chemical Engineering, The University of Melbourne Parkville, Victoria, 3010, Australia
| |
Collapse
|
37
|
Guo Z, Zhang T, Yang H, Zhu X, Lu S, Chen A, Fan M, Qu J. Unraveling tetracycline and its degradation product: Induction mechanisms of antibiotic resistance in Escherichia coli. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 970:178959. [PMID: 40023879 DOI: 10.1016/j.scitotenv.2025.178959] [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: 08/02/2024] [Revised: 02/11/2025] [Accepted: 02/22/2025] [Indexed: 03/04/2025]
Abstract
In aquatic environments, antibiotics degrade into byproducts, potentially enhancing bacterial resistance. However, the specific mechanisms by which these byproducts induce bacterial resistance remain elusive. This study conducted experimental evolution experiments to explore how E. coli adapts to tetracycline (TC) and its primary degradation products-anhydrotetracycline (ATC), epitetracycline (ETC), and 4-epianhydrotetracycline (EATC)-through evolution experiments. Prolonged exposure to TC and its byproducts significantly increased frequency of resistant mutants in E. coli ATCC25922, with a maximum 106-fold increase. Resistant mutants exhibited markedly elevated minimum inhibitory concentrations (MICs) for TC, ampicillin (AMP), and ciprofloxacin (CIP), indicating multidrug resistance. Transcriptomic analysis showed that the antibiotic resistance phenotype could be related to enhanced target protection, metabolic adaptations, and reduced membrane permeability. The induction pathways between TC and its byproducts were distinct. Specifically, TC20d (where TC20d represents the mutants collected after 20 days of continuous exposure to TC) was associated with more alterations in ribosome-associated genes, which was correlated with an enhanced defensive response as shown by the data. Moreover, variations in energy metabolism gene expression suggest a robust metabolic defense in ATC20d and ETC20d. When TC and its byproducts-ATC, ETC, and EATC-act together, they induce antibiotic resistant mutants at rates of 29.8 %, 18.9 %, 18.3 %, and 31.9 %, respectively. This study provides a descriptive overview of the possible adaptive mechanisms and pathways that may be involved in antibiotic resistance due to environmental exposure.
Collapse
Affiliation(s)
- Zhengfeng Guo
- State Environmental Protection Key Laboratory of Wetland Ecology and Vegetation Restoration, School of Environment, Northeast Normal University, Changchun 130117, China
| | - Tingting Zhang
- State Environmental Protection Key Laboratory of Wetland Ecology and Vegetation Restoration, School of Environment, Northeast Normal University, Changchun 130117, China
| | - Hao Yang
- State Environmental Protection Key Laboratory of Wetland Ecology and Vegetation Restoration, School of Environment, Northeast Normal University, Changchun 130117, China
| | - Xiaolin Zhu
- State Environmental Protection Key Laboratory of Wetland Ecology and Vegetation Restoration, School of Environment, Northeast Normal University, Changchun 130117, China
| | - Siyuan Lu
- State Environmental Protection Key Laboratory of Wetland Ecology and Vegetation Restoration, School of Environment, Northeast Normal University, Changchun 130117, China
| | - Anjie Chen
- State Environmental Protection Key Laboratory of Wetland Ecology and Vegetation Restoration, School of Environment, Northeast Normal University, Changchun 130117, China
| | - Mingyu Fan
- College of art, Hebei University of Economics and Business, Shijiazhuang 050000, China
| | - Jiao Qu
- State Environmental Protection Key Laboratory of Wetland Ecology and Vegetation Restoration, School of Environment, Northeast Normal University, Changchun 130117, China.
| |
Collapse
|
38
|
Pruitt HM, Zhu JC, Riley SP, Shi M. The Hidden Fortress: A Comprehensive Review of Fungal Biofilms with Emphasis on Cryptococcus neoformans. J Fungi (Basel) 2025; 11:236. [PMID: 40137272 PMCID: PMC11943451 DOI: 10.3390/jof11030236] [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: 02/21/2025] [Revised: 03/14/2025] [Accepted: 03/17/2025] [Indexed: 03/27/2025] Open
Abstract
Biofilms are structurally organized communities of microorganisms that adhere to a variety of surfaces. These communities produce protective matrices consisting of polymeric polysaccharides, proteins, nucleic acids, and/or lipids that promote shared resistance to various environmental threats, including chemical, antibiotic, and immune insults. While algal and bacterial biofilms are more apparent in the scientific zeitgeist, many fungal pathogens also form biofilms. These surprisingly common biofilms are morphologically distinct from the multicellular molds and mushrooms normally associated with fungi and are instead an assemblage of single-celled organisms. As a collection of yeast and filamentous cells cloaked in an extracellular matrix, fungal biofilms are an extreme threat to public health, especially in conjunction with surgical implants. The encapsulated yeast, Cryptococcus neoformans, is an opportunistic pathogen that causes both pulmonary and disseminated infections, particularly in immunocompromised individuals. However, there is an emerging trend of cryptococcosis among otherwise healthy individuals. C. neoformans forms biofilms in diverse environments, including within human hosts. Notably, biofilm association correlates with increased expression of multiple virulence factors and increased resistance to both host defenses and antifungal treatments. Thus, it is crucial to develop novel strategies to combat fungal biofilms. In this review, we discuss the development and treatment of fungal biofilms, with a particular focus on C. neoformans.
Collapse
Affiliation(s)
| | | | - Sean P. Riley
- Department of Veterinary Medicine, Virginia-Maryland College of Veterinary Medicine, University of Maryland, College Park, MD 20742, USA; (H.M.P.); (J.C.Z.)
| | - Meiqing Shi
- Department of Veterinary Medicine, Virginia-Maryland College of Veterinary Medicine, University of Maryland, College Park, MD 20742, USA; (H.M.P.); (J.C.Z.)
| |
Collapse
|
39
|
Iobbi V, Parisi V, Giacomini M, De Riccardis F, Brun P, Núñez-Pons L, Drava G, Giordani P, Monti MC, Poggi R, Murgia Y, De Tommasi N, Bisio A. Sesterterpenoids: sources, structural diversity, biological activity, and data management. Nat Prod Rep 2025; 42:443-481. [PMID: 39832137 DOI: 10.1039/d4np00041b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Reviewing the literature published up to October 2024.Sesterterpenoids are one of the most chemically diverse and biologically promising subgroup of terpenoids, the largest family of secondary metabolites. The present review article summarizes more than seven decades of studies on isolation and characterization of more than 1600 structurally novel sesterterpenoids, supplemented by biological, pharmacological, ecological, and geographic distribution data. All the information have been implemented in eight tables available on the web and a relational database https://sesterterpenoids.unige.net/. The interface has two sections, one open to the public for reading only and the other, protected by an authentication mechanism, for timely updating of published results.
Collapse
Affiliation(s)
- Valeria Iobbi
- Department of Pharmacy, University of Genova, Viale Cembrano 4, 16148 Genova, Italy.
| | - Valentina Parisi
- Department of Pharmacy, Via Giovanni Paolo II, 132, 84084 Fisciano, Salerno, Italy.
- NBFC, National Biodiversity Future Center, 90133 Palermo, Italy
| | - Mauro Giacomini
- Department of Informatics, Bioengineering, Robotics and System Science, University of Genova, Via all'Opera Pia 13, 16146 Genova, Italy
| | - Francesco De Riccardis
- Department of Chemistry and Biology "A. Zambelli", Via Giovanni Paolo II, 132, 84084 Fisciano, Salerno, Italy
| | - Paola Brun
- Department of Molecular Medicine, Section of Microbiology, University of Padova, Via A. Gabelli, 63, 35121 Padova, Italy
| | - Laura Núñez-Pons
- Department of Integrative Marine Ecology (EMI), Stazione Zoologica Anton Dohrn, Villa Comunale, 80121 Napoli, Italy
- NBFC, National Biodiversity Future Center, 90133 Palermo, Italy
| | - Giuliana Drava
- Department of Pharmacy, University of Genova, Viale Cembrano 4, 16148 Genova, Italy.
| | - Paolo Giordani
- Department of Pharmacy, University of Genova, Viale Cembrano 4, 16148 Genova, Italy.
| | - Maria Chiara Monti
- Department of Pharmacy, University of Napoli "Federico II", Via T. De Amicis 95, 80131 Napoli, Italy
| | - Roberto Poggi
- Museo Civico di Storia Naturale Giacomo Doria, Via Brigata Liguria 9, 16121 Genova, Italy
| | - Ylenia Murgia
- Department of Informatics, Bioengineering, Robotics and System Science, University of Genova, Via all'Opera Pia 13, 16146 Genova, Italy
| | - Nunziatina De Tommasi
- Department of Pharmacy, Via Giovanni Paolo II, 132, 84084 Fisciano, Salerno, Italy.
- NBFC, National Biodiversity Future Center, 90133 Palermo, Italy
| | - Angela Bisio
- Department of Pharmacy, University of Genova, Viale Cembrano 4, 16148 Genova, Italy.
| |
Collapse
|
40
|
Gonzalez-Henao S, Schrenk MO. An astrobiological perspective on microbial biofilms: their importance for habitability and production of detectable and lasting biosignatures. Appl Environ Microbiol 2025; 91:e0177824. [PMID: 39927769 PMCID: PMC11921390 DOI: 10.1128/aem.01778-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2025] Open
Abstract
The search for life elsewhere in the universe has remained one of the main goals of astrobiological exploration. In this quest, extreme environments on Earth have served as analogs to study the potential habitability of Mars and icy moons, which include but are not limited to hydrothermal vent systems, acid lakes, deserts, and polar ice, among others. Within the various forms that life manifests, biofilms constitute one of the most widespread phenotypes and are ubiquitous in extreme environments. Biofilms are structured communities of microorganisms enclosed in a matrix of extracellular polymeric substances (EPS) that protect against unfavorable and dynamic conditions. These concentrated structures and their associated chemistry may serve as unique and persistent signatures of life processes that may aid in their detection. Here we propose biofilms as a model system to understand the habitability of extraterrestrial systems and as sources of recognizable and persistent biosignatures for life detection. By testing these ideas in extreme analog environments on Earth, this approach could be used to guide and focus future exploration of samples encompassing the geologic record of early Earth as well as other planets and moons of our solar system.
Collapse
Affiliation(s)
- Sarah Gonzalez-Henao
- Department of Microbiology, Genetics, and Immunology, Michigan State University, East Lansing, Michigan, USA
- Department of Earth and Environmental Sciences, Michigan State University, East Lansing, Michigan, USA
| | - Matthew O. Schrenk
- Department of Microbiology, Genetics, and Immunology, Michigan State University, East Lansing, Michigan, USA
- Department of Earth and Environmental Sciences, Michigan State University, East Lansing, Michigan, USA
| |
Collapse
|
41
|
Rahman MA, Akter P, Habib MR, Rahman MA, Mahiuddin M, Rahman MM, Islam MS, Miah MAJ, Ahmad H. Functionalization of Biomimetic Polydopamine Shells Constructed onto Bismuth-Core Particles for pH-Mediated Drug Targeting to Heal Bacterial Infections. Bioconjug Chem 2025; 36:563-577. [PMID: 39940090 DOI: 10.1021/acs.bioconjchem.5c00003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2025]
Abstract
Nonhealing chronic bacterial infections are very challenging to both patients and the healthcare-providing system. Multimodal therapy enhances the antibiotic efficacy to treat infections via combating multidrug resistance through cumulative therapeutic effects. Functionalized polydopamine (PDA)-coated Bi particles having a core-shell structure may treat such chronic infections. We fabricated a new advanced material based on Tris-functionalized PDA and Bi using a facile three-step protocol for healing drug-resistant bacterial infections. The fabrication of Bi particles, PDA coating on Bi particles, and their Tris functionalization were confirmed by X-ray diffraction, and spectroscopic and thermogravimetric analyses. Tris-functionalized PDA-coated Bi particles, abbreviated as Bi/PDA-Tris, exhibited a higher average diameter, improved hydrophilicity, aqueous dispersity, and colloidal stability. Bi/PDA-Tris showed a delicate surface morphology, narrow size distribution, spherical shape, and core-shell structure. In vitro bovine serum albumin and hemolysis assays showed minimal protein adsorption and the desirable hemocompatibility of Bi/PDA-Tris. Antibacterial gentamicin (GM)-immobilized Bi/PDA-Tris showed pH-mediated sustained drug release kinetics under acidic conditions. The in vitro study of GM-loaded Bi/PDA-Tris particles exhibited significant bacterial growth inhibition and bactericidal activity. Tris functionalization effectively enhances the antibacterial efficacy of the PDA shell under acidic conditions to target and heal bacterial infections. This approach has introduced economic, nontoxic, easy-to-use, relatively more biocompatible Bi particles as a substituent for precise metals like Pt, Au, and Ag for the development of core-shell composite materials.
Collapse
Affiliation(s)
- Md Abdur Rahman
- Polymer Colloids and Nanomaterials Research Lab, Department of Chemistry, Faculty of Science, University of Rajshahi, Rajshahi 6205, Bangladesh
| | - Pinky Akter
- Polymer Colloids and Nanomaterials Research Lab, Department of Chemistry, Faculty of Science, University of Rajshahi, Rajshahi 6205, Bangladesh
| | - Md Rowshanul Habib
- Department of Biochemistry and Molecular Biology, Faculty of Science, Rajshahi University, Rajshahi 6205, Bangladesh
| | - Md Ataur Rahman
- Polymer Colloids and Nanomaterials Research Lab, Department of Chemistry, Faculty of Science, University of Rajshahi, Rajshahi 6205, Bangladesh
| | - Md Mahiuddin
- Chemistry Discipline, University of Khulna, Khulna 9208, Bangladesh
| | - Md Mahbubor Rahman
- Polymer Colloids and Nanomaterials Research Lab, Department of Chemistry, Faculty of Science, University of Rajshahi, Rajshahi 6205, Bangladesh
| | - Md Shahidul Islam
- Polymer Colloids and Nanomaterials Research Lab, Department of Chemistry, Faculty of Science, University of Rajshahi, Rajshahi 6205, Bangladesh
| | - M A Jalil Miah
- Polymer Colloids and Nanomaterials Research Lab, Department of Chemistry, Faculty of Science, University of Rajshahi, Rajshahi 6205, Bangladesh
| | - Hasan Ahmad
- Polymer Colloids and Nanomaterials Research Lab, Department of Chemistry, Faculty of Science, University of Rajshahi, Rajshahi 6205, Bangladesh
| |
Collapse
|
42
|
Biswas P, Bose S, Chakraborty S. Vitamin D3 potentiates antimicrobial and antibiofilm activities of streptomycin and thymoquinone against Pseudomonas aeruginosa. World J Microbiol Biotechnol 2025; 41:104. [PMID: 40074989 DOI: 10.1007/s11274-025-04304-y] [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: 12/31/2024] [Accepted: 02/22/2025] [Indexed: 03/14/2025]
Abstract
Biofilm formed by Pseudomonas aeruginosa is a three dimensional microbial matrix that confers multidrug resistance properties along with the proficiency to evade the host immune system. The present study aims to determine the combinatorial effects of vitamin D3 (cholecalciferol) with two already reported antibiofilm agents: streptomycin and thymoquinone separately against P. aeruginosa biofilms. The minimum inhibitory concentration of streptomycin, thymoquinone and D3 was found to be 20, 10 and 100 μg/mL respectively. The inhibition of biofilm formation and pre-formed biofilm disintegration properties of streptomycin and thymoquinone alone or in combination with D3 at their sub-MIC concentration was determined by crystal violet staining and confocal laser scanning microscopy. A significant inhibition of metabolic activities like oxygen consumption rate and reduction in quorum sensing related cellular activities like swarming motilities, pyocyanin production and extracellular protease secretion by P. aeruginosa were also observed as a result of this combinatorial effect. Both of these combinatorial applications were found to accumulate ROS in bacterial cells, which has been proved to be one of the main causes of their antibiofilm activity. Effect of these two drug combinations on bacterial lettuce leaf infection was also evaluated. Molecular docking analysis indicated that thymoquinone combined D3 can interact more efficiently with the quorum sensing proteins LasI and LasR. The host cell cytotoxicity of these two combinations was found to be negligible on the murine macrophage cell line. These findings suggest that D3 potentiates the antimicrobial and antibiofilm efficacy of both streptomycin and thymoquinone against P. aeruginosa. Although both combinations have shown significant antibiofilm and antimicrobial potential, combinatorial performances of D3 combined thymoquinone were found to be more promising.
Collapse
Affiliation(s)
- Priyam Biswas
- Department of Biochemistry, University of Calcutta, Kolkata, West Bengal, 700 019, India
| | - Soham Bose
- Department of Biochemistry, University of Calcutta, Kolkata, West Bengal, 700 019, India
| | - Sudipta Chakraborty
- Department of Microbiology, Government General Degree College, Narayangarh, Rathipur, Narayangarh, Midnapore, West Bengal, 721437, India.
| |
Collapse
|
43
|
Sleutel M, Sogues A, Van Gerven N, Jonsmoen UL, Aspholm M, Van Molle I, Fislage M, Theunissen L, Bellis N, Baquero D, Egelman E, Krupovic M, Wang J, Remaut H. Cryo-EM analysis of the Bacillus thuringiensis extrasporal matrix identifies F-ENA as a widespread family of endospore appendages across Firmicutes. RESEARCH SQUARE 2025:rs.3.rs-6050303. [PMID: 40162231 PMCID: PMC11952670 DOI: 10.21203/rs.3.rs-6050303/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
For over 100 years, Bacillus thuringiensis (Bt) has been used as an agricultural biopesticide to control pests caused by insect species in the orders of Lepidoptera, Diptera and Coleoptera. Under nutrient starvation, Bt cells differentiate into spores and associated toxin crystals that can adopt biofilm-like aggregates. We reveal that such Bt spore/toxin biofilms are embedded in a fibrous extrasporal matrix (ESM), and using cryoID, we resolved the structure and molecular identity of an uncharacterized type of pili, referred to here as Fibrillar ENdospore Appendages or 'F-ENA'. F-ENA are monomolecular protein polymers tethered to the exosporium of Bt and are decorated with a flexible tip fibrillum. Phylogenetic analysis reveals that F-ENA is widespread not only in the class Bacilli, but also in the class Clostridia, and the cryoEM structures of F-ENA filaments from Bacillus, Anaerovorax and Paenibaccilus reveal subunits with a generic head-neck domain structure, where the b-barrel neck of variable length latch onto a preceding head domain through short N-terminal hook peptides. In Bacillus, two collagen-like proteins (CLP) respectively tether F-ENA to the exosporium (F-Anchor), or constitute the tip fibrillum at the distal terminus of F-ENA (F-BclA). Sedimentation assays point towards F-ENA involvement in spore-spore clustering, likely mediated via F-BclA contacts and F-ENA bundling through the antiparallel interlocking of the head-neck units.
Collapse
|
44
|
Zheng S, Tu Y, Li B, Qu G, Li A, Peng X, Li S, Shao C. Antimicrobial peptide biological activity, delivery systems and clinical translation status and challenges. J Transl Med 2025; 23:292. [PMID: 40055730 PMCID: PMC11887333 DOI: 10.1186/s12967-025-06321-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 02/25/2025] [Indexed: 05/13/2025] Open
Abstract
Antibiotic resistance is currently one of the most significant threats to global public health and safety. And studies have found that over the next 25 years, 39 million people will die directly and 169 million indirectly due to antibiotic-resistant diseases. Consequently, the development of new types of antimicrobial drugs is urgently needed. Antimicrobial peptides (AMPs) constitute an essential component of the innate immune response in all organisms. They exhibit a distinctive mechanism of action that endows them with a broad spectrum of biological activities, including antimicrobial, antibiofilm, antiviral, and anti-inflammatory effects. However, AMPs also present certain limitations, such as cytotoxicity, susceptibility to protein hydrolysis, and poor pharmacokinetic properties, which have impeded their clinical application. The development of delivery systems can address these challenges by modifying AMP delivery and enabling precise, controlled release at the site of infection or disease. This review offers a comprehensive analysis of the mechanisms of action and biological advantages of AMPs. and systematically evaluate how emerging drug delivery systems, such as nanoparticles and hydrogels, enhance the stability and bioavailability of AMPs, discussing both their strengths and limitations. Moreover, unlike previous reviews, this review highlight the most recent clinically approved AMP-based drugs and those currently in development, emphasizing the key challenges in translating these drugs into clinical practice. With these perspectives, it is hoped that this review will provide some insights into overcoming translational barriers and advancing AMPs drugs into clinical practice.
Collapse
Affiliation(s)
- Sainan Zheng
- Department of Pharmacy, Yueqing Third People's Hospital, Wenzhou, 325604, People's Republic of China
| | - Yuhan Tu
- Department of Pharmacy, Yueqing Third People's Hospital, Wenzhou, 325604, People's Republic of China
- Institute of Life Sciences, Wenzhou University, Wenzhou, 325035, People's Republic of China
| | - Bin Li
- Institute of Life Sciences, Wenzhou University, Wenzhou, 325035, People's Republic of China
| | - Gaoer Qu
- Department of Pharmacy, Yueqing Third People's Hospital, Wenzhou, 325604, People's Republic of China
| | - Anqi Li
- Institute of Life Sciences, Wenzhou University, Wenzhou, 325035, People's Republic of China
| | - Xuemei Peng
- Institute of Life Sciences, Wenzhou University, Wenzhou, 325035, People's Republic of China
| | - Shijun Li
- Institute of Life Sciences, Wenzhou University, Wenzhou, 325035, People's Republic of China.
| | - Chuanfeng Shao
- Department of Pharmacy, Yueqing Third People's Hospital, Wenzhou, 325604, People's Republic of China.
| |
Collapse
|
45
|
Yuan VG, Xia A, Santa Maria PL. Chronic suppurative otitis media: disrupted host-microbial interactions and immune dysregulation. Front Immunol 2025; 16:1547206. [PMID: 40114926 PMCID: PMC11923626 DOI: 10.3389/fimmu.2025.1547206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 02/17/2025] [Indexed: 03/22/2025] Open
Abstract
Recent research has uncovered new mechanisms that disrupt the balance between the host and microbes in the middle ear, potentially leading to dysbiosis and chronic suppurative otitis media (CSOM). Dysbiotic microbial communities, including core pathogens such as persister cells, are recognized for displaying cooperative virulence. These microbial communities not only evade the host's immune defenses but also promote inflammation that leads to tissue damage. This leads to uncontrolled disorder and pathogen proliferation, potentially causing hearing loss and systemic complications. In this discussion, we examine emerging paradigms in the study of CSOM that could provide insights into other polymicrobial inflammatory diseases. Additionally, we underscore critical knowledge gaps essential for developing a comprehensive understanding of how microbes interact with both the innate and adaptive immune systems to trigger and maintain CSOM.
Collapse
Affiliation(s)
- Vincent G. Yuan
- Department of Otolaryngology-Head and Neck Surgery, University of Pittsburgh Medical Center, Pittsburg, PA, United States
| | - Anping Xia
- Department of Otolaryngology-Head and Neck Surgery, University of Pittsburgh Medical Center, Pittsburg, PA, United States
| | - Peter L. Santa Maria
- Department of Otolaryngology-Head and Neck Surgery, University of Pittsburgh Medical Center, Pittsburg, PA, United States
| |
Collapse
|
46
|
Kang HED, Costalonga M, Vandereydt B, Varanasi KK. Design of Antibiofouling Lubricant-Impregnated Surfaces Robust to Cell-Growth-Induced Instability. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2025; 41:5000-5008. [PMID: 39983042 DOI: 10.1021/acs.langmuir.4c03783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/23/2025]
Abstract
Biofouling, commonly referred to as the unwanted deposition of cells on wetted solids, is a serious operational and environmental issue in many underwater and biomedical applications. Over the past decade, lubricant-impregnated surfaces (LIS) arose as a potential solution to prevent fouling, owing to their unique layer of lubricant masking the solid from the outer environment, thereby preventing biofouling. However, living microorganisms alter their environment by reproducing and secreting biomolecules, which can threaten the stability of such coatings over time. In this paper, we show that secretion of biomolecules from aquatic cells and subsequent changes in the interfacial tension of the surrounding media can trigger dewetting of the lubricant, ultimately exposing the surface to the outer solution and therefore becoming prone to fouling. By observing LIS immersed in Nannochloropsis oculata algae solutions at various stages of population growth, we establish a correlation between the decrease in interfacial tension and wetting states of the surface. We also visualize dewetting of the lubricant through confocal imaging performed in situ. Finally, we establish a diagram providing fundamental insights to design sturdy LIS circumventing such dewetting, therefore ensuring long-term protection against biofouling upon extended immersion in living cell solutions.
Collapse
Affiliation(s)
- Ha Eun David Kang
- Department of Mechanical Engineering, MIT, Cambridge, Massachusetts 02139, United States
| | - Maxime Costalonga
- Department of Mechanical Engineering, MIT, Cambridge, Massachusetts 02139, United States
| | - Bert Vandereydt
- Department of Mechanical Engineering, MIT, Cambridge, Massachusetts 02139, United States
| | - Kripa K Varanasi
- Department of Mechanical Engineering, MIT, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
47
|
Martin A, Doyle N, O'Mahony TF. Sodium dichloroisocyanurate: a promising candidate for the disinfection of resilient drain biofilm. Infect Prev Pract 2025; 7:100446. [PMID: 40008271 PMCID: PMC11850130 DOI: 10.1016/j.infpip.2025.100446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 01/03/2025] [Indexed: 02/27/2025] Open
Abstract
Background Biofilms are complex multicellular communities of microorganisms embedded within a protective matrix which confers resistance to various antimicrobials, including biocides. Biofilms can cause a range of human diseases and are responsible for 1.7 million hospital-acquired infections in the US annually, providing an economic burden of $11.5 billion in treatment costs. Biofilm contained within drain and plumbing systems may contain pathogenic viruses and bacteria which pose a significant risk to patient safety within healthcare environments. Aim The aim of this study was to determine if three hospital-grade disinfectants (sodium dichloroisocyanurate, peracetic acid and sodium hypochlorite) were capable of killing microorganisms within biofilm, and thus, determining their potential as candidates for drain biofilm disinfection. Methods Pseudomonas aeruginosa biofilms were cultivated using the CDC biofilm reactor, a standardised method for determining disinfectant efficacy against biofilm within the United States of America. Each disinfectant was tested using a one-minute contact time, using the highest concentration available on the product label. Findings The sodium dichloroisocyanurate product was successful in killing biofilm microorganisms, resulting in a log reduction of ≥ 8.70. Peracetic acid reduced biofilm by 3.82 log10 units, followed by sodium hypochlorite, which produced a reduction of 3.78 log10 units. Conclusions The use of a highly effective disinfectant with proven biofilm efficacy can help ensure patient safety and reduce infection levels. Drains and plumbing systems provide a reservoir for potential pathogens and biofilm; thus, drain disinfection is critical in reducing the instance of hospital-acquired infections. Sodium dichloroisocyanurate may provide a reliable solution for drain applications and subsequently, patient wellbeing and safety.
Collapse
Affiliation(s)
- Abbie Martin
- Microbiology & Validation Technician, R&D Department, Kersia Healthcare, Wexford, Ireland
| | - Natasha Doyle
- Principal R&D Scientist, R&D Department, Kersia Healthcare, Wexford, Ireland
| | - Tom F. O'Mahony
- R&D Manager Healthcare, R&D Department, Kersia Healthcare, Wexford, Ireland
| |
Collapse
|
48
|
Higashihira S, Simpson SJ, Arnold CJ, Deckard ER, Meneghini RM, Greenfield EM, Buller LT. Biofilm Formation is Durably Prevented on Pre-Fabricated Antibiotic Cement Spacers Compared to Cobalt Chrome and Polyethylene. J Arthroplasty 2025; 40:779-785. [PMID: 39233097 DOI: 10.1016/j.arth.2024.08.046] [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: 02/06/2024] [Revised: 08/26/2024] [Accepted: 08/28/2024] [Indexed: 09/06/2024] Open
Abstract
BACKGROUND A 2-stage revision remains the standard for managing chronic periprosthetic joint infection. Despite multiple spacer options, whether a particular one better resists biofilm formation remains unclear. Prefabricated polymethylmethacrylate (PMMA) articulating spacers containing antibiotics and a proprietary pore structure were developed to increase antibiotic elution characterized by a rapid burst phase for the initial one to two days and an extended slow-release phase for > 28 days. This in vitro study determined whether biofilm formation is prevented during the initial rapid burst phase and/or the slow-release phase. METHODS S. aureus-Xen36 was incubated in 1.5 mL of Luria-Bertani broth with PMMA discs with the proprietary pore structure either with or without gentamycin and vancomycin or with 'Hoffman style' positive-control discs (ultra-high molecular weight polyethylene or cobalt-chrome). Nonadherent bacteria were removed by three phosphate buffered saline rinses every 20 to 24 hours. Planktonic bacterial growth in the culture broth and biofilm formation on the discs were measured by colony forming unit (CFU) counting and resazurin reduction assays. Experiments were repeated > four times. RESULTS No detectable planktonic bacterial growth or biofilm formation occurred in cultures containing PMMA with antibiotics (≤ 15 CFUs/disc), whereas biofilms formed on PMMA without antibiotics, ultra-high molecular weight polyethylene, and cobalt-chrome (1 × 107 to 4 × 108 CFUs/disc, P < 0.0001). Biofilm formation was confirmed by a 100-fold decrease in sensitivity to vancomycin. To determine whether the antibiotic slow-release phase is sufficient to block biofilm formation, PMMA discs with antibiotics were preeluted for 14 days with multiple saline changes prior to bacterial inoculation. After antibiotic elution, still no detectable biofilms formed on PMMA discs with antibiotics (≤ 15 CFUs/disc, P < 0.0001). CONCLUSIONS Antibiotic release during both the initial and slow-release phases prevented biofilm formation on PMMA with the proprietary pore structure. This may translate into improved infection eradication rates clinically.
Collapse
Affiliation(s)
- Shota Higashihira
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana; Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, Indiana; Department of Orthopaedic Surgery, Yokohama City University Medical Center, Yokohama, Kanagawa, Japan
| | - Stefanie J Simpson
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana; Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, Indiana
| | - Christopher J Arnold
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Evan R Deckard
- Indiana Joint Replacement Institute, Indianapolis, Indiana
| | | | - Edward M Greenfield
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana; Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, Indiana
| | - Leonard T Buller
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
49
|
Wang C, Shahriar SMS, Su Y, Xie J. Versatile nanomaterials used in combatting biofilm infections. Nanomedicine (Lond) 2025; 20:501-518. [PMID: 39887017 PMCID: PMC11875486 DOI: 10.1080/17435889.2025.2459049] [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: 11/27/2024] [Accepted: 01/23/2025] [Indexed: 02/01/2025] Open
Abstract
Microbial infections are a pressing global health issue, exacerbated by the rise of antibiotic-resistant bacteria due to widespread antibiotic overuse. This resistance diminishes the effectiveness of current treatments, intensifying the need for new antimicrobial agents and innovative drug delivery strategies. Nanotechnology presents promising solutions, leveraging the unique properties of nanomaterials such as tunable optical and electronic characteristics, nanoscale size, and high surface-to-volume ratios. These features enhance their effectiveness as innovative antimicrobial agents and versatile drug delivery systems. This minireview classifies antimicrobial nanomaterials into four categories based on their mechanisms of action: thermal generation, reactive oxygen species generation, gas generation, and nanocarrier systems such as liposomes, polymersomes, and metal-organic frameworks. Uniquely, this review integrates a comparative analysis of these mechanisms, highlighting their relative advantages, limitations, and applications across diverse microbial targets. Additionally, it identifies emerging trends in the field, providing a forward-looking perspective on how recent advancements in nanotechnology can be leveraged to address unmet clinical needs. Finally, this article discusses future directions and emerging opportunities in antimicrobial nanotechnology.
Collapse
Affiliation(s)
- Chenlong Wang
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - S. M. Shatil Shahriar
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Yajuan Su
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jingwei Xie
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Mechanical and Materials Engineering, University of Nebraska Lincoln, Lincoln, NE, USA
| |
Collapse
|
50
|
Padmavathi AR, Karthikeyan B, Rao TS, Senthil Kumar J, Murthy PS. Polydimethylsiloxane loaded capsaicin afflicts membrane integrity, metabolic activity and biofilm formation of nosocomial pathogens. Microb Pathog 2025; 200:107282. [PMID: 39761772 DOI: 10.1016/j.micpath.2025.107282] [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: 06/07/2024] [Revised: 12/03/2024] [Accepted: 01/04/2025] [Indexed: 01/11/2025]
Abstract
Biofilms constitute 80 % of all nosocomial infections associated with invasive medical devices. Polydimethylsiloxane, a highly elastic, inert, non-reactive, biocompatible silicone polymer is widely used as implant biomaterial due to its non-toxic and low-immunogenic nature. Owing to its hydrophobicity, PDMS suffers from microbial adhesion. Inhibition of biofilm formation on PDMS surfaces is imperative to prevent morbidity, mortality and replacement of implants. The present study investigates the efficacy of capsaicin (0.5 % w/v) loaded PDMS as a broad spectrum antimicrobial surface against Staphylococcus aureus, Escherichia coli and Candida albicans. Capsaicin exhibited minimum inhibitory concentration of 1024 μg mL-1 for S. aureus, E. coli and 256 μg mL-1 for C. albicans. Capsaicin inhibited biofilms of S. aureus, E. coli and C. albicans at much lower concentrations of 2, 64 and 8 μg mL-1 respectively. The minimum capsaicin concentrations required for total biofilm eradication was found to be 256, 512, 128 μg mL-1 for S. aureus, E. coli and C. albicans respectively. Probing sub-lethal concentrations of capsaicin revealed 38, 32, 30 % reduction in metabolic activity of S. aureus, E. coli & C. albicans planktonic cells respectively. Similarly, there was an increase in permeability of cells to propidium iodide compared to control. By reducing the metabolic activity and perturbing membrane integrity, capsaicin could prevent biofilm formation and this was also observed with capsaicin-PDMS surfaces that exhibited 1 log (∼90 %) reduction of viable bacterial counts.
Collapse
Affiliation(s)
- Alwar Ramanujam Padmavathi
- Biofouling and Biofilm Processes Section, Water and Steam Chemistry Division, Bhabha Atomic Research Centre Facilities, Kalpakkam, 603 102, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai, 400 094, India
| | - B Karthikeyan
- Department of Biotechnology, Sri Krishna Arts and Science College, Coimbatore, 641008, India
| | - Toleti Subba Rao
- Biofouling and Biofilm Processes Section, Water and Steam Chemistry Division, Bhabha Atomic Research Centre Facilities, Kalpakkam, 603 102, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai, 400 094, India
| | - J Senthil Kumar
- Department of Biotechnology, PSG College of Arts & Science, Coimbatore, 641014, India
| | - P Sriyutha Murthy
- Biofouling and Biofilm Processes Section, Water and Steam Chemistry Division, Bhabha Atomic Research Centre Facilities, Kalpakkam, 603 102, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai, 400 094, India.
| |
Collapse
|