1
|
Shingade JA, Padalkar NS, Shin JH, Kim YH, Park TJ, Park JP, Patil AR. Electrostatically assembled maghemite nanoparticles-Lactobacillus plantarum: A novel hybrid for enhanced antioxidant, antimicrobial, and antibiofilm efficacy. BIORESOURCE TECHNOLOGY 2025; 430:132538. [PMID: 40228722 DOI: 10.1016/j.biortech.2025.132538] [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: 12/01/2024] [Revised: 04/10/2025] [Accepted: 04/11/2025] [Indexed: 04/16/2025]
Abstract
Excessive antibiotic use contributes to oxidative stress and microbial imbalance, leading to increased growth of pathogens and biofilm formation. To address this, we developed a novel electrostatically assembled hybrid of maghemite nanoparticles and Lactobacillus plantarum (MNPs-LAB) as a multifunctional agent. Structural and surface interactions were confirmed through X-ray diffraction (XRD), Fourier-transform infrared spectroscopy (FTIR), and X-ray photoelectron spectroscopy (XPS), while transmission electron microscopy (TEM) and field emission scanning electron microscopy (FESEM) analyses revealed a uniform distribution of MNPs on the LAB surface. The MNPs-LAB hybrid exhibited strong antioxidant activity (71.45 % at 500 µg/mL) and enhanced antimicrobial performance against Listeria monocytogenes. In addition, the hybrid inhibited biofilm formation and effectively eradicated preformed biofilms of Staphylococcus succinus, Listeria monocytogenes, Escherichia coli, and Salmonella thompson. Notably, the low cytotoxicity observed in Caco-2 cells indicated good biocompatibility with intestinal epithelial cells. These results highlight the potential of MNPs-LAB hybrid as a safe and effective therapeutic candidate for combating oxidative stress, microbial infections, and biofilm-associated challenges.
Collapse
Affiliation(s)
- Jayshri A Shingade
- Centre for Interdisciplinary Research, D. Y. Patil Education Society (Deemed to be University), Kolhapur 416 006, India; Department of Food Science and Technology, GreenTech-based Food Safety Research Group, BK21 Four, Chung-Ang University, Anseong 17546, Republic of Korea
| | - Navnath S Padalkar
- Department of Food Science and Technology, GreenTech-based Food Safety Research Group, BK21 Four, Chung-Ang University, Anseong 17546, Republic of Korea
| | - Jae Hwan Shin
- Department of Food Science and Technology, GreenTech-based Food Safety Research Group, BK21 Four, Chung-Ang University, Anseong 17546, Republic of Korea
| | - Yeong Hyeock Kim
- Department of Chemistry, Research Institute of Chem-Bio Diagnostic Technology, Chung-Ang University, 84, Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Tae Jung Park
- Department of Chemistry, Research Institute of Chem-Bio Diagnostic Technology, Chung-Ang University, 84, Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Jong Pil Park
- Department of Food Science and Technology, GreenTech-based Food Safety Research Group, BK21 Four, Chung-Ang University, Anseong 17546, Republic of Korea.
| | - Abhinandan R Patil
- Department of Pharmaceutics, D. Y. Patil Education Society (Deemed to Be University), Kolhapur 416 006, India.
| |
Collapse
|
2
|
Xu Y, Da X, Jian Y, Zhou W, Wu A, Wu Y, Peng Y, Liu X, Shi Y, Wang X, Zhou Q. A highly positively charged Ru(II) complex with photo-labile ligands for selective and efficient photo-inactivation of intracellular Staphylococcus aureus. J Inorg Biochem 2025; 268:112908. [PMID: 40209460 DOI: 10.1016/j.jinorgbio.2025.112908] [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: 12/14/2024] [Revised: 03/12/2025] [Accepted: 04/01/2025] [Indexed: 04/12/2025]
Abstract
Due to the protection afforded by host cells, intracellular Staphylococcus aureus (S. aureus), particularly methicillin-resistant S. aureus (MRSA), poses a significantly greater challenge to eliminate compared to the extracellular counterparts. It is highly desirable to develop novel antibacterial agents which are capable of selectively and efficiently eradicating intracellular bacteria, including drug-resistant strains, while being less prone to induce bacterial resistance. In this work, two Ru(II) complexes (Ru1 and Ru2) with photo-labile ligands were designed and synthesized. Both Ru1 and Ru2 could covalently bind to DNA after photo-induced ligand dissociation. Compared to Ru1, the incorporation of a triphenylamine group adorned with two positively charged cationic pyridine units significantly boosts the DNA binding constant, bacterial binding/uptake level, and subsequently, the antibacterial activity of Ru2. Ru2 could selectively photo-inactivate intracellular S. aureus and MRSA, being more efficient than vancomycin both in vitro and in vivo. Interestingly, after 20 days' treatment at sublethal concentrations, S. aureus cells exhibited no obvious drug resistance towards Ru2 upon irradiation. Such appealing results may provide new sights for developing novel antibacterial agents against intractable intracellular pathogens and also prevalent drug resistance.
Collapse
Affiliation(s)
- Yunli Xu
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xuwen Da
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Yao Jian
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wanpeng Zhou
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Aifeng Wu
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yao Wu
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yatong Peng
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiulian Liu
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yu Shi
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xuesong Wang
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Qianxiong Zhou
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China.
| |
Collapse
|
3
|
Horng YT, Chien CC, Dewi Panjaitan NS, Tseng SW, Chen HW, Yang HC, Chen YY, Soo PC. Sucrose reduces biofilm formation by Klebsiella pneumoniae through the PTS components ScrA and Crr. Biofilm 2025; 9:100269. [PMID: 40130064 PMCID: PMC11932656 DOI: 10.1016/j.bioflm.2025.100269] [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: 10/24/2024] [Revised: 02/21/2025] [Accepted: 03/03/2025] [Indexed: 03/26/2025] Open
Abstract
The presence of sucrose at concentrations of 0.5-5% can either increase bacterial biofilms (Streptococcus mutans and Escherichia coli) or have no significant effect on biofilms (Pseudomonas aeruginosa and Staphylococcus aureus). However, our study revealed that 1 % sucrose reduced the biofilm formation by Klebsiella pneumoniae STU1. To explore the role of the phosphoenolpyruvate-dependent-carbohydrate: phosphotransferase system (PTS) in regulating this process, the scrA gene, which encodes the sucrose-specific EIIBC of the PTS, was deleted in K. pneumoniae to create a scrA mutant (ΔscrA). Thereafter, we observed that the biofilm formation and type 3 fimbriae production were not affected by sucrose in the ΔscrA while sucrose reduced these processes in the wild type. Furthermore, we discovered that Crr, the glucose-specific EIIA of PTS, was the primary but not the sole EIIA of ScrA in K. pneumoniae by sucrose fermentation test. In addition, deficiency of Crr reduced the biofilm formation in K. pneumoniae. Our proposed model suggests that, through the action of Crr in the absence of sucrose, the transcription of the mrk operon, which produces type 3 fimbriae, was increased, thereby influencing biofilm formation by K. pneumoniae and bacterial number in the gut of nematode. This observation differs from the regulation of polysaccharide and biofilm by sucrose in other bacteria. Our findings extend the understanding of the effects of sucrose on biofilm formation.
Collapse
Affiliation(s)
- Yu-Tze Horng
- Department of Laboratory Medicine and Biotechnology, College of Medicine, Tzu Chi University, Hualien, 970374, Taiwan
| | - Chih-Ching Chien
- Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Zhongli Dist., Taoyuan City, 320315, Taiwan
| | - Novaria Sari Dewi Panjaitan
- Department of Laboratory Medicine and Biotechnology, College of Medicine, Tzu Chi University, Hualien, 970374, Taiwan
- Center for Biomedical Research, Research Organization for Health, National Research and Innovation Agency (BRIN), Cibinong Science Center. Jl. Raya Jakarta-Bogor Km. 46, Cibinong, Bogor, West Java, 16911, Indonesia
| | - Shih-Wen Tseng
- Department of Laboratory Medicine and Biotechnology, College of Medicine, Tzu Chi University, Hualien, 970374, Taiwan
| | - Hsueh-Wen Chen
- Department of Laboratory Medicine and Biotechnology, College of Medicine, Tzu Chi University, Hualien, 970374, Taiwan
| | - Hung-Chi Yang
- Department of Medical Laboratory Science and Biotechnology, Yuanpei University of Medical Technology, Hsinchu, 30015, Taiwan
| | - Yih-Yuan Chen
- Department of Biochemical Science and Technology, National Chiayi University, Chiayi, 60004, Taiwan
| | - Po-Chi Soo
- Department of Laboratory Medicine and Biotechnology, College of Medicine, Tzu Chi University, Hualien, 970374, Taiwan
| |
Collapse
|
4
|
Ourtirane R, Connil N, Bendali F. Evaluation of the virulence of enterococcal strains isolated from high touch surfaces in a hospital in Bejaia (Algeria). Microb Pathog 2025; 205:107736. [PMID: 40419202 DOI: 10.1016/j.micpath.2025.107736] [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: 12/06/2024] [Revised: 05/19/2025] [Accepted: 05/21/2025] [Indexed: 05/28/2025]
Abstract
Biofilms are key drivers of healthcare-associated infections (HAIs). Recently, the emergence of Enterococcus strains as major contributors to HAIs has been linked to their high antibiotic resistance and biofilm formation capabilities. In this study, we report for the first time the isolation and characterisation of Enterococcus strains (5 E. faecalis, 2 E. faecium and 1 E. hirae), from hospital surfaces in Algeria. We evaluated their biofilm-forming ability under different culture conditions, cytotoxicity on Caco-2/TC7 epithelial cells, antibiotic resistance profiles and the presence of key virulence genes (agg, gelE, vanA, vanB). Results showed that the biofilm formation capacity was enhanced in the presence of sucrose and all strains were slime-positive. They were non-hemolytic but cytotoxic on epithelial cells; 75 % and 62.5 % of the strains harbored the gelE and agg genes respectively. No strain carried vanA and vanB genes. Seven strains were resistant to cefotaxime and penicillin G, and all of them were resistant to ciprofloxacin but sensitive to ofloxacin. Importantly, we evaluated the anti-biofilm efficacy of benzalkonium chloride (BAK) against mature biofilms on stainless steel coupons, complete biofilm removal required BAK >1250 × MIC, highlighting the limitations of standard biocide use. This study provides one of the few comprehensive characterisations of environmental Enterococcus strains, including E. hirae, with a focus on their resistance, virulence and tolerance to biocides. Our findings highlight the importance of contamination control of inanimate surfaces and equipment in hospitals and inefficacy of commonly used biocides against biofilm-embedded cells, posing a threat for HAIs and call for improved disinfection strategies targeting biofilm-embedded bacteria.
Collapse
Affiliation(s)
- Roza Ourtirane
- Université de Bejaia, Faculté des Sciences de la Nature et de la Vie, Laboratoire de Microbiologie Appliquée, Bejaia, 06000, Algeria
| | - Nathalie Connil
- Laboratoire Communication Bactérienne et Stratégies Anti-infectieuses (CBSA), Université de Rouen, Evreux, France.
| | - Farida Bendali
- Université de Bejaia, Faculté des Sciences de la Nature et de la Vie, Laboratoire de Microbiologie Appliquée, Bejaia, 06000, Algeria.
| |
Collapse
|
5
|
Pastén SR, Quezada CP, Arellano C, Vidal RM, Escobar A, Alonso F, Villarroel J, Montero DA, Paredes MC. Antimicrobial Properties of a Novel PEGylated Copper Nanoparticle-Embedded Silicone Rubber with Potential for Use in Biomedical Applications. Polymers (Basel) 2025; 17:1404. [PMID: 40430700 PMCID: PMC12115074 DOI: 10.3390/polym17101404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 03/17/2025] [Accepted: 03/19/2025] [Indexed: 05/29/2025] Open
Abstract
Background: Healthcare-associated infections (HAIs) significantly increase morbidity, mortality, and healthcare costs. Among HAIs, catheter-associated infections are particularly prevalent due to the susceptibility of catheters to microbial contamination and biofilm formation, especially with prolonged use. Biofilms act as infection reservoirs, complicating treatment and often requiring catheter removal, thus extending hospital stays and increasing costs. Recent technological advances in catheter design have focused on integrating antifouling and antimicrobial coatings to mitigate or prevent biofilm formation. Methods: We developed COPESIL®, a novel silicone rubber embedded with PEGylated copper nanoparticles designed to reduce microbial contamination on catheter surfaces. We conducted in vitro assays to evaluate the antimicrobial and antibiofilm efficacy of COPESIL® against pathogens commonly implicated in catheter-associated urinary tract infections. Additionally, the safety profile of the material was assessed through cytotoxicity evaluations using HepG2 cells. Results: COPESIL® demonstrated substantial antimicrobial activity, reducing contamination with Escherichia coli and Klebsiella pneumoniae by >99.9% and between 93.2% and 99.8%, respectively. Biofilm formation was reduced by 5.2- to 7.9-fold for E. coli and 2.7- to 2.8-fold for K. pneumoniae compared to controls. Cytotoxicity assays suggest the material is non-toxic, with cell viability remaining above 95% after 24 h of exposure. Conclusions: The integration of PEGylated copper nanoparticles into a silicone matrix in COPESIL® represents a promising strategy to enhance the antimicrobial properties of catheters. Future studies should rigorously evaluate the long-term antimicrobial efficacy and clinical safety of COPESIL®-coated catheters, with a focus on their impact on patient outcomes and infection rates in clinical settings.
Collapse
Affiliation(s)
- Sara Ramírez Pastén
- Centro Integrativo de Biología y Química Aplicada (CIBQA), Universidad Bernardo O’Higgins, Santiago 8370993, Chile; (S.R.P.); (C.P.Q.)
| | - Carolina Paz Quezada
- Centro Integrativo de Biología y Química Aplicada (CIBQA), Universidad Bernardo O’Higgins, Santiago 8370993, Chile; (S.R.P.); (C.P.Q.)
- Departamento de Química Ambiental, Facultad de Ciencias, Universidad Católica de la Santísima Concepción, Concepción 4070129, Chile
| | - Carolina Arellano
- Programa de Microbiología y Micología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; (C.A.); (R.M.V.)
| | - Roberto M. Vidal
- Programa de Microbiología y Micología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; (C.A.); (R.M.V.)
| | - Alejandro Escobar
- Instituto de Investigación en Ciencias Odontológicas, Facultad de Odontología, Universidad de Chile, Santiago 8380544, Chile;
| | - Faustino Alonso
- Instituto de Salud Poblacional, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile;
| | - Javier Villarroel
- Unidad de Infectología, Servicio de Medicina, Hospital del Salvador, Servicio de Salud Metropolitano Oriente, Santiago 7500922, Chile;
| | - David A. Montero
- Programa de Microbiología y Micología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; (C.A.); (R.M.V.)
| | - María C. Paredes
- Carrera de Enfermería, Facultad de Salud, Sede Santiago, Universidad Santo Tomás, Santiago 8370003, Chile
| |
Collapse
|
6
|
Coppola F, Fratianni F, Bianco V, Wang Z, Pellegrini M, Coppola R, Nazzaro F. New Methodologies as Opportunities in the Study of Bacterial Biofilms, Including Food-Related Applications. Microorganisms 2025; 13:1062. [PMID: 40431235 PMCID: PMC12114119 DOI: 10.3390/microorganisms13051062] [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: 04/04/2025] [Revised: 04/19/2025] [Accepted: 04/30/2025] [Indexed: 05/29/2025] Open
Abstract
Traditional food technologies, while essential, often face limitations in sensitivity, real-time detection, and adaptability to complex biological systems such as microbial biofilms. These constraints have created a growing demand for more advanced, precise, and non-invasive tools to ensure food safety and quality. In response to these challenges, cross-disciplinary technological integration has opened new opportunities for the food industry and public health, leveraging methods originally developed in other scientific fields. Although their industrial-scale implementation is still evolving, their application in research and pilot settings has already significantly improved our ability to detect and control biofilms, thereby strengthening food safety protocols. Advanced analytical techniques, the identification of pathogenic species and their virulence markers, and the screening of "natural" antimicrobial compounds can now be conceptualized as interconnected elements within a virtual framework centered on "food" and "biofilm". In this short review, starting from the basic concepts of biofilm and associated microorganisms, we highlight a selection of emerging analytical approaches-from optical methods, microfluidics, Atomic Force Microscopy (AFM), and biospeckle techniques to molecular strategies like CRISPR, qPCR, and NGS, and the use of organoids. Initially conceived for biomedical and biotechnological applications, these tools have recently demonstrated their value in food science by enhancing our understanding of biofilm behavior and supporting the discovery of novel anti-biofilm strategies.
Collapse
Affiliation(s)
- Francesca Coppola
- Institute of Food Science, CNR-ISA, 83100 Avellino, Italy; (F.F.); (R.C.)
- Department of Agricultural Sciences, University of Naples “Federico II”, Piazza Carlo di Borbone 1, 80055 Portici, Italy
| | - Florinda Fratianni
- Institute of Food Science, CNR-ISA, 83100 Avellino, Italy; (F.F.); (R.C.)
| | - Vittorio Bianco
- Institute of Applied Sciences and Intelligent Systems “Eduardo Caianiello”, Via Campi Flegrei, 80078 Pozzuoli, Italy; (V.B.); (Z.W.)
| | - Zhe Wang
- Institute of Applied Sciences and Intelligent Systems “Eduardo Caianiello”, Via Campi Flegrei, 80078 Pozzuoli, Italy; (V.B.); (Z.W.)
- Dipartimento di Ingegneria Chimica, Dei Materiali e della Produzione Industriale, University of Napoli Federico II, Piazzale Vincenzo Tecchio 80, 80125 Napoli, Italy
| | - Michela Pellegrini
- Department of Agricultural, Food, Environmental and Animal Science, University of Udine, Via Sondrio 2/a, 33100 Udine, Italy
| | - Raffaele Coppola
- Institute of Food Science, CNR-ISA, 83100 Avellino, Italy; (F.F.); (R.C.)
- DiAAA, University of Molise, Via De Sanctis s.n.c., 86100 Campobasso, Italy
| | - Filomena Nazzaro
- Institute of Food Science, CNR-ISA, 83100 Avellino, Italy; (F.F.); (R.C.)
| |
Collapse
|
7
|
Sandu AM, Chifiriuc MC, Vrancianu CO, Cristian RE, Alistar CF, Constantin M, Paun M, Alistar A, Popa LG, Popa MI, Tantu AC, Sidoroff ME, Mihai MM, Marcu A, Popescu G, Tantu MM. Healthcare-Associated Infections: The Role of Microbial and Environmental Factors in Infection Control-A Narrative Review. Infect Dis Ther 2025; 14:933-971. [PMID: 40208412 PMCID: PMC12084486 DOI: 10.1007/s40121-025-01143-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Accepted: 03/20/2025] [Indexed: 04/11/2025] Open
Abstract
Healthcare-associated infections (HAIs), previously known as nosocomial infections, represent a significant threat to healthcare systems worldwide, prolonging patient hospital stays and the duration of antimicrobial therapy. One of the most serious consequences of HAIs is the increase in the rate of antibiotic resistance (AR) generated by the prolonged, frequent, and sometimes incorrect use of antibiotics, which leads to the selection of resistant bacteria, making treatment difficult and expensive, with direct consequences for the safety of patients and healthcare personnel. Therefore, timely and accurate diagnosis of HAIs is mandatory to develop appropriate infection prevention and control practices (IPC) and new therapeutic strategies. This review aimed to present the prevalence, risk factors, current diagnosis, including artificial intelligence (AI) and machine learning approaches, future perspectives in combating HAIs causative bacteria (phage therapy, microbiome-based interventions, and vaccination), and HAIs surveillance strategies. Also, we discussed the latest findings regarding the relationships of AR with climate change and environmental pollution in the context of the One Health approach. Phage therapy is an emerging option that can offer an alternative to ineffective antibiotic treatments for antibiotic-resistant bacteria causing HAIs. Clinical trials dealing with vaccine development for resistant bacteria have yielded conflicting results. Two promising strategies, fecal microbiota transplantation and probiotic therapy, proved highly effective against recurrent Clostridium difficile infections and have been shown to reduce HAI incidence in hospitalized patients undergoing antibiotic therapy. Artificial intelligence and machine learning systems offer promising predictive capabilities in processing large volumes of clinical, microbiological, and patient data but require robust data integration. Our paper argues that HAIs are still a global challenge, requiring stringent IPC policies, computer vision, and AI-powered tools. Despite promising avenues like integrated One Health approaches, optimized phage therapy, microbiome-based interventions, and targeted vaccine development, several knowledge gaps in clinical efficacy, standardization, and pathogen complexity remain to be answered.
Collapse
Grants
- CNFIS-FDI-2024-F-0484 INOVEX University of Bucharest
- Pillar III Ministry of Research, Innovation and Digitalization through the National Recovery and Resilience Plan (PNRR) of Romania
- Component C9/Investment no. 8 (I8) - contract CF 68 Ministry of Research, Innovation and Digitalization through the National Recovery and Resilience Plan (PNRR) of Romania
- Project No. RO1567-IBB05/2023 Institute of Biology Bucharest of the Romanian Academy
- project no. 23020101 The core program within the National Research Development and Innovation Plan, 2022-2027', carried out with the support of the Ministry of Research, Innovation and Digitalization (MCID),
- Contract no. 7N from 3 January 2023 The core program within the National Research Development and Innovation Plan, 2022-2027', carried out with the support of the Ministry of Research, Innovation and Digitalization (MCID),
- Dezvoltarea cercetării genomice în România - ROGEN" (Development of genomic research in Romania -ROGEN). ROGEN
- The core program within the National Research Development and Innovation Plan, 2022–2027’, carried out with the support of the Ministry of Research, Innovation and Digitalization (MCID),
Collapse
Affiliation(s)
- Andreea M Sandu
- Doctoral School, Carol Davila University of Medicine and Pharmacy, Eroii Sanitari 8, District 5, 050474, Bucharest, Romania
- The County Emergency Hospital, Aleea Spitalului 36, 110283, Pitești, Romania
| | - Mariana C Chifiriuc
- The Research Institute of the University of Bucharest, ICUB, Șoseaua Panduri 90, District 5, 050663, Bucharest, Romania
- Microbiology-Immunology Department, Faculty of Biology, University of Bucharest, 050095, Bucharest, Romania
- Biological Sciences Division, Romanian Academy, 125 Calea Victoriei, 010071, Bucharest, Romania
| | - Corneliu O Vrancianu
- Doctoral School, Carol Davila University of Medicine and Pharmacy, Eroii Sanitari 8, District 5, 050474, Bucharest, Romania.
- The Research Institute of the University of Bucharest, ICUB, Șoseaua Panduri 90, District 5, 050663, Bucharest, Romania.
- National Institute of Research and Development for Biological Sciences, 296 Splaiul Independentei, District 6, 060031, Bucharest, Romania.
| | - Roxana-E Cristian
- The Research Institute of the University of Bucharest, ICUB, Șoseaua Panduri 90, District 5, 050663, Bucharest, Romania
- National Institute of Research and Development for Biological Sciences, 296 Splaiul Independentei, District 6, 060031, Bucharest, Romania
| | - Cristina F Alistar
- The Research Institute of the University of Bucharest, ICUB, Șoseaua Panduri 90, District 5, 050663, Bucharest, Romania
| | - Marian Constantin
- The Research Institute of the University of Bucharest, ICUB, Șoseaua Panduri 90, District 5, 050663, Bucharest, Romania
- Institute of Biology of Romanian Academy, 296 Splaiul Independentei, District 6, 060031, Bucharest, Romania
| | - Mihaela Paun
- National Institute of Research and Development for Biological Sciences, 296 Splaiul Independentei, District 6, 060031, Bucharest, Romania
- Faculty of Administration and Business, University of Bucharest, Bucharest, Romania
| | - Alexandru Alistar
- The Research Institute of the University of Bucharest, ICUB, Șoseaua Panduri 90, District 5, 050663, Bucharest, Romania
| | - Loredana G Popa
- Faculty of Medicine, Microbiology Discipline II, Carol Davila University of Medicine and Pharmacy, 020021, Bucharest, Romania
| | - Mircea I Popa
- Faculty of Medicine, Microbiology Discipline II, Carol Davila University of Medicine and Pharmacy, 020021, Bucharest, Romania
- Preclinical Testing Unit, Cantacuzino National Military Medical Institute for Research and Development, 050096, Bucharest, Romania
| | - Ana C Tantu
- University of Medicine and Pharmacy of Craiova, Petru Rareș 2, 200349, Craiova, Romania
- Emergency Clinical County Hospital of Craiova, Tabaci 1, 200642, Craiova, Romania
| | - Manuela E Sidoroff
- National Institute of Research and Development for Biological Sciences, 296 Splaiul Independentei, District 6, 060031, Bucharest, Romania
| | - Mara M Mihai
- Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy, 020021, Bucharest, Romania
- Department of Oncologic Dermatology, "Elias" University Emergency Hospital, 010024, Bucharest, Romania
| | - Andreea Marcu
- Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy, 020021, Bucharest, Romania
| | - George Popescu
- Department of Neurosurgery 4, Bagdasar-Arseni Emergency Clinical Hospital, Şoseaua Berceni 12, 041915, Bucharest, Romania
| | - Monica M Tantu
- Department of Medical Assistance and Physical Therapy, Pitesti University Center, Târgu din Vale 1, 110040, Piteşti, Romania
- Faculty of Science, Physical Education and Informatics, National University of Science and Technology, Politehnica, Splaiul Independenţei 313, District 6, 060042, Bucharest, Romania
| |
Collapse
|
8
|
Zhang T, Liang T, Pan Q, Zhang S, Zhang S, Geng Z, Zhu B. A Universal and Versatile Zwitterionic Coating for Blood-Contacting Catheters with Long Lengths and Complex Geometries. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2502411. [PMID: 40125798 PMCID: PMC12097014 DOI: 10.1002/advs.202502411] [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] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Indexed: 03/25/2025]
Abstract
Blood-contacting catheters are highly susceptible to thrombus formation, making heparin coating essential for reducing clinical complications. However, the limitations of heparin coatings have spurred significant efforts to develop alternative strategies. This study demonstrates a cost-efficient, mechanically viable, and universal zwitterion coating approach for long and complex catheters with near-zero fouling, super anticoagulation, and selective biocapturing. Leveraging the synergistic action of side groups, a wet-adhesive initiator-bearing polymer rapidly assembles on catheter surfaces in aqueous environments, facilitating the grafting of superhydrophilic and zwitterionic polymers onto catheter inner walls. This strategy demonstrates broad adaptability, successfully applying to ten substrates and showing exceptional versatility in modifying catheters and joints of various shapes and sizes. These coatings exhibit near-zero protein fouling across a broad pH range, and superior resistance to blood cells and bacteria. Furthermore, they maintain excellent stability under simulated bloodstream without compromising anticoagulant performance. Beyond antifouling properties, this method enables the construction of highly selective bio-interaction networks on catheter inner walls, allowing precise capture of circulating tumor cells from blood. This zwitterion coating technique, with its rapid modification, robust anticoagulant properties, and customizable bio-functionality, provides an attractive solution for, beyond catheters, a wide range of medical devices that must perform in challenging biological environments.
Collapse
Affiliation(s)
- Tong Zhang
- School of Materials Science and EngineeringShanghai University99 Shangda RoadBaoshanShanghai200444China
- Shanghai Engineering Research Center of Organ RepairShanghai UniversityShanghai200444China
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ RepairMinistry of EducationShanghai UniversityShanghai200444China
| | - Tian Liang
- School of Materials Science and EngineeringShanghai University99 Shangda RoadBaoshanShanghai200444China
- Shanghai Engineering Research Center of Organ RepairShanghai UniversityShanghai200444China
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ RepairMinistry of EducationShanghai UniversityShanghai200444China
| | - Qichao Pan
- School of Materials Science and EngineeringShanghai University99 Shangda RoadBaoshanShanghai200444China
- Shanghai Engineering Research Center of Organ RepairShanghai UniversityShanghai200444China
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ RepairMinistry of EducationShanghai UniversityShanghai200444China
| | - Shouyan Zhang
- School of Materials Science and EngineeringShanghai University99 Shangda RoadBaoshanShanghai200444China
- Shanghai Engineering Research Center of Organ RepairShanghai UniversityShanghai200444China
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ RepairMinistry of EducationShanghai UniversityShanghai200444China
| | - Shuhua Zhang
- School of Materials Science and EngineeringShanghai University99 Shangda RoadBaoshanShanghai200444China
- Shanghai Engineering Research Center of Organ RepairShanghai UniversityShanghai200444China
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ RepairMinistry of EducationShanghai UniversityShanghai200444China
| | - Zhi Geng
- School of Materials Science and EngineeringShanghai University99 Shangda RoadBaoshanShanghai200444China
- Shanghai Engineering Research Center of Organ RepairShanghai UniversityShanghai200444China
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ RepairMinistry of EducationShanghai UniversityShanghai200444China
| | - Bo Zhu
- School of Materials Science and EngineeringShanghai University99 Shangda RoadBaoshanShanghai200444China
- Shanghai Engineering Research Center of Organ RepairShanghai UniversityShanghai200444China
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ RepairMinistry of EducationShanghai UniversityShanghai200444China
| |
Collapse
|
9
|
Zhai X, Liu Y, Hao X, Luo M, Gao Z, Wu J, Yang Z, Gan Y, Zhao S, Song Z, Guan J. Photothermal-Driven α-Amylase-Modified Polydopamine Pot-Like Nanomotors for Enhancing Penetration and Elimination of Drug-Resistant Biofilms. Adv Healthc Mater 2025; 14:e2403033. [PMID: 39901377 DOI: 10.1002/adhm.202403033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 01/17/2025] [Indexed: 02/05/2025]
Abstract
Biological enzyme-functionalized antibacterial nanoparticles, which can degrade biofilm and kill bacteria under mild reaction conditions, have attracted much attention for the elimination of deep-seated bacterial infections. However, the poor diffusion and penetration capabilities of recently developed biological enzyme-functionalized antibacterial nanoparticles in biofilm severely impair the eradication efficacy of deep-seated bacteria. Herein, a photothermal-driven nanomotor (denoted as APPNM) is developed for enhancing the elimination of drug-resistant biofilms and the eradication of deep-seated bacteria. The nanomotor contained a pot-like polydopamine (PDA) nanostructure and its outer surface is chemically immobilized with a layer of α-amylases. Under exposure to 808 nm near-infrared (NIR) laser irradiation, the self-propelled nanomotors, integrating the α-amylases to destroy the compact structure of biofilms, can penetrate deeply into biofilms and effectively eliminate them. Subsequently, they can accumulate on the surface of bacteria using the inherent bio-adhesion property of PDA, thereby completely eradicating deep-seated bacteria by photothermal effect. These synergistic effects enable them to exhibit superior antibiofilm effects and produce remarkable therapeutic efficacy with accelerated wound healing in vivo. With excellent biocompatibility, the as-developed nanomotors have great potential to be applied for treating biofilm-related infections.
Collapse
Affiliation(s)
- Xiangxiang Zhai
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, International School of Materials Science and Engineering, Wuhan University of Technology, Wuhan, 430070, China
| | - Yi Liu
- National Key Laboratory of Agricultural Microbiology, College of Chemistry, Huazhong Agricultural University, Wuhan, 430070, China
| | - Xiaomeng Hao
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, International School of Materials Science and Engineering, Wuhan University of Technology, Wuhan, 430070, China
| | - Ming Luo
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, International School of Materials Science and Engineering, Wuhan University of Technology, Wuhan, 430070, China
| | - Zhixue Gao
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, International School of Materials Science and Engineering, Wuhan University of Technology, Wuhan, 430070, China
| | - Jinmei Wu
- National Key Laboratory of Agricultural Microbiology, College of Chemistry, Huazhong Agricultural University, Wuhan, 430070, China
| | - Zili Yang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, International School of Materials Science and Engineering, Wuhan University of Technology, Wuhan, 430070, China
| | - Ying Gan
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, International School of Materials Science and Engineering, Wuhan University of Technology, Wuhan, 430070, China
| | - Suling Zhao
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, International School of Materials Science and Engineering, Wuhan University of Technology, Wuhan, 430070, China
| | - Zhiyong Song
- National Key Laboratory of Agricultural Microbiology, College of Chemistry, Huazhong Agricultural University, Wuhan, 430070, China
| | - Jianguo Guan
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, International School of Materials Science and Engineering, Wuhan University of Technology, Wuhan, 430070, China
| |
Collapse
|
10
|
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
|
11
|
Kemal M, Demeke G, Adugna A, Dilnessa T, Abebaw A, Esmael A. Prevalence, antimicrobial resistance profiles, and determinants of Acinetobacter baumannii and Pseudomonas aeruginosa isolates among nosocomial infection--suspected patients in the northwestern region of Ethiopia. Am J Infect Control 2025:S0196-6553(25)00102-6. [PMID: 40058598 DOI: 10.1016/j.ajic.2025.03.004] [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: 01/17/2025] [Revised: 03/01/2025] [Accepted: 03/01/2025] [Indexed: 03/26/2025]
Abstract
BACKGROUND Pseudomonas aeruginosa and Acinetobacter baumannii are common causes of nosocomial infections. Furthermore, antimicrobial resistance is frequently observed in these pathogens, posing significant challenges to treatment. OBJECTIVE This study aimed to determine the prevalence, antimicrobial resistance profiles, and determinants of A baumannii and P aeruginosa isolates among nosocomial infection--suspected patients at Debre Markos Comprehensive Specialized Hospital. METHODS An institutional-based cross-sectional study was conducted from March 01, 2021 to May 30, 2021. A consecutive convenient sampling technique was applied to select 200 nosocomial infection--suspected patients. Clinical samples were collected and inoculated on MacConkey agar, blood agar, and Tryptic Soy broth. Testing for antibiotic susceptibility was done on Mueller Hinton agar. RESULTS The overall prevalence of A baumannii and P aeruginosa isolates among nosocomial infection--suspected patients was 11.5%. All isolates of P aeruginosa and A baumannii were 100% resistant to cefepime and piperacillin. Prolonged hospitalization and antibiotics use were significant determinants of P aeruginosa and A baumannii--induced nosocomial infection (adjusted odds ratio: 5.691, 95% CI: 1.069-7.296, P<.050 and adjusted odds ratio: 4.199, 95% CI: 0.997-6.675, P<.042, respectively). CONCLUSIONS The prevalence of A baumannii and P aeruginosa isolates was high in the study area. Therefore, there should be serious attention to control the spread of drug-resistant nosocomial infections in the study area.
Collapse
Affiliation(s)
- Medina Kemal
- Medical Laboratory Science, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Gebereselassie Demeke
- Medical Laboratory Science, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Adane Adugna
- Medical Laboratory Science, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia.
| | - Tebelay Dilnessa
- Medical Laboratory Science, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Abtie Abebaw
- Medical Laboratory Science, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Ahmed Esmael
- Medical Laboratory Science, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| |
Collapse
|
12
|
Fakher S, Westenberg D. Properties and antibacterial effectiveness of metal-ion doped borate-based bioactive glasses. Future Microbiol 2025; 20:315-331. [PMID: 40079871 PMCID: PMC11938980 DOI: 10.1080/17460913.2025.2470029] [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: 01/15/2025] [Accepted: 02/18/2025] [Indexed: 03/15/2025] Open
Abstract
Bioactive glasses (BGs) are physiologically reactive surface biomaterials widely used in biomedical applications and various treatments. Borate bioactive glasses (BBGs) are third-generation BGs, and they exhibit superior biodegradable, bioactive, osteoconductive, antibacterial, and biocompatible properties compared to other types of BGs. Certain concentrations of dopant ions can be incorporated into the chemical structure of BBGs to enhance their biological functionalities and antimicrobial properties. It was demonstrated that those ions play a crucial role in the biological responsiveness in vitro and in vivo once in contact with a physiological environment. The dissolution products of ion-doped BBGs were noted in their ability to stimulate gene expression related to cell differentiation and proliferation, promote angiogenesis, display anti-inflammatory effects, and inhibit bacterial growth within a few hours. Thus, metal-ion-doped BBGs address several limitations encountered by biomedical, tissue engineering, and infection control applications. Considering the research studies on BBGs to date, this review aims to analyze metal-ion-doped BBGs based on their primary antibacterial properties and effectiveness.
Collapse
Affiliation(s)
- Sarah Fakher
- Department of Biological Sciences, Missouri University of Science and Technology, Rolla, MO, USA
| | - David Westenberg
- Department of Biological Sciences, Missouri University of Science and Technology, Rolla, MO, USA
| |
Collapse
|
13
|
Soni N, Patel T, Dhandhukia P, Thakker JN. Characterization and antibiofilm activity of carotenoids derived from marine Bacillus infantis against Staphylococcus aureus and Pseudomonas aeruginosa. INTERNATIONAL JOURNAL OF ENVIRONMENTAL HEALTH RESEARCH 2025:1-10. [PMID: 39925180 DOI: 10.1080/09603123.2025.2464082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 02/04/2025] [Indexed: 02/11/2025]
Abstract
Antibiotic resistance and biofilm pose significant challenges in healthcare, impacting economic growth and human well-being. The quest must be conducted for plausible natural anti-biofilm agents. Liquid chromatography mass spectroscopy was used for identification of carotenoids. Thebiofilm inhibition and eradication activity were evaluated against P. aeruginosa and S. aureus using crystal violet, Congo Red agar, and Scanning electron microscopy (SEM). Along with apocarotenoids, di-O-demethylspirilloxanthin, dihydroxylycopene glucoside, 2,2'-dihydroxy astaxanthin, and all-trans-Rhodovibrin were detected. Carotenoids at a concentration of 200 µg/ml showed 89.42 ± 3.42%, and 44.72 ± 6.18% biofilm inhibition of S. aureus, and P. aeruginosa, respectively, and was able to eradicate 25.68 ± 1.87% preformed biofilm in S. aureus. SEM and Congo Red Agar confirmed that carotenoids inhibited bacterial growth, exopolysaccharide production and prevented biofilm formation. Our investigation indicated that carotenoids from B. infantis could be an effective inhibitor for biofilm formed by both organisms and also had good eradication activity against S. aureus.
Collapse
Affiliation(s)
- Nidhi Soni
- Department of Biological Sciences, P.D. Patel Institute of Applied Sciences, Charotar University of Science and Technology, Changa, Anand, India
| | - Tanvi Patel
- Department of Biological Sciences, P.D. Patel Institute of Applied Sciences, Charotar University of Science and Technology, Changa, Anand, India
| | - Pinakin Dhandhukia
- Department of Microbiology, School of Science and Technology, Vanita Vishram Women's University, Surat, India
| | - Janki N Thakker
- Department of Biological Sciences, P.D. Patel Institute of Applied Sciences, Charotar University of Science and Technology, Changa, Anand, India
| |
Collapse
|
14
|
Wu X, Pan B, Chu C, Zhang Y, Ma J, Xing Y, Ma Y, Zhu W, Zhong H, Alimu A, Zhou G, Liu S, Chen W, Li X, Puyi S. CXCL16/CXCR6/TGF-β Feedback Loop Between M-MDSCs and Treg Inhibits Anti-Bacterial Immunity During Biofilm Infection. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409537. [PMID: 39716908 PMCID: PMC11831521 DOI: 10.1002/advs.202409537] [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: 08/12/2024] [Revised: 10/29/2024] [Indexed: 12/25/2024]
Abstract
Staphylococcus aureus (S. aureus) is a leading cause of Periprosthetic joint infection (PJI), a severe complication after joint arthroplasty. Immunosuppression is a major factor contributing to the infection chronicity of S. aureus PJI, posing significant treatment challenges. This study investigates the relationship between the immunosuppressive biofilm milieu and S. aureus PJI outcomes in both discovery and validation cohorts. This scRNA-seq analysis of synovium from PJI patients reveals an expansion and heightened activity of monocyte-related myeloid-derived suppressor cells (M-MDSCs) and regulatory T cells (Treg). Importantly, CXCL16 is significantly upregulated in M-MDSCs, with its corresponding CXCR6 receptor also elevated on Treg. M-MDSCs recruit Treg and enhance its activity via CXCL16-CXCR6 interactions, while Treg secretes TGF-β, inducing M-MDSCs proliferation and immunosuppressive activity. Interfering with this cross-talk in vivo using Treg-specific CXCR6 knockout PJI mouse model reduces M-MDSCs/Treg-mediated immunosuppression and alleviates bacterial burden. Immunohistochemistry and recurrence analysis show that PJI patients with CXCR6high synovium have poor prognosis. This findings highlight the critical role of CXCR6 in Treg in orchestrating an immunosuppressive microenvironment and biofilm persistence during PJI, offering potential targets for therapeutic intervention.
Collapse
Affiliation(s)
- Xiaoyu Wu
- Department of Joint SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Clinical Research Center for Orthopedic DiseasesThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Key Laboratory of Orthopaedics and TraumatologyGuangzhouGuangdong510080China
| | - Baiqi Pan
- Department of Joint SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Clinical Research Center for Orthopedic DiseasesThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Key Laboratory of Orthopaedics and TraumatologyGuangzhouGuangdong510080China
| | - Chenghan Chu
- Department of Joint SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Clinical Research Center for Orthopedic DiseasesThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Key Laboratory of Orthopaedics and TraumatologyGuangzhouGuangdong510080China
| | - Yangchun Zhang
- Department of OrthopedicsThe People's Hospital of Baoan ShenzhenShenzhenGuangdong518101China
- Department of OrthopedicsThe Second Affiliated Hospital of Shenzhen UniversityShenzhenGuangdong518101China
| | - Jinjin Ma
- Technology School of MedicineSouth China University of TechnologyGuangzhouGuangdong510640China
- Shien‐ming Wu School of Intelligent EngineeringSouth China University of TechnologyGuangzhouGuangdong510640China
| | - Yang Xing
- Department of Joint SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Clinical Research Center for Orthopedic DiseasesThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Key Laboratory of Orthopaedics and TraumatologyGuangzhouGuangdong510080China
| | - Yuanchen Ma
- Department of OrthopedicsGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouGuangdong519041China
| | - Wengang Zhu
- Department of Joint OrthopedicsYuebei People's HospitalShaoguanGuangdong512099China
| | - Huan Zhong
- Department of Joint SurgeryAffiliated Hospital of Guangdong Medical UniversityZhanjiangGuangdong524002China
| | - Aerman Alimu
- Department of Joint SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Clinical Research Center for Orthopedic DiseasesThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Key Laboratory of Orthopaedics and TraumatologyGuangzhouGuangdong510080China
| | - Guanming Zhou
- Department of OrthopedicsFoshan Hospital of Traditional Chinese MedicineGuangzhouGuangdong528051China
| | - Shuying Liu
- Department of Histology and EmbryologyZhongshan School of MedicineSun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Weishen Chen
- Department of Joint SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Clinical Research Center for Orthopedic DiseasesThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Key Laboratory of Orthopaedics and TraumatologyGuangzhouGuangdong510080China
| | - Xiang Li
- Guangdong Provincial Clinical Research Center for Orthopedic DiseasesThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Department of Spine SurgeryThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Sheng Puyi
- Department of Joint SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Clinical Research Center for Orthopedic DiseasesThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Key Laboratory of Orthopaedics and TraumatologyGuangzhouGuangdong510080China
| |
Collapse
|
15
|
Zhao R, Du B, Hu L, Li C, Xue F, Wang X, Jiang C, Wang J, Zhao Y. Distribution of sasX, mupA, and qacA/B genes and determination of genetic relatedness of epidemic methicillin-resistant Staphylococcus aureus strains associated with bloodstream infections in southern China. Front Cell Infect Microbiol 2025; 15:1491658. [PMID: 39949720 PMCID: PMC11821609 DOI: 10.3389/fcimb.2025.1491658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 01/13/2025] [Indexed: 02/16/2025] Open
Abstract
Introduction Methicillin-resistant Staphylococcus aureus (MRSA) exhibits diverse genotypes with varying virulence and resistance profiles, particularly in the context of bloodstream infections (BSI). This study investigates the prevalence of the sasX, mupA, and qacA/B genes among MRSA isolates from bloodstream infections in southern China and analyzes their genetic relatedness. Methods A polymerase chain reaction (PCR) assay was developed to detect the presence of the sasX gene, which is associated with nasal colonization, immune evasion, and virulence, the mupirocin resistance gene mupA, and the chlorhexidine tolerance gene qacA/B in a total of 77 MRSA isolates. Multilocus sequence typing (MLST) was performed to determine the sequence types (STs) and assess the genetic relatedness of the isolates. The resistance of these strains to 16 antibiotics was also analyzed. The distribution of these genes and their association with epidemic STs were analyzed. Results A total of 26 STs were identified, with notable prevalence in five epidemic clones: ST59, ST5, and ST764. The prevalence of the sasX, mupA, and qacA/B genes across all isolates was 23.4%, 33.8%, and 79.2%, respectively. Specifically, the frequency of the sasX gene was highest in ST59 (29.4%), ST239 (100%), and ST764 (37.5%); mupA was most prevalent in ST5 (66.7%), ST59 (17.6%), ST764 (37.5%), and ST15 (100%); qacA/B was predominantly found in ST59 (88.2%), ST5 (66.7%), ST398 (85.7%), ST764 (50.0%), and ST239 (100%). The gene distribution patterns revealed that sasX+ qacA/B+ mupA+ strains were closely associated with epidemic clones ST6290 and ST88, whereas sasX+ qacA/B+ mupA- strains were linked to ST59, ST239, and ST764. Discussion Notably, forty-seven (61%) MRSA BSI strains were multidrug-resistant, with the majority exhibiting resistance to penicillin, erythromycin, and clindamycin. Major MRSA clones in southern China include ST59, ST5, ST764, and ST398. In this study, sasX, mupA and qacA/B genes were present in the MRSA isolates, with the mupA gene being the most prevalent. Variations in the prevalence of virulence and resistance genes among these epidemic strains underscore the need for targeted infection control measures. These findings contribute to a better understanding of the genetic epidemiology of MRSA in the region, facilitating the development of effective prevention and control strategies for BSI.
Collapse
Affiliation(s)
- Rui Zhao
- Clinical Microbiology Laboratory, Shanghai Center for Clinical Laboratory, Shanghai, China
| | - Bingyu Du
- Laboratory Medicine Center, Second Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Lingling Hu
- Laboratory Medicine Center, Second Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Chenxi Li
- Laboratory Medicine Center, Second Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Fen Xue
- Laboratory Medicine Center, Second Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Xing Wang
- Department of Laboratory Medicine, Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Changhong Jiang
- Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jinghua Wang
- Clinical Microbiology Laboratory, Shanghai Center for Clinical Laboratory, Shanghai, China
| | - Yanfeng Zhao
- Laboratory Medicine Center, Second Affiliated Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
16
|
Zhang J, Pandit S, Rahimi S, Cao Z, Mijakovic I. Vertical graphene nanoarray decorated with Ag nanoparticles exhibits enhanced antibacterial effects. J Colloid Interface Sci 2024; 676:808-816. [PMID: 39067216 DOI: 10.1016/j.jcis.2024.07.173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/16/2024] [Accepted: 07/21/2024] [Indexed: 07/30/2024]
Abstract
Bacterial infection of biomedical implants is an important clinical challenge, driving the development of novel antimicrobial materials. The antibacterial effect of vertically aligned graphene as a nanoarray coating has been reported. In this study, vertically aligned graphene nanosheets decorated with silver nanoparticles were fabricated to enhance antibacterial effectiveness. Vertical graphene (VG) nanoflakes were synthesized by plasma-enhanced chemical vapor deposition (PECVD). Ag nanoparticles were attached to the surface of VG through using polydopamine and achieving a sustained release of Ag+. VG loaded with Ag nanoparticles (VGP/Ag) not only prevented bacterial adhesion for a long time, but also exhibited good biocompatibility. This work provides a new venue for designing antibacterial surfaces based on combination of graphene nanoarrays with other nanomaterials, and the results indicate that this approach could be very successful in preventing implant associated infections.
Collapse
Affiliation(s)
- Jian Zhang
- Systems and Synthetic Biology Division, Department of Life Sciences, Chalmers University of Technology, SE-412 96 Gothenburg, Sweden
| | - Santosh Pandit
- Systems and Synthetic Biology Division, Department of Life Sciences, Chalmers University of Technology, SE-412 96 Gothenburg, Sweden.
| | - Shadi Rahimi
- Systems and Synthetic Biology Division, Department of Life Sciences, Chalmers University of Technology, SE-412 96 Gothenburg, Sweden
| | - Zhejian Cao
- Systems and Synthetic Biology Division, Department of Life Sciences, Chalmers University of Technology, SE-412 96 Gothenburg, Sweden
| | - Ivan Mijakovic
- Systems and Synthetic Biology Division, Department of Life Sciences, Chalmers University of Technology, SE-412 96 Gothenburg, Sweden; The Novo Nordisk Foundation, Center for Biosustainability, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark.
| |
Collapse
|
17
|
Timoncini A, Lorenzetti L, Turner RJ, McGibbon A, Martini C, Cofini E, Bernardi E, Chiavari C. Inhibition of Pseudomonas aeruginosa biofilm formation on copper-based thin foils. PLoS One 2024; 19:e0314684. [PMID: 39636955 PMCID: PMC11620393 DOI: 10.1371/journal.pone.0314684] [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: 08/14/2024] [Accepted: 11/14/2024] [Indexed: 12/07/2024] Open
Abstract
The development of Healthcare-Associated Infections (HAIs) represents an increasing threat to patient health. In this context, Pseudomonas aeruginosa is responsible for various HAIs, determining about 20% of the infections in hospitalized patients, which makes it one of the most effective pathogens due to its strong ability to form biofilms. Using Cu-based materials as foils on high-touch surfaces can help to prevent and mitigate P. aeruginosa contamination in biohazardous settings. However, the antibiofilm properties of Cu-based surfaces against P. aeruginosa may vary due to frequent touches combined with indoor environmental exposure. The main aim of this study is to investigate the impact of accelerated ageing, mimicking a high-touch frequency by cyclic exposure to artificial sweat solution as well as to temperature and relative humidity variations, on the efficacy of Cu-based thin foils against P. aeruginosa biofilms. Three Cu-based materials (rolled and annealed Phosphorous High-Conductivity (PHC) Cu, Cu15Zn brass, and Cu18Ni20Zn nickel silver) were evaluated. The ageing process enhanced the antibiofilm properties, due to an increment in Cu ion release: aged PHC Cu and Cu15Zn exhibited the highest Cu ion release and hence the highest biofilm inhibition (decrease in colony forming unit (CFU)) in comparison to their pristine counterpart, while aged Cu18Ni20Zn displayed the lowest biofilm formation reduction, despite showing the highest aesthetic and morphological stability. The Cu-based surface, which highlited the highest biofilm formation inhibition due to accelerated ageing, was Cu15Zn.
Collapse
Affiliation(s)
- Andrea Timoncini
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Luca Lorenzetti
- Department of Industrial Engineering, University of Bologna, Bologna, Italy
| | - Raymond J. Turner
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
| | - Ashley McGibbon
- Department of Biomedical Sciences, McGill University, Montreal, QC, Canada
| | - Carla Martini
- Department of Industrial Engineering, University of Bologna, Bologna, Italy
| | - Elena Cofini
- Department of Cultural Heritage, University of Bologna, Ravenna, Italy
| | - Elena Bernardi
- Department of Industrial Chemistry “Toso Montanari”, University of Bologna, Bologna, Italy
| | - Cristina Chiavari
- Department of Cultural Heritage, University of Bologna, Ravenna, Italy
| |
Collapse
|
18
|
Moheb N, Mohamed AF, Elbaghdady KZ, Saeed AM, Abu-Elghait M. Monitoring and controlling bacteria in cleanrooms of pharmaceutical plant model: an in vitro study. ENVIRONMENTAL MONITORING AND ASSESSMENT 2024; 197:3. [PMID: 39621119 DOI: 10.1007/s10661-024-13445-w] [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: 11/06/2023] [Accepted: 11/16/2024] [Indexed: 01/23/2025]
Abstract
This work aims to screen the major species of bacteria distributed in the filling area in one of the new pharmaceutical facilities in the 6th of October city in Egypt and their phylogenic relationship. One hundred percent of collected Gram-positive and Gram-negative isolates of bacteria were sensitive to Levofloxacin. There were five Gram-positive multidrug-resistant (MDR) bacterial isolates and one Gram-negative (MDR) bacterial isolate (three (from personnel), two (from surface), and one (from air)). The five Gram-positive MDR bacterial isolates were resistant to Tobramycin, Gentamicin, Piperacillin, Cefaclor, and Amikacin while the one Gram-negative MDR bacterial isolate was resistant to Ceftazidime, Cefotaxime, Tobramycin, Gentamicin, Piperacillin, Cefoperazone/Sulbactam, Ofloxacin, and Polymixin b. The existence of multidrug-resistant bacteria inside cleanrooms of pharmaceutical plants signifies a life-threatening danger on human through generating contaminated drugs and/or vaccines that undoubtedly harm the consumer's healthiness. The technique of 16SrRNA gene sequencing was used to identify multidrug-resistant bacterial isolates. All tested disinfectants were bactericidal except Dettol that was found to be a bacteriostatic agent and had an anti-biofilm effect. Clorox was the most potent disinfectant that had the least MIC and MBC of 0.0002% and 0.0004%, respectively. Ethanol and Klericide were excellent sanitizing agents. The strongest biofilm formed by Staphylococcus gallinarum strain MN1812 was disrupted by Clorox with a concentration of 0.000098%. Only Dettol with a concentration of 6.3% achieved the highest disruption for the biofilm of Staphylococcus gallinarum strain NM2009. Staphylococcus gallinarum strain MN1812 followed by Bacillus amyloliquefaciens showed the highest adhesion and invasion efficiencies to Caco-cells among the investigated bacterial strains. Klericide and Dettol mixture showed more anti adhesion and invasion effects against Staphylococcus gallinarum strain NM2009 and strain MN1812 and Pseudomonas putida compared to using Klericide alone. Ethanol and Klericide had the least contact time (30 s) against most of the tested bacteria.
Collapse
Affiliation(s)
- Nahla Moheb
- Microbiology Department, Faculty of Science, Ain Shams University, Cairo, Egypt
- The Holding Company for Production of Vaccines Sera, and Drug (EGYVAC, VACSERA), Giza, Egypt
| | - Aly Fahmy Mohamed
- The International Center for Advanced Research (ICTAR), Cairo, Egypt
- The Holding Company for Production of Vaccines Sera, and Drug (EGYVAC, VACSERA), Giza, Egypt
| | | | - Ali M Saeed
- Microbiology Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Mohammed Abu-Elghait
- Department of Botany and Microbiology, Faculty of Science, Al-Azhar University, Cairo, Egypt.
| |
Collapse
|
19
|
Schlichter Kadosh Y, Muthuraman S, Nisaa K, Ben-Zvi A, Karsagi Byron DL, Shagan M, Brandis A, Mehlman T, Gopas J, Saravana Kumar R, Kushmaro A. Pseudomonas aeruginosa quorum sensing and biofilm attenuation by a di-hydroxy derivative of piperlongumine (PL-18). Biofilm 2024; 8:100215. [PMID: 39148892 PMCID: PMC11326495 DOI: 10.1016/j.bioflm.2024.100215] [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: 01/10/2024] [Revised: 07/01/2024] [Accepted: 07/13/2024] [Indexed: 08/17/2024] Open
Abstract
Bacterial communication, Quorum Sensing (QS), is a target against virulence and prevention of antibiotic-resistant infections. 16 derivatives of Piperlongumine (PL), an amide alkaloid from Piper longum L., were screened for QS inhibition. PL-18 had the best QSI activity. PL-18 inhibited the lasR-lasI, rhlR-rhlI, and pqs QS systems of Pseudomonas aeruginosa. PL-18 inhibited pyocyanin and rhamnolipids that are QS-controlled virulence elements. Iron is an essential element for pathogenicity, biofilm formation and resilience in harsh environments, its uptake was inhibited by PL-18. Pl-18 significantly reduced the biofilm biovolume including in established biofilms. PL-18-coated silicon tubes significantly inhibited biofilm formation. The transcriptome study of treated P. aeruginosa showed that PL-18 indeed reduced the expression of QS and iron homeostasis related genes, and up regulated sulfur metabolism related genes. Altogether, PL-18 inhibits QS, virulence, iron uptake, and biofilm formation. Thus, PL-18 should be further developed against bacterial infection, antibiotic resistance, and biofilm formation.
Collapse
Affiliation(s)
- Yael Schlichter Kadosh
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Ben Gurion University of the Negev, Beer Sheva, Israel
| | | | - Khairun Nisaa
- Department of Life Science, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Anat Ben-Zvi
- Department of Life Science, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Danit Lisa Karsagi Byron
- Department of Civil and Environmental Engineering, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Marilou Shagan
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Ben Gurion University of the Negev, Beer Sheva, Israel
| | - Alexander Brandis
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Tevie Mehlman
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Jacob Gopas
- Department of Microbiology, Immunology and Genetics Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheva, Israel
| | | | - Ariel Kushmaro
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Ben Gurion University of the Negev, Beer Sheva, Israel
- The Ilse Katz Center for Nanoscale Science and Technology, Ben Gurion University of the Negev, Beer Sheva, Israel
- School of Sustainability and Climate Change, Ben Gurion University of the Negev, Beer Sheva, Israel
| |
Collapse
|
20
|
Lamba S, Wang K, Lu J, Phillips ARJ, Swift S, Sarojini V. Polydopamine-Mediated Antimicrobial Lipopeptide Surface Coating for Medical Devices. ACS APPLIED BIO MATERIALS 2024; 7:7574-7584. [PMID: 39475192 DOI: 10.1021/acsabm.4c01132] [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: 11/19/2024]
Abstract
Biofilm formation on medical implants such as catheters is a major issue which needs to be addressed as it leads to severe health care associated infections. This study explored the design and synthesis of a polydopamine-lipopeptide based antimicrobial coating. The coating was used to modify the surface of Ultrathane Catheters. The lipopeptide SL1.15 with an N-terminal cysteine was covalently conjugated to the polydopamine modified catheters via a Michael addition reaction between the thiol moiety in the peptide and the aromatic ring in the polydopamine layer. The immobilization of the peptide on the polydopamine coated catheters was confirmed using water contact angle, X-ray photoelectron spectroscopy, atomic force microscopy, and scanning electron microscopy (SEM). The antimicrobial activity of the coated catheters investigated using drug resistant and clinical strains of Gram-positive (MRSA and S. aureus) and Gram-negative (E. coli, A. baumannii, and P. aeruginosa) bacteria revealed that lipopeptide immobilization inhibited >90% bacterial adhesion to the catheter surface. Additionally, biofilm assays against MRSA and E. coli revealed that the lipopeptide immobilized catheters inhibited >85% bacterial growth after 1 week incubation. Finally, the cytotoxicity profile of the catheters using the human dermal fibroblast, and the human embryonic kidney cell lines demonstrated that the polydopamine-lipopeptide coating was not toxic after 72 h incubation.
Collapse
Affiliation(s)
- Saurabh Lamba
- School of Chemical Sciences and The Centre for Green Chemical Science, University of Auckland, Auckland 1142, New Zealand
- The MacDiarmid Institute for Advanced Materials and Nanotechnology, Wellington 6012, New Zealand
| | - Kelvin Wang
- Auckland Bioengineering Institute, University of Auckland, Auckland 1142, New Zealand
| | - Jun Lu
- Auckland Bioengineering Institute, University of Auckland, Auckland 1142, New Zealand
| | - Anthony R J Phillips
- Surgical and Translational Research Centre, School of Biological Sciences, University of Auckland, Auckland 1142, New Zealand
| | - Simon Swift
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland 1142, New Zealand
| | - Vijayalekshmi Sarojini
- School of Chemical Sciences and The Centre for Green Chemical Science, University of Auckland, Auckland 1142, New Zealand
- The MacDiarmid Institute for Advanced Materials and Nanotechnology, Wellington 6012, New Zealand
| |
Collapse
|
21
|
Epelle EI, Amaeze N, Mackay WG, Yaseen M. Dry biofilms on polystyrene surfaces: the role of oxidative treatments for their mitigation. BIOFOULING 2024; 40:772-784. [PMID: 39377105 DOI: 10.1080/08927014.2024.2411389] [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: 06/13/2024] [Revised: 08/25/2024] [Accepted: 09/26/2024] [Indexed: 10/09/2024]
Abstract
Candida auris and Staphylococcus aureus are associated with a wide range of infections, as they exhibit multidrug resistance - a growing health concern. In this study, gaseous ozone, and ultraviolet-C (UVC) radiation are applied as infection control measures to inactivate dry biofilms of these organisms on polystyrene surfaces. The dosages utilised herein are 1000 and 3000 ppm.min for ozone and 2864 and 11592 mJ.cm-2 for UVC. Both organisms showed an increased sensitivity to UVC relative to ozone exposure in a bespoke decontamination chamber. While complete inactivation of both organisms (>7.5 CFU log) was realized after 60 mins of UVC application, this could not be achieved with ozonation for the same duration. However, a combined application of ozone and UVC yielded complete inactivation in only 20 mins. For both treatment methods, it was observed that dry biofilms of S. aureus were more difficult to inactivate than dry biofilms of C. auris. Compared to dry biofilms of C. auris, micrographs of wet C. auris biofilms revealed the presence of an abundance of extracellular material after treatments. Interestingly, wet biofilms were more difficult to inactivate than dry biofilms. These insights are crucial to preventing recalcitrant and recurrent infections via contact with contaminated polymeric surfaces.
Collapse
Affiliation(s)
- Emmanuel I Epelle
- Institute for Infrastructure and Environment, School of Engineering, The University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Ngozi Amaeze
- School of Health & Life Sciences, University of the West of Scotland, Blantyre, United Kingdom
| | - William G Mackay
- School of Health & Life Sciences, University of the West of Scotland, Blantyre, United Kingdom
| | - Mohammed Yaseen
- School of Computing, Engineering & Physical Sciences, University of the West of Scotland, Paisley, United Kingdom
| |
Collapse
|
22
|
Singh A, Tanwar M, Singh TP, Sharma S, Sharma P. An escape from ESKAPE pathogens: A comprehensive review on current and emerging therapeutics against antibiotic resistance. Int J Biol Macromol 2024; 279:135253. [PMID: 39244118 DOI: 10.1016/j.ijbiomac.2024.135253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/09/2024]
Abstract
The rise of antimicrobial resistance has positioned ESKAPE pathogens as a serious global health threat, primarily due to the limitations and frequent failures of current treatment options. This growing risk has spurred the scientific community to seek innovative antibiotic therapies and improved oversight strategies. This review aims to provide a comprehensive overview of the origins and resistance mechanisms of ESKAPE pathogens, while also exploring next-generation treatment strategies for these infections. In addition, it will address both traditional and novel approaches to combating antibiotic resistance, offering insights into potential new therapeutic avenues. Emerging research underscores the urgency of developing new antimicrobial agents and strategies to overcome resistance, highlighting the need for novel drug classes and combination therapies. Advances in genomic technologies and a deeper understanding of microbial pathogenesis are crucial in identifying effective treatments. Integrating precision medicine and personalized approaches could enhance therapeutic efficacy. The review also emphasizes the importance of global collaboration in surveillance and stewardship, as well as policy reforms, enhanced diagnostic tools, and public awareness initiatives, to address resistance on a worldwide scale.
Collapse
Affiliation(s)
- Anamika Singh
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Mansi Tanwar
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India
| | - T P Singh
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Sujata Sharma
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India.
| | - Pradeep Sharma
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India.
| |
Collapse
|
23
|
Igizeneza A, Bitunguhari L, Masaisa F, Hahirwa I, Uwamahoro LD, Sebatunzi O, Umugwaneza N, Pauwels I, Versporten A, Vlieghe E, Ahmed A, Ngabonziza JCS, Theunissen C. Prescription Practices and Usage of Antimicrobials in a Tertiary Teaching Hospital in Rwanda: A Call for Antimicrobial Stewardship. Antibiotics (Basel) 2024; 13:1032. [PMID: 39596727 PMCID: PMC11591426 DOI: 10.3390/antibiotics13111032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 09/02/2024] [Accepted: 09/04/2024] [Indexed: 11/29/2024] Open
Abstract
Background: Antimicrobial resistance (AMR) is a global problem that results in high morbidity and mortality, particularly in low- and middle-income countries. Inappropriate use of antimicrobials is a major driver of AMR. This study aimed to evaluate the rate and quality of antimicrobial prescription and use at the University Teaching Hospital of Kigali (CHUK), a tertiary-referral teaching hospital. Methodology: A point prevalence survey (PPS) of antimicrobial prescription was conducted using the Global PPS tool, including a healthcare-acquired infection (HAI) module. Results: On the day of the PPS, 39.3% (145/369) of inpatients were prescribed at least one antimicrobial. Out of the 259 prescribed antimicrobials, 232 (89.6%) were antibacterials, of which 151 (65.1%) belonged to the watch group of the WHO AWaRe classification. The top three antibiotics prescribed were cefotaxime (87; 37.5%), parenteral metronidazole (31; 13.4%), and meropenem (23; 9.9%). Stop or review dates for the prescribed antimicrobials were documented in 27/259 prescriptions (10.4%). Surgical prophylaxis (SP) was prescribed for longer than one day in 83.3% of 61 patients. Samples for culture were sent for 27.1% (63/232) of all the patients prescribed antibiotics. Conclusion: This PPS demonstrates multiple indicators of the poor use of antimicrobials, including the high prevalence usage of watch antibiotics and prolonged surgical prophylaxis and other poor-quality indicators. Thus, there is an urgent need for intervention to improve antimicrobial stewardship.
Collapse
Affiliation(s)
- Acsa Igizeneza
- Department of Microbiology and Parasitology, University of Rwanda, Huye P.O. Box 117, Rwanda
| | - Leopold Bitunguhari
- Department of Clinical Biology, University of Rwanda, Kigali P.O. Box 3286, Rwanda; (L.B.); (F.M.); (L.D.U.); (O.S.); (J.C.S.N.)
- Department of Internal Medicine, University Teaching Hospital of Kigali, Kigali P.O. Box 655, Rwanda
| | - Florence Masaisa
- Department of Clinical Biology, University of Rwanda, Kigali P.O. Box 3286, Rwanda; (L.B.); (F.M.); (L.D.U.); (O.S.); (J.C.S.N.)
- Department of Internal Medicine, University Teaching Hospital of Kigali, Kigali P.O. Box 655, Rwanda
| | - Innocent Hahirwa
- Department of Pharmacology and Toxicology, University of Rwanda, Kigali P.O. Box 655, Rwanda;
- Department of Pharmacy, University Teaching Hospital of Kigali, Kigali P.O. Box 655, Rwanda
| | - Lorette D. Uwamahoro
- Department of Clinical Biology, University of Rwanda, Kigali P.O. Box 3286, Rwanda; (L.B.); (F.M.); (L.D.U.); (O.S.); (J.C.S.N.)
- Department of Accident and Emergency, University Teaching Hospital of Kigali, Kigali P.O. Box 655, Rwanda
| | - Osee Sebatunzi
- Department of Clinical Biology, University of Rwanda, Kigali P.O. Box 3286, Rwanda; (L.B.); (F.M.); (L.D.U.); (O.S.); (J.C.S.N.)
- Department of Internal Medicine, University Teaching Hospital of Kigali, Kigali P.O. Box 655, Rwanda
| | - Nathalie Umugwaneza
- Department of Surgery, University Teaching Hospital of Kigali, Kigali P.O. Box 655, Rwanda;
| | - Ines Pauwels
- Laboratory of Medical Microbiology, Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium; (I.P.); (A.V.); (E.V.)
| | - Ann Versporten
- Laboratory of Medical Microbiology, Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium; (I.P.); (A.V.); (E.V.)
| | - Erika Vlieghe
- Laboratory of Medical Microbiology, Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium; (I.P.); (A.V.); (E.V.)
- Department of General Internal Medicine, Infectious and Tropical Diseases, Antwerp University Hospital, 2650 Edegem, Belgium
- Faculty of Medicine and Health Sciences, University of Antwerp, 2000 Antwerp, Belgium
| | - Ayman Ahmed
- Unit of Applied Medical Sciences, Faculty of Medical Laboratory Sciences, University of Khartoum, Khartoum 11111, Sudan;
- Rwanda Biomedical Centre, Kigali P.O. Box 7162, Rwanda
| | - Jean Claude S. Ngabonziza
- Department of Clinical Biology, University of Rwanda, Kigali P.O. Box 3286, Rwanda; (L.B.); (F.M.); (L.D.U.); (O.S.); (J.C.S.N.)
- Research, Innovation and Data Science Division, Rwanda Biomedical Centre, Kigali P.O. Box 7162, Rwanda
- Department of Biomedical Sciences, Institute of Tropical Medicine, 2000 Antwerp, Belgium
| | - Caroline Theunissen
- Department of Clinical Sciences, Institute of Tropical Medicine, 2000 Antwerp, Belgium;
| |
Collapse
|
24
|
Xu Y, Xiao Y, Zhao H, Wang B, Yu J, Shang Y, Zhou Y, Wu X, Guo Y, Yu F. Phenotypic and genetic characterization of daptomycin non-susceptible Staphylococcus aureus strains selected by adaptive laboratory evolution. Front Cell Infect Microbiol 2024; 14:1453233. [PMID: 39512591 PMCID: PMC11540788 DOI: 10.3389/fcimb.2024.1453233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 09/30/2024] [Indexed: 11/15/2024] Open
Abstract
Background Daptomycin non-susceptible Staphylococcus aureus (DNS) strains pose a serious clinical threat, yet their characteristics remain poorly understood. Methods DNS derivatives were generated by exposing S. aureus strains to subinhibitory concentrations of daptomycin. Competition experiment and growth kinetics experiment were used to observe the growth of bacteria. Galleria mellonella larvae and mouse skin abscess models were used to observe the virulence of bacteria. Transmission electron microscopy (TEM), cytochrome C experiment and biofilm formation experiment were used to observe the drug resistance phenotype. And homologous recombination was used to study the role of mutations. Results Phenotypic profiling of DNS strains revealed impaired growth, increased cell wall thickness, enhanced biofilm formation, reduced negative surface charge, and attenuated virulence compared to their wild-type strains. Whole genome sequencing identified mutations in mprF, cls2, and saeR in DNS strains. Allelic replacement experiments validated the roles of MprF L341F and Cls2 F60S substitutions in augmenting daptomycin non-susceptibility in Newman. Deletion of saeR in the NewmanMprFL341F strain and complementation of saeR in the Newman-DNS strain did not directly alter daptomycin susceptibility. However, the deletion of saeR was found to enhance competitive fitness under daptomycin pressure. Conclusion This work validates adaptive laboratory evolution (ALE) for modeling clinical DNS strains and uncovers contributions of mprF, cls2, and saeR mutations to the adaptation and resistance mechanisms of S. aureus against daptomycin. These findings enrich our understanding of how S. aureus acquired resistance to daptomycin, thus paving the way for the development of more effective treatment strategies and offering potential molecular markers for resistance surveillance.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Yinjuan Guo
- Department of Clinical Laboratory Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Fangyou Yu
- Department of Clinical Laboratory Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
25
|
Khan SA, Rahman ZU, Cai Z, Jiang O, Xu G. Drug-eluting ureteral stents: An overview. J Drug Deliv Sci Technol 2024; 100:106039. [DOI: 10.1016/j.jddst.2024.106039] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
|
26
|
Liu T, Li M, Tang L, Wang B, Li T, Huang Y, Xu Y, Li Y. Epidemiological, clinical and microbiological characteristics of patients with biliary tract diseases with positive bile culture in a tertiary hospital. BMC Infect Dis 2024; 24:1010. [PMID: 39300331 DOI: 10.1186/s12879-024-09799-8] [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: 02/29/2024] [Accepted: 08/22/2024] [Indexed: 09/22/2024] Open
Abstract
PURPOSE The prevalence of biliary tract diseases, which are common gastrointestinal disorders, is steadily rising. If it progresses to sepsis or septic shock, it can endanger the patient's life. Therefore, it is crucial to promptly diagnose bacterial infection in individuals suffering from biliary diseases and comprehend the risk factors associated with infection. The objective of this study was to examine the types of bacteria present in the bile of patients with biliary tract diseases, assess any alterations in their susceptibility to antimicrobial agents, and identify the risk factors contributing to the development of infection in these patients. PATIENTS AND METHODS From June 2019 to November 2022, 317 patients of biliary tract diseases with positive bile culture were included in this hospital-based descriptive analysis. The hospital's computerized medical records were used to collect data on demographic information (including gender, age, and occupation), laboratory, and clinical findings, physical examination results, comorbidities, basic diseases, treatment history, complications, and in-hospital outcomes. The study followed the Strengthening the Reporting of Observational Studies in Epidemiology (STROBE) principles. RESULTS Of the 317 patients with positive biliary tract diseases, 247 had benign diseases and 70 had malignant diseases. Patients with benign disease experienced a higher prevalence of statistically significant symptoms such as abdominal pain (81.4% vs. 57.1%, P = 0.000), nausea (31.2% vs. 14.3%, P = 0.005), vomiting (30.0% vs. 12.9%, P = 0.004), and chills (10.9% vs. 2.9%, P = 0.039), while jaundice (12.6% vs. 37.1%, P = 0.000) was more common in patients with malignant disease. At the species level, Escherichia coli (105; 40.5%), Klebsiella pneumoniae (41; 15.8%), and Pseudomonas aeruginosa (30; 11.6%) were the most commonly found Gram-negative bacterial strains in biliary tract infection. Escherichia coli, Klebsiella pneumoniae, and Pseudomonas aeruginosa were most susceptible to tigecycline, ertapenem and ceftazidime/avibactam, respectively. CONCLUSION Gram-negative bacteria are the most commonly isolated biliary bacteria. Clinical doctors should pay attention to patients with malignant diseases with low hemoglobin, high total bilirubin and high alkaline phosphatase. Carbapenems, tigecycline, and minocycline are the recommended antibiotics for Enterobacteriaceae. In recent years, the proportion of enterococcus has gradually increased, and clinical attention should be paid to enterococcus infection. Linezolid and vancomycin were recommended for the treatment of Enterococci infections. Overall, this work can provide reference for clinical diagnosis, treatment and effective interventions.
Collapse
Affiliation(s)
- Tingting Liu
- Department of Clinical Laboratory, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Moyan Li
- Department of Clinical Laboratory, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ling Tang
- Department of Clinical Laboratory, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Bo Wang
- Department of Clinical Laboratory, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Tingting Li
- Department of Clinical Laboratory, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ying Huang
- Department of Clinical Laboratory, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yuanhong Xu
- Department of Clinical Laboratory, First Affiliated Hospital of Anhui Medical University, Hefei, China.
| | - Yajuan Li
- Department of Clinical Laboratory, First Affiliated Hospital of Anhui Medical University, Hefei, China.
| |
Collapse
|
27
|
Pradhan L, Hazra S, Manna S, Pal BN, Mukherjee S. Screening of Lithium Substituted Ag-TiO 2 Nanoparticle Coating for Antibiofilm Application. ACS APPLIED BIO MATERIALS 2024; 7:6101-6113. [PMID: 39121349 DOI: 10.1021/acsabm.4c00711] [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: 08/11/2024]
Abstract
Bacterial infections and biofilm growth are common mishaps associated with medical devices, and they contribute significantly to ill health and mortality. Removal of bacterial deposition from these devices is a major challenge, resulting in an immediate necessity for developing antibacterial coatings on the surfaces of medical implants. In this context, we developed an innovative coating strategy that can operate at low temperatures (80 °C) and preserve the devices' integrity and functionality. An innovative Ag-TiO2 based coating was developed by ion exchange between silver nitrate (AgNO3) and lithium titanate (Li4Ti5O12) on glass substrates for different periods, ranging from 10 to 60 min. The differently coated samples were tested for their antibacterial and antibiofilm efficacy.
Collapse
Affiliation(s)
- Lipi Pradhan
- School of Biomedical Engineering, IIT (BHU), Varanasi 221005, Uttar Pradesh, India
| | - Sobhan Hazra
- School of Materials Science and Technology, Indian Institute of Technology (BHU), Varanasi 221005, Uttar Pradesh, India
| | - Sumit Manna
- School of Biomedical Engineering, IIT (BHU), Varanasi 221005, Uttar Pradesh, India
| | - Bhola Nath Pal
- School of Materials Science and Technology, Indian Institute of Technology (BHU), Varanasi 221005, Uttar Pradesh, India
| | - Sudip Mukherjee
- School of Biomedical Engineering, IIT (BHU), Varanasi 221005, Uttar Pradesh, India
| |
Collapse
|
28
|
Xia L, Chen M, Li G, An T. Can photocatalysis inhibit interspecies bacterial cooperation to quench the formation of robust complex bacterial biofilms in water environments? WATER RESEARCH 2024; 262:122137. [PMID: 39059198 DOI: 10.1016/j.watres.2024.122137] [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: 02/24/2024] [Revised: 07/14/2024] [Accepted: 07/22/2024] [Indexed: 07/28/2024]
Abstract
Bacterial biofilms pose significant a public health risk as an environmental reservoir for opportunistic aquatic bacterial pathogens. Understanding the interspecies roles of complex bacterial biofilms under different stimuli and regulatory mechanisms of stress responses is the key to controlling their dissemination. Herein, two-species mixture (TSM) biofilms (Staphylococcus aureus and Pseudomonas aeruginosa) were constructed in a flowthrough reactor. Compared with the single-species biofilms, the TSM biofilm had higher growth activity to reach maturity faster, forming a staggered community structure. Moreover, the TSM biofilm exhibited greatly improved resistance to different antibiotics (16-128 times higher), especially to those that act on protein synthesis and cell membrane integrity, when compared to single planktonic microorganisms. In the presence of stimuli, photocatalysis effectively inactivated the TSM biofilm within 10 h, a 4-fold shorter inactivation time compared to UVC irradiation. In addition, photocatalysis effectively depleted the extracellular polymers of the TSM biofilm and inhibited secretion of their interspecies quorum sensing signaling molecule autoinducer-2 (AI-2). However, the expression of AI-2 induced related virulence factors, and biofilm growth-related genes were initially up-regulated 3 - 10 fold for the TSM biofilm within the first 2 - 4 h of photocatalysis, followed by significant down-regulation. Furthermore, the addition of the AI-2 precursor 4,5-dihydroxy-2,3-pentanedione effectively delayed the photocatalytic inactivation efficiency of the TSM biofilm compared to the control. These results suggest that photocatalysis can effectively inactivate biofilms by inhibiting interspecies cooperation by quenching AI-2 in the TSM biofilm. This work sheds light on controlling biofilms in public health engineering systems.
Collapse
Affiliation(s)
- Longji Xia
- Guangdong-Hong Kong-Macao Joint Laboratory for Contaminants Exposure and Health, Guangdong Key Laboratory of Environmental Catalysis and Health Risk Control, Institute of Environmental Health and Pollution Control, Guangdong University of Technology, Guangzhou, 510006, China; Guangzhou Key Laboratory of Environmental Catalysis and Pollution Control, Guangdong Technology Research Center for Photocatalytic Technology Integration and Equipment Engineering, School of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou, 510006, China
| | - Min Chen
- Guangdong-Hong Kong-Macao Joint Laboratory for Contaminants Exposure and Health, Guangdong Key Laboratory of Environmental Catalysis and Health Risk Control, Institute of Environmental Health and Pollution Control, Guangdong University of Technology, Guangzhou, 510006, China; Guangzhou Key Laboratory of Environmental Catalysis and Pollution Control, Guangdong Technology Research Center for Photocatalytic Technology Integration and Equipment Engineering, School of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou, 510006, China
| | - Guiying Li
- Guangdong-Hong Kong-Macao Joint Laboratory for Contaminants Exposure and Health, Guangdong Key Laboratory of Environmental Catalysis and Health Risk Control, Institute of Environmental Health and Pollution Control, Guangdong University of Technology, Guangzhou, 510006, China; Guangzhou Key Laboratory of Environmental Catalysis and Pollution Control, Guangdong Technology Research Center for Photocatalytic Technology Integration and Equipment Engineering, School of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou, 510006, China.
| | - Taicheng An
- Guangdong-Hong Kong-Macao Joint Laboratory for Contaminants Exposure and Health, Guangdong Key Laboratory of Environmental Catalysis and Health Risk Control, Institute of Environmental Health and Pollution Control, Guangdong University of Technology, Guangzhou, 510006, China; Guangzhou Key Laboratory of Environmental Catalysis and Pollution Control, Guangdong Technology Research Center for Photocatalytic Technology Integration and Equipment Engineering, School of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou, 510006, China
| |
Collapse
|
29
|
Mishra A, Tabassum N, Aggarwal A, Kim YM, Khan F. Artificial Intelligence-Driven Analysis of Antimicrobial-Resistant and Biofilm-Forming Pathogens on Biotic and Abiotic Surfaces. Antibiotics (Basel) 2024; 13:788. [PMID: 39200087 PMCID: PMC11351874 DOI: 10.3390/antibiotics13080788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/14/2024] [Accepted: 08/19/2024] [Indexed: 09/01/2024] Open
Abstract
The growing threat of antimicrobial-resistant (AMR) pathogens to human health worldwide emphasizes the need for more effective infection control strategies. Bacterial and fungal biofilms pose a major challenge in treating AMR pathogen infections. Biofilms are formed by pathogenic microbes encased in extracellular polymeric substances to confer protection from antimicrobials and the host immune system. Biofilms also promote the growth of antibiotic-resistant mutants and latent persister cells and thus complicate therapeutic approaches. Biofilms are ubiquitous and cause serious health risks due to their ability to colonize various surfaces, including human tissues, medical devices, and food-processing equipment. Detection and characterization of biofilms are crucial for prompt intervention and infection control. To this end, traditional approaches are often effective, yet they fail to identify the microbial species inside biofilms. Recent advances in artificial intelligence (AI) have provided new avenues to improve biofilm identification. Machine-learning algorithms and image-processing techniques have shown promise for the accurate and efficient detection of biofilm-forming microorganisms on biotic and abiotic surfaces. These advancements have the potential to transform biofilm research and clinical practice by allowing faster diagnosis and more tailored therapy. This comprehensive review focuses on the application of AI techniques for the identification of biofilm-forming pathogens in various industries, including healthcare, food safety, and agriculture. The review discusses the existing approaches, challenges, and potential applications of AI in biofilm research, with a particular focus on the role of AI in improving diagnostic capacities and guiding preventative actions. The synthesis of the current knowledge and future directions, as described in this review, will guide future research and development efforts in combating biofilm-associated infections.
Collapse
Affiliation(s)
- Akanksha Mishra
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144001, Punjab, India;
| | - Nazia Tabassum
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea; (N.T.); (Y.-M.K.)
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea
| | - Ashish Aggarwal
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144001, Punjab, India;
| | - Young-Mog Kim
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea; (N.T.); (Y.-M.K.)
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea
- Department of Food Science and Technology, Pukyong National University, Busan 48513, Republic of Korea
| | - Fazlurrahman Khan
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea; (N.T.); (Y.-M.K.)
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea
- Institute of Fisheries Science, Pukyong National University, Busan 48513, Republic of Korea
- International Graduate Program of Fisheries Science, Pukyong National University, Busan 48513, Republic of Korea
| |
Collapse
|
30
|
Marzullo P, Gruttadauria M, D’Anna F. Quaternary Ammonium Salts-Based Materials: A Review on Environmental Toxicity, Anti-Fouling Mechanisms and Applications in Marine and Water Treatment Industries. Biomolecules 2024; 14:957. [PMID: 39199346 PMCID: PMC11352365 DOI: 10.3390/biom14080957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/03/2024] [Accepted: 08/06/2024] [Indexed: 09/01/2024] Open
Abstract
The adherence of pathogenic microorganisms to surfaces and their association to form antibiotic-resistant biofilms threatens public health and affects several industrial sectors with significant economic losses. For this reason, the medical, pharmaceutical and materials science communities are exploring more effective anti-fouling approaches. This review focuses on the anti-fouling properties, structure-activity relationships and environmental toxicity of quaternary ammonium salts (QAS) and, as a subclass, ionic liquid compounds. Greener alternatives such as QAS-based antimicrobial polymers with biocide release, non-fouling (i.e., PEG, zwitterions), fouling release (i.e., poly(dimethylsiloxanes), fluorocarbon) and contact killing properties are highlighted. We also report on dual-functional polymers and stimuli-responsive materials. Given the economic and environmental impacts of biofilms in submerged surfaces, we emphasize the importance of less explored QAS-based anti-fouling approaches in the marine industry and in developing efficient membranes for water treatment systems.
Collapse
Affiliation(s)
- Paola Marzullo
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, 90128 Palermo, Italy; (M.G.); (F.D.)
- Sustainable Mobility Center (Centro Nazionale per la Mobilità Sostenibile—CNMS), Via Durando 39, 20158 Milano, Italy
| | - Michelangelo Gruttadauria
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, 90128 Palermo, Italy; (M.G.); (F.D.)
- Sustainable Mobility Center (Centro Nazionale per la Mobilità Sostenibile—CNMS), Via Durando 39, 20158 Milano, Italy
| | - Francesca D’Anna
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, 90128 Palermo, Italy; (M.G.); (F.D.)
- Sustainable Mobility Center (Centro Nazionale per la Mobilità Sostenibile—CNMS), Via Durando 39, 20158 Milano, Italy
| |
Collapse
|
31
|
Jeong GJ, Khan F, Tabassum N, Cho KJ, Kim YM. Strategies for controlling polymicrobial biofilms: A focus on antibiofilm agents. Int J Antimicrob Agents 2024; 64:107243. [PMID: 38908533 DOI: 10.1016/j.ijantimicag.2024.107243] [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: 03/13/2024] [Revised: 04/29/2024] [Accepted: 06/13/2024] [Indexed: 06/24/2024]
Abstract
Polymicrobial biofilms are among the leading causes of antimicrobial treatment failure. In these biofilms, bacterial and fungal pathogens interact synergistically at the interspecies, intraspecies, and interkingdom levels. Consequently, combating polymicrobial biofilms is substantially more difficult compared to single-species biofilms due to their distinct properties and the resulting potential variation in antimicrobial drug efficiency. In recent years, there has been an increased focus on developing alternative strategies for controlling polymicrobial biofilms formed by bacterial and fungal pathogens. Current approaches for controlling polymicrobial biofilms include monotherapy (using either natural or synthetic compounds), combination treatments, and nanomaterials. Here, a comprehensive review of different types of polymicrobial interactions between pathogenic bacterial species or bacteria and fungi is provided along with a discussion of their relevance. The mechanisms of action of individual compounds, combination treatments, and nanomaterials against polymicrobial biofilms are thoroughly explored. This review provides various future perspectives that can advance the strategies used to control polymicrobial biofilms and their likely modes of action. Since the majority of research on combating polymicrobial biofilms has been conducted in vitro, it would be an essential step in performing in vivo tests to determine the clinical effectiveness of different treatments against polymicrobial biofilms.
Collapse
Affiliation(s)
- Geum-Jae Jeong
- Department of Food Science and Technology, Pukyong National University, Busan, 48513, Republic of Korea; Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, 48513, Republic of Korea
| | - Fazlurrahman Khan
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, 48513, Republic of Korea; Institute of Fisheries Science, Pukyong National University, Busan, 48513, Republic of Korea; International Graduate Program of Fisheries Science, Pukyong National University, Busan, 48513, Republic of Korea.
| | - Nazia Tabassum
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, 48513, Republic of Korea
| | - Kyung-Jin Cho
- Department of Food Science and Technology, Pukyong National University, Busan, 48513, Republic of Korea; Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, 48513, Republic of Korea
| | - Young-Mog Kim
- Department of Food Science and Technology, Pukyong National University, Busan, 48513, Republic of Korea; Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, 48513, Republic of Korea.
| |
Collapse
|
32
|
Maake R, Achilonu I. Expression, Purification and Biophysical Characterisation of Klebsiella Pneumoniae Protein Adenylyltransferase: A Systematic Integration of Empirical and Computational Modelling Approaches. Protein J 2024; 43:751-770. [PMID: 38981945 PMCID: PMC11345332 DOI: 10.1007/s10930-024-10210-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/25/2024] [Indexed: 07/11/2024]
Abstract
Infections that are acquired due to a prolonged hospital stay and manifest 2 days following the admission of a patient to a health-care institution can be classified as hospital-acquired infections. Klebsiella pneumoniae (K. pneumoniae) has become a critical pathogen, posing serious concern globally due to the rising incidences of hypervirulent and carbapenem-resistant strains. Glutaredoxin is a redox protein that protects cells from oxidative stress as it associates with glutathione to reduce mixed disulfides. Protein adenylyltransferase (PrAT) is a pseudokinase with a proposed mechanism of transferring an AMP group from ATP to glutaredoxin. Inducing oxidative stress to the bacterium by inhibiting the activity of PrAT is a promising approach to combating its contribution to hospital-acquired infections. Thus, this study aims to overexpress, purify, and analyse the effects of ATP and Mg2+ binding to Klebsiella pneumoniae PrAT (KpPrAT). The pET expression system and nickel affinity chromatography were effective in expressing and purifying KpPrAT. Far-UV CD spectroscopy demonstrates that the protein is predominantly α-helical, even in the presence of Mg2+. Extrinsic fluorescence spectroscopy with ANS indicates the presence of a hydrophobic pocket in the presence of ATP and Mg2+, while mant-ATP studies allude to the potential nucleotide binding ability of KpPrAT. The presence of Mg2+ increases the thermostability of the protein. Isothermal titration calorimetry provides insight into the binding affinity and thermodynamic parameters associated with the binding of ATP to KpPrAT, with or without Mg2+. Conclusively, the presence of Mg2+ induces a conformation in KpPrAT that favours nucleotide binding.
Collapse
Affiliation(s)
- Reabetswe Maake
- Protein Structure‑Function and Research Unit, School of Molecular and Cell Biology, Faculty of Science, University of the Witwatersrand, Braamfontein, Johannesburg, 2050, South Africa
| | - Ikechukwu Achilonu
- Protein Structure‑Function and Research Unit, School of Molecular and Cell Biology, Faculty of Science, University of the Witwatersrand, Braamfontein, Johannesburg, 2050, South Africa.
| |
Collapse
|
33
|
Ansari MA, Alomary MN. Bioinspired ferromagnetic NiFe 2O 4 nanoparticles: Eradication of fungal and drug-resistant bacterial pathogens and their established biofilm. Microb Pathog 2024; 193:106729. [PMID: 38851363 DOI: 10.1016/j.micpath.2024.106729] [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: 03/16/2024] [Revised: 05/05/2024] [Accepted: 06/04/2024] [Indexed: 06/10/2024]
Abstract
Nickel ferrite nanoparticles (NiFe2O4 NPs) were synthesized using the medicinally important plant Aloe vera leaf extract, and their structural, morphological, and magnetic properties were characterized by x-ray diffraction (XRD), fourier transform infrared (FTIR), scanning electron microscopy (SEM), energy dispersive x-ray (EDX), and vibrating sample magnetometer (VSM). The synthesized NPs were soft ferromagnetic and spinel in nature, with an average particle size of 22.2 nm. To the best of our understanding, this is the first comprehensive investigation into the antibacterial, anticandidal, antibiofilm, and antihyphal properties of NiFe2O4 NPs against C. albicans as well as drug-resistant gram-positive methicillin-resistant Staphylococcus aureus (MRSA) and gram-negative multidrug resistant Pseudomonas aeruginosa (MDR-P. aeruginosa) bacteria. NiFe2O4 NPs showed potent antimicrobial activity (MIC 1.6-2 mg/mL) against the test pathogens. NiFe2O4 NPs at 0.5 mg/mL suppressed biofilm formation by 49.5-53.1 % in test pathogens. The study found that the NPs not only prevent the formation of biofilm, but also eliminate existing mature biofilms by 50.5-75.79 % at 0.5 mg/mL, which was further validated by SEM. SEM examination revealed a reduction in the number of cells that form biofilms and adhere to the surface. Additionally, it considerably impeded the colonization and aggregation of the biofilm strains on the glass surface. Light microscopic examination demonstrated that NPs effectively prevent the expansion of hyphae, filaments, and yeast-to-hyphae transformation in C. albicans, resulting in a substantial decrease in their ability to cause infection. Moreover, SEM images of the treated cells exhibited the presence of wrinkles, deformities, and impaired cell walls, which suggests an alteration and instability of the membrane. This study demonstrated the efficacy of the greenly manufactured NPs in suppressing the proliferation of candida, drug-resistant bacteria, and their preexisting biofilms, as well as yeast-to-hyphae transformation. Therefore, these NPs with broad spectrum applications could be utilized in health settings to mitigate biofilm-related health conditions caused by pathogenic microbial strains.
Collapse
Affiliation(s)
- Mohammad Azam Ansari
- Department of Epidemic Disease Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, 31441, Dammam, Saudi Arabia.
| | - Mohammad N Alomary
- Advanced Diagnostic and Therapeutic Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh, 11442, Saudi Arabia
| |
Collapse
|
34
|
Asmare Z, Erkihun M, Abebe W, Ashagre A, Misganaw T, Feleke SF. Catheter-associated urinary tract infections in Africa: Systematic review and meta-analysis. Infect Dis Health 2024; 29:172-179. [PMID: 38485529 DOI: 10.1016/j.idh.2024.02.005] [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: 11/07/2023] [Revised: 02/22/2024] [Accepted: 02/27/2024] [Indexed: 07/16/2024]
Abstract
BACKGROUND Catheter-associated urinary tract infections (CAUTIs) account for the majority of device-associated healthcare-acquired infections with significant morbidity and mortality worldwide. In developing countries with limited resources, the burden of CAUTI have substantial burden owing to the lack of well-organized infection prevention and control. Although there are studies in African countries, the magnitude of CAUTI is inconsistent. Therefore this systematic review and meta-analysis aimed to determine the pooled prevalence of CAUTI in Africa and identify the pathogens involved. METHODS Systematic review of articles from different databases and search engines such as Medline/PubMed, Google Scholar, Science Direct, and African Journal online were systematically searched to identify potential studies. Data were extracted on Microsoft Excel spreadsheet and analyzed using STATA 17.0. The pooled prevalence of CAUTI was estimated using a random effects model, inverse of variance was used to assess statistical heterogeneity across studies. Egger's tests was performed to identify possible publication bias. RESULTS This systematic review and meta-analysis incorporated twenty studies, revealing a pooled prevalence of CAUTI at 43.28%. Gram-negative bacteria were the leading cause of CAUTI accounts for 82.9%. Escherichia coli (45.06%) was the most frequent gram-negative bacterial isolate involved in CAUTI followed by Klebsiella spp (24.17%). Staphylococcus aureus was the predominant gram-positive bacterial isolate, accounting for 53.24% of gram-positive associated cases in CAUTI. CONCLUSION AND RECOMMENDATIONS In conclusion, the high prevalence of CAUTI in Africa underlines a pressing healthcare challenge. Addressing this issue requires a concerted effort, encompassing health education, infection prevention measures, resource allocation, and collaborative initiatives to enhance patient safety and mitigate the impact of CAUTI on healthcare systems in the region. As prolonged catheterization increases the risk of infection, catheters should only be used for proper indications and removed promptly when no longer needed.
Collapse
Affiliation(s)
- Zelalem Asmare
- Department of Medical Laboratory Science, College of Health Sciences, Woldia University, Woldia, Ethiopia.
| | - Mulat Erkihun
- Department of Medical Laboratory Science, College of Medicine and Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Wagaw Abebe
- Department of Medical Laboratory Science, College of Health Sciences, Woldia University, Woldia, Ethiopia
| | - Agenagnew Ashagre
- Department of Medical Laboratory Science, College of Health Sciences, Woldia University, Woldia, Ethiopia
| | - Tadesse Misganaw
- Department of Medical Laboratory Science, College of Health Sciences, Woldia University, Woldia, Ethiopia
| | - Sefineh Fenta Feleke
- Department of Public Health, College of Health Science, Woldia University, Woldia, Ethiopia
| |
Collapse
|
35
|
Fabrizio G, Sivori F, Cavallo I, Truglio M, Toma L, Sperati F, Francalancia M, Obregon F, Pamparau L, Kovacs D, Pimpinelli F, Di Domenico EG. Efficacy of sodium hypochlorite in overcoming antimicrobial resistance and eradicating biofilms in clinical pathogens from pressure ulcers. Front Microbiol 2024; 15:1432883. [PMID: 39050624 PMCID: PMC11266179 DOI: 10.3389/fmicb.2024.1432883] [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: 05/14/2024] [Accepted: 06/25/2024] [Indexed: 07/27/2024] Open
Abstract
Sodium hypochlorite (NaOCl) is widely recognized for its broad-spectrum antimicrobial efficacy in skin wound care. This study investigates the effectiveness of NaOCl against a range of bacterial and fungal isolates from pressure ulcer (PU) patients. We analyzed 20 bacterial isolates from PU patients, comprising carbapenem-resistant Klebsiella pneumoniae (CRKP), multidrug-resistant Acinetobacter baumannii (MDRAB), methicillin-resistant Staphylococcus aureus (MRSA), methicillin-susceptible Staphylococcus aureus (MSSA), along with 5 Candida albicans isolates. Antibiotic resistance profiles were determined using standard susceptibility testing. Whole-genome sequencing (WGS) was employed to identify antimicrobial resistance genes (ARGs) and disinfectant resistance genes (DRGs). Genetic determinants of biofilm formation were also assessed. The antimicrobial activity of NaOCl was evaluated by determining the minimum inhibitory concentration (MIC) and the minimal biofilm eradication concentration (MBEC) for both planktonic and biofilm-associated cells. CRKP and MDRAB showed resistance to fluoroquinolones and carbapenems, while MRSA exhibited resistance to β-lactams and levofloxacin. MSSA displayed a comparatively lower resistance profile. WGS identified significant numbers of ARGs in CRKP and MDRAB, with fewer DRGs compared to MRSA and MSSA. All isolates possessed genes associated with fimbriae production and adhesion, correlating with pronounced biofilm biomass production. NaOCl demonstrated substantial antimicrobial activity against both planktonic cells and biofilms. The MIC90 for planktonic bacterial cells was 0.125 mg/mL, and the MBEC90 ranged from 0.225 to 0.5 mg/mL. For planktonic C. albicans, the MIC90 was 0.150 mg/mL, and the MBEC90 was 0.250 mg/mL. These results highlight the challenge in treating biofilm-associated infections and underscore the potential of NaOCl as a robust antimicrobial agent against difficult-to-treat biofilm infections at concentrations lower than those typically found in commercial disinfectants.
Collapse
Affiliation(s)
- Giorgia Fabrizio
- Department of Biology and Biotechnology "C. Darwin", Sapienza University of Rome, Rome, Italy
| | - Francesca Sivori
- Microbiology and Virology, San Gallicano Dermatological Institute, IRCCS, Rome, Italy
| | - Ilaria Cavallo
- Microbiology and Virology, San Gallicano Dermatological Institute, IRCCS, Rome, Italy
| | - Mauro Truglio
- Microbiology and Virology, San Gallicano Dermatological Institute, IRCCS, Rome, Italy
| | - Luigi Toma
- Medical Directorate, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Francesca Sperati
- UOSD Clinical Trial Center, Biostatistics and Bioinformatics, IRCCS San Gallicano Dermatological Institute, Rome, Italy
| | - Massimo Francalancia
- Microbiology and Virology, San Gallicano Dermatological Institute, IRCCS, Rome, Italy
| | - Francisco Obregon
- Microbiology and Virology, San Gallicano Dermatological Institute, IRCCS, Rome, Italy
| | - Luisa Pamparau
- Microbiology and Virology, San Gallicano Dermatological Institute, IRCCS, Rome, Italy
| | - Daniela Kovacs
- Cutaneous Physiopathology, San Gallicano Dermatological Institute, IRCCS, Rome, Italy
| | - Fulvia Pimpinelli
- Microbiology and Virology, San Gallicano Dermatological Institute, IRCCS, Rome, Italy
| | - Enea Gino Di Domenico
- Department of Biology and Biotechnology "C. Darwin", Sapienza University of Rome, Rome, Italy
| |
Collapse
|
36
|
Khursheed A, Xu LC, Siedlecki CA. The effects of submicron-textured surface topography on antibiotic efficacy against biofilms. J Biomed Mater Res B Appl Biomater 2024; 112:e35436. [PMID: 38961592 PMCID: PMC11239140 DOI: 10.1002/jbm.b.35436] [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: 02/29/2024] [Revised: 04/23/2024] [Accepted: 05/14/2024] [Indexed: 07/05/2024]
Abstract
Submicron-textured surfaces have been a promising approach to mitigate biofilm development and control microbial infection. However, the use of the single surface texturing approach is still far from ideal for achieving complete control of microbial infections on implanted biomedical devices. The use of a surface topographic modification that might improve the utility of standard antibiotic therapy could alleviate the complications of biofilms on devices. In this study, we characterized the biofilms of Staphylococcus aureus and Pseudomonas aeruginosa on smooth and submicron-textured polyurethane surfaces after 1, 2, 3, and 7 days, and measured the efficacy of common antibiotics against these biofilms. Results show that the submicron-textured surfaces significantly reduced biofilm formation and growth, and that the efficacy of antibiotics against biofilms grown on textured surfaces was improved compared with smooth surfaces. The antibiotic efficacy appears to be related to the degree of biofilm development. At early time points in biofilm formation, antibiotic treatment reveals reasonably good antibiotic efficacy against biofilms on both smooth and textured surfaces, but as biofilms mature, the efficacy of antibiotics drops dramatically on smooth surfaces, with lesser decreases seen for the textured surfaces. The results demonstrate that surface texturing with submicron patterns is able to improve the use of standard antibiotic therapy to treat device-centered biofilms by slowing the development of the biofilm, thereby offering less resistance to antibiotic delivery to the bacteria within the biofilm community.
Collapse
Affiliation(s)
- Asma Khursheed
- Department of Surgery, The Pennsylvania State University, College of Medicine, Hershey, PA 17033
| | - Li-Chong Xu
- Department of Surgery, The Pennsylvania State University, College of Medicine, Hershey, PA 17033
| | - Christopher A. Siedlecki
- Department of Surgery, The Pennsylvania State University, College of Medicine, Hershey, PA 17033
- Department of Biomedical Engineering, The Pennsylvania State University, College of Medicine, Hershey, PA 17033
| |
Collapse
|
37
|
Kelly L, Jameson E. Bacteriophage cocktail shows no toxicity and improves the survival of Galleria mellonella infected with Klebsiella spp. J Virol 2024; 98:e0027224. [PMID: 38771043 PMCID: PMC11237459 DOI: 10.1128/jvi.00272-24] [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: 02/07/2024] [Accepted: 04/22/2024] [Indexed: 05/22/2024] Open
Abstract
Klebsiella spp. are causative agents of healthcare-associated infections in patients who are immunocompromised and use medical devices. The antibiotic resistance crisis has led to an increase in infections caused by these bacteria, which can develop into potentially life-threatening illnesses if not treated swiftly and effectively. Thus, new treatment options for Klebsiella are urgently required. Phage therapy can offer an alternative to ineffective antibiotic treatments for antibiotic-resistant bacteria infections. The aim of the present study was to produce a safe and effective phage cocktail treatment against Klebsiella pneumoniae and Klebsiella oxytoca, both in liquid in vitro culture and an in vivo Galleria mellonella infection model. The phage cocktail was significantly more effective at killing K. pneumoniae and K. oxytoca strains compared with monophage treatments. Preliminary phage cocktail safety was demonstrated through application in the in vivo G. mellonella model: where the phage cocktail induced no toxic side effects in G. mellonella. In addition, the phage cocktail significantly improved the survival of G. mellonella when administered as a prophylactic treatment, compared with controls. In conclusion, our phage cocktail was demonstrated to be safe and effective against Klebsiella spp. in the G. mellonella infection model. This provides a strong case for future treatment for Klebsiella infections, either as an alternative or adjunct to antibiotics.IMPORTANCEKlebsiella infections are a concern in individuals who are immunocompromised and are becoming increasingly difficult to treat with antibiotics due to their drug-resistant properties. Bacteriophage is one potential alternative therapy that could be used to tackle these infections. The present study describes the design of a non-toxic phage cocktail that improved the survival of Galleria mellonella infected with Klebsiella. This phage cocktail demonstrates potential for the safe and effective treatment of Klebsiella infections, as an adjunct or alternative to antibiotics.
Collapse
Affiliation(s)
- Lucy Kelly
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
| | - Eleanor Jameson
- School of Environmental and Natural Sciences, Bangor University, Gwynedd, United Kingdom
| |
Collapse
|
38
|
Patra D, Ghosh S, Mukherjee S, Acharya Y, Mukherjee R, Haldar J. Antimicrobial nanocomposite coatings for rapid intervention against catheter-associated urinary tract infections. NANOSCALE 2024; 16:11109-11125. [PMID: 38787647 DOI: 10.1039/d4nr00653d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2024]
Abstract
Catheter-associated urinary tract infections (CAUTIs) pose a significant challenge in hospital settings. Current solutions available on the market involve incorporating antimicrobials and antiseptics into catheters. However, challenges such as uncontrolled release leading to undesirable toxicity, as well as the prevalence of antimicrobial resistance reduce the effectiveness of these solutions. Additionally, conventional antibiotics fail to effectively eradicate entrenched bacteria and metabolically suppressed bacteria present in the biofilm, necessitating the exploration of alternative strategies. Here, we introduce a novel polymer-nanocomposite coating that imparts rapid antimicrobial and anti-biofilm properties to coated urinary catheters. We have coated silicone-based urinary catheters with an organo-soluble antimicrobial polymer nanocomposite (APN), containing hydrophobic quaternized polyethyleneimine and zinc oxide nanoparticles, in a single step coating process. The coated surfaces exhibited rapid eradication of drug-resistant bacteria within 10-15 min, including E. coli, K. pneumoniae, MRSA, and S. epidermidis, as well as drug-resistant C. albicans fungi. APN coated catheters exhibited potent bactericidal activity against uropathogenic strains of E. coli, even when incubated in human urine. Furthermore, the stability of the coating and retention of antimicrobial activity was validated even after multiple washes. More importantly, this coating deterred biofilm formation on the catheter surface, and displayed rapid inactivation of metabolically repressed stationary phase and persister cells. The ability of the coated surfaces to disrupt bacterial membranes and induce the generation of intracellular reactive oxygen species (ROS) was assessed through different techniques, such as electron microscopy imaging, flow cytometry as well as fluorescence spectroscopy and microscopy. The surface coatings were found to be biocompatible in an in vivo mice model. Our simple one-step coating approach for catheters holds significant potential owing to its ability to tackle multidrug resistant bacteria and fungi, and the challenge of biofilm formation. This work brings us one step closer to enhancing patient care and safety in hospitals.
Collapse
Affiliation(s)
- Dipanjana Patra
- Chemistry and Physics of Materials Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur, Bengaluru-560064, Karnataka, India
| | - Sreyan Ghosh
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur, Bengaluru-560064, Karnataka, India.
| | - Sudip Mukherjee
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur, Bengaluru-560064, Karnataka, India.
| | - Yash Acharya
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur, Bengaluru-560064, Karnataka, India.
| | - Riya Mukherjee
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur, Bengaluru-560064, Karnataka, India.
| | - Jayanta Haldar
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur, Bengaluru-560064, Karnataka, India.
- School of Advanced Materials, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur, Bengaluru-560064, Karnataka, India
| |
Collapse
|
39
|
Sung K, Park M, Chon J, Kweon O, Paredes A, Khan SA. Chicken Juice Enhances C. jejuni NCTC 11168 Biofilm Formation with Distinct Morphological Features and Altered Protein Expression. Foods 2024; 13:1828. [PMID: 38928770 PMCID: PMC11202532 DOI: 10.3390/foods13121828] [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: 05/24/2024] [Revised: 06/05/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Campylobacter jejuni is the foodborne pathogen causing most gastrointestinal infections. Understanding its ability to form biofilms is crucial for devising effective control strategies in food processing environments. In this study, we investigated the growth dynamics and biofilm formation of C. jejuni NCTC 11168 in various culture media, including chicken juice (CJ), brain heart infusion (BHI), and Mueller Hinton (MH) broth. Our results demonstrated that C. jejuni exhibited a higher growth rate and enhanced biofilm formation in CJ and in 1:1 mixtures of CJ with BHI or MH broth compared to these measures in BHI or MH broth alone. Electron microscopy unveiled distinct morphological attributes of late-stage biofilm cells in CJ, including the presence of elongated spiral-shaped cells, thinner stretched structures compared to regular cells, and extended thread-like structures within the biofilms. Proteomic analysis identified significant alterations in protein expression profiles in C. jejuni biofilms, with a predominance of downregulated proteins associated with vital functions like metabolism, energy production, and amino acid and protein biosynthesis. Additionally, a significant proportion of proteins linked to biofilm formation, virulence, and iron uptake were suppressed. This shift toward a predominantly coccoid morphology echoed the reduced energy demands of these biofilm communities. Our study unlocks valuable insights into C. jejuni's biofilm in CJ, demonstrating its adaptation and survival.
Collapse
Affiliation(s)
- Kidon Sung
- Division of Microbiology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, USA; (M.P.); (O.K.); (S.A.K.)
| | - Miseon Park
- Division of Microbiology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, USA; (M.P.); (O.K.); (S.A.K.)
| | - Jungwhan Chon
- Department of Companion Animal Health, Inje University, Gimhae 50834, Republic of Korea;
| | - Ohgew Kweon
- Division of Microbiology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, USA; (M.P.); (O.K.); (S.A.K.)
| | - Angel Paredes
- Office of Scientific Coordination, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, USA;
| | - Saeed A. Khan
- Division of Microbiology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, USA; (M.P.); (O.K.); (S.A.K.)
| |
Collapse
|
40
|
Song K, Hwang SJ, Jeon Y, Yoon Y. The Biomedical Applications of Biomolecule Integrated Biosensors for Cell Monitoring. Int J Mol Sci 2024; 25:6336. [PMID: 38928042 PMCID: PMC11204277 DOI: 10.3390/ijms25126336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 06/04/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
Cell monitoring is essential for understanding the physiological conditions and cell abnormalities induced by various stimuli, such as stress factors, microbial invasion, and diseases. Currently, various techniques for detecting cell abnormalities and metabolites originating from specific cells are employed to obtain information on cells in terms of human health. Although the states of cells have traditionally been accessed using instrument-based analysis, this has been replaced by various sensor systems equipped with new materials and technologies. Various sensor systems have been developed for monitoring cells by recognizing biological markers such as proteins on cell surfaces, components on plasma membranes, secreted metabolites, and DNA sequences. Sensor systems are classified into subclasses, such as chemical sensors and biosensors, based on the components used to recognize the targets. In this review, we aim to outline the fundamental principles of sensor systems used for monitoring cells, encompassing both biosensors and chemical sensors. Specifically, we focus on biosensing systems in terms of the types of sensing and signal-transducing elements and introduce recent advancements and applications of biosensors. Finally, we address the present challenges in biosensor systems and the prospects that should be considered to enhance biosensor performance. Although this review covers the application of biosensors for monitoring cells, we believe that it can provide valuable insights for researchers and general readers interested in the advancements of biosensing and its further applications in biomedical fields.
Collapse
Affiliation(s)
| | | | | | - Youngdae Yoon
- Department of Environmental Health Science, Konkuk University, Seoul 05029, Republic of Korea; (K.S.); (S.-J.H.)
| |
Collapse
|
41
|
Wang S, Courreges MC, Xu L, Gurung B, Berryman M, Gu T. Revealing roles of S-layer protein (SlpA) in Clostridioides difficile pathogenicity by generating the first slpA gene deletion mutant. Microbiol Spectr 2024; 12:e0400523. [PMID: 38709045 PMCID: PMC11237437 DOI: 10.1128/spectrum.04005-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 04/16/2024] [Indexed: 05/07/2024] Open
Abstract
Clostridioides difficile infection (CDI) with high morbidity and high mortality is an urgent threat to public health, and C. difficile pathogenesis studies are eagerly required for CDI therapy. The major surface layer protein, SlpA, was supposed to play a key role in C. difficile pathogenesis; however, a lack of isogenic slpA mutants has greatly hampered analysis of SlpA functions. In this study, the whole slpA gene was successfully deleted for the first time via CRISPR-Cas9 system. Deletion of slpA in C. difficile resulted in smaller, smother-edged colonies, shorter bacterial cell size, and aggregation in suspension. For life cycle, the mutant demonstrated lower growth (changes of optical density at 600 nm, OD600) but higher cell density (colony-forming unit, CFU), decreased toxins production, and inhibited sporulation. Moreover, the mutant was more impaired in motility, more sensitive to vancomycin and Triton X-100-induced autolysis, releasing more lactate dehydrogenase. In addition, SlpA deficiency led to robust biofilm formation but weak adhesion to human host cells.IMPORTANCEClostridioides difficile infection (CDI) has been the most common hospital-acquired infection, with a high rate of antibiotic resistance and recurrence incidences, become a debilitating public health threat. It is urgently needed to study C. difficile pathogenesis for developing efficient strategies as CDI therapy. SlpA was indicated to play a key role in C. difficile pathogenesis. However, analysis of SlpA functions was hampered due to lack of isogenic slpA mutants. Surprisingly, the first slpA deletion C. difficile strain was generated in this study via CRISPR-Cas9, further negating the previous thought about slpA being essential. Results in this study will provide direct proof for roles of SlpA in C. difficile pathogenesis, which will facilitate future investigations for new targets as vaccines, new therapeutic agents, and intervention strategies in combating CDI.
Collapse
Affiliation(s)
- Shaohua Wang
- Department of Biomedical Sciences, Ohio University Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA
- Infectious and Tropical Disease Institute, Ohio University, Athens, Ohio, USA
| | - Maria C. Courreges
- Department of Biomedical Sciences, Ohio University Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA
| | - Lingjun Xu
- Department of Chemical and Biomolecular Engineering, Institute for Corrosion and Multiphase Technology, Ohio University, Athens, Ohio, USA
| | - Bijay Gurung
- Department of Biomedical Sciences, Ohio University Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA
| | - Mark Berryman
- Department of Biomedical Sciences, Ohio University Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA
| | - Tingyue Gu
- Department of Chemical and Biomolecular Engineering, Institute for Corrosion and Multiphase Technology, Ohio University, Athens, Ohio, USA
| |
Collapse
|
42
|
Park KH, Kim D, Jung M, Kim DY, Lee YM, Lee MS, Hong KW, Bae IG, Hong SI, Cho OH. Effects of sub-inhibitory concentrations of nafcillin, vancomycin, ciprofloxacin, and rifampin on biofilm formation of clinical methicillin-resistant Staphylococcus aureus. Microbiol Spectr 2024; 12:e0341223. [PMID: 38651875 PMCID: PMC11237638 DOI: 10.1128/spectrum.03412-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 04/08/2024] [Indexed: 04/25/2024] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) infections are often difficult to treat because of their biofilm-forming ability and antimicrobial resistance. We investigated the effects of sub-minimal inhibitory concentrations (MICs) of antibiotics on MRSA biofilm formation. Clinical MRSA isolates were grown with sub-MICs (1/256-1/2 × MICs) of nafcillin, vancomycin, ciprofloxacin, and rifampin. The biofilm biomass was measured using crystal violet staining. Of the 107 MRSA isolates tested, 63 (58.9%) belonged to sequence type 5 (ST5), and 44 (41.1%) belonged to ST72. The MIC50/MIC90 values of nafcillin, vancomycin, ciprofloxacin, and rifampin were 256/512, 1/2, 64/512, and 0.008/0.03 mg/L, respectively. The sub-MICs of nafcillin, vancomycin, ciprofloxacin, and rifampin promoted biofilm formation in 75 (70.1%), 49 (45.8%), 89 (83.2%), and 89 (83.2%) isolates, respectively. At sub-MICs of nafcillin, the factors associated with strong biofilm induction were the ST5 strain (P = 0.001) and agr dysfunction (P = 0.005). For the sub-MICs of ciprofloxacin, the associated factors were the ST5 strain (P = 0.002), staphylococcal protein A type t002 strain (P < 0.001), and ciprofloxacin resistance (P < 0.001). Among the sub-MICs of rifampin, only ST5 was associated with strong biofilm induction (P = 0.006). Because the sub-MICs of rifampin were much lower than clinically relevant concentrations, we further tested the capability of biofilm induction in 0.03[Formula: see text]32 mg/L of rifampin. At these concentrations, rifampin-induced biofilm formation was rare in rifampin-susceptible MRSA [1.0% (1 of 100)] but common in rifampin-resistant MRSA [71.4% (5 of 7), P < 0.001]. Induction of biofilm biomass at sub-MICs of antibiotics is common in clinical MRSA isolates and is differentially affected by the MRSA strain and antibiotic class. IMPORTANCE Bacteria can be exposed to sub-MICs of antibiotics at the beginning and end of a dosing regimen, between doses, or during low-dose therapies. Growing evidence suggests that sub-MICs of antimicrobials can stimulate MRSA biofilm formation and alter the composition of the biofilm matrix. Pevious studies have found that sub-MICs of oxacillin, methicillin, and amoxicillin promote biofilm formation in some community-acquired MRSA (CA-MRSA). We evaluated biofilm induction by sub-MICs of four different classes of antibiotics in 44 CA-MRSA and 63 healthcare-associated MRSA (HA-MRSA) strains. Our study indicated that sub-MICs of nafcillin, vancomycin, ciprofloxacin, and rifampin frequently promote biofilm induction in clinical MRSA isolates. Strong biofilm induction in sub-MICs of nafcillin, ciprofloxacin, and rifampin was more frequent in HA-MRSA than in CA-MRSA. Antibiotic-induced biofilm formation depends on the antibiotic class, MRSA strain, and antibiotic resistance. Our results emphasize the importance of maintaining effective bactericidal concentrations of antibiotics to treat biofilm-related infections.
Collapse
Affiliation(s)
- Ki-Ho Park
- Division of Infectious Diseases, Department of Internal Medicine, Kyung Hee University College of Medicine, Kyung Hee University Hospital, Seoul, South Korea
| | - Dokyoung Kim
- Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Seoul, South Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, South Korea
- Center for Converging Humanities, Kyung Hee University, Seoul, South Korea
- Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, Graduate School, Kyung Hee University, Seoul, South Korea
| | - Minji Jung
- Division of Infectious Diseases, Department of Internal Medicine, Kyung Hee University Hospital, Seoul, South Korea
| | - Dong Youn Kim
- Division of Infectious Diseases, Department of Internal Medicine, Kyung Hee University Hospital, Seoul, South Korea
| | - Yu-Mi Lee
- Division of Infectious Diseases, Department of Internal Medicine, Kyung Hee University College of Medicine, Kyung Hee University Hospital, Seoul, South Korea
| | - Mi Suk Lee
- Division of Infectious Diseases, Department of Internal Medicine, Kyung Hee University College of Medicine, Kyung Hee University Hospital, Seoul, South Korea
| | - Kyung-Wook Hong
- Department of Internal Medicine, Gyeongsang National University Hospital, Gyeongsang National University College of Medicine, Jinju, South Korea
| | - In-Gyu Bae
- Department of Internal Medicine, Gyeongsang National University Hospital, Gyeongsang National University College of Medicine, Jinju, South Korea
| | - Sun In Hong
- Division of Infectious Diseases, Department of Internal Medicine, Soonchunhyang University Cheonan Hospital, Soonchunhyang University College of Medicine, Cheonan, South Korea
| | - Oh-Hyun Cho
- Division of Infectious Diseases, Department of Internal Medicine, Soonchunhyang University Cheonan Hospital, Soonchunhyang University College of Medicine, Cheonan, South Korea
| |
Collapse
|
43
|
Centeleghe I, Norville P, Maillard JY, Hughes L. Infection prevention control in practice: a survey of healthcare professionals' knowledge and experiences. Infect Prev Pract 2024; 6:100357. [PMID: 38854706 PMCID: PMC11156693 DOI: 10.1016/j.infpip.2024.100357] [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: 10/23/2023] [Accepted: 02/24/2024] [Indexed: 06/11/2024] Open
Abstract
Background Laboratory experiments are crucial in understanding efficacy of disinfectant products, but without compliance and appropriate application, the effectiveness of products is compromised. This study aims to understand current perceptions and knowledge of healthcare professionals (HCPs) to common cleaning and disinfection routines and microbial contamination, including biofilms, in healthcare environments. Methods An online survey, including open and closed questions, was developed. Non-probability convenience and purposive sampling were used: those currently or previously in a healthcare profession were eligible. Survey responses were taken over 24 months, including the COVID-19 pandemic. Discussion 137 participants completed the survey; over 50% were nurses. Surface cleaning frequency increased post COVID-19 from 'twice a day' to 'three/more times a day'. Disinfection frequency reduced from 'between every patient' before COVID-19 to 'twice a day' afterwards. A multimethod approach to cleaning and disinfection (70.8%) was predominant when considering the best method to deliver infection control. Most areas of clinical settings were identified as high risk (13/19). Most (87.6%) participants had heard the term 'biofilm', mainly at conference/study days (60%). 39.1% said they were aware of dry surface biofilms (DSB) in the healthcare environment. Conclusions There remain mixed views on surface cleaning and disinfection within healthcare. Education is important for understanding microbial contamination and tackling problems. More people than expected had heard the term DSB. Infection control practices seemed consistent across responses, however whether this is reality is unknown. This study provides an initial insight into current opinions/knowledge of HCPs and can form basis for further in-depth investigation.
Collapse
Affiliation(s)
- Isabella Centeleghe
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK
| | | | - Jean-Yves Maillard
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK
| | - Louise Hughes
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK
| |
Collapse
|
44
|
Kumar J, Onteru SK, Singh D. Deciphering the Drug Delivery Potential of Milk Exosome Nanovesicles for Aminobenzylpenicillin Therapeutic Efficacy against Contagious Staphylococcus Aureus in Bovine Mastitis. Adv Biol (Weinh) 2024; 8:e2300519. [PMID: 38573624 DOI: 10.1002/adbi.202300519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 01/21/2024] [Indexed: 04/05/2024]
Abstract
The emergence of antimicrobial resistance and failure of antibiotic treatment are challenging tasks for managing bovine mastitis, which is mainly caused by the contagious Staphylococcus aureus (S. aureus).To overcome these difficulties, there is an urgent need for a novel drug system. In the present study, the aim is to develop next-generation therapeutics against S. aureus by harnessing the drug delivery potential of milk nanovesicles called milk exosomes (mENs). In the present work, a drug system is developed by encapsulating aminobenzylpenicillin (AMP) in mENs (mENs-AMP). Electron microscopy and zeta-sizer results indicate that the size of mENs-AMP ranged from 55.79 ± 2.8 to 85.53 ± 7.4 nm. The AMP loading efficiency in mENs is 88.61% with its sustained release. Fluorescence spectroscopy results indicated that mENs are biocompatible with mammary epithelial cells. In vitro studies show that the antibacterial activity and the minimum inhibitory concentrations of mENs-AMP are eleven times greater and four times lower than that of unencapsulated AMP, respectively. The mENs-AMP exhibit significantly higher therapeutic efficacy than AMP at the same dosage and treatment frequency. Validation of this approach is demonstrated in mastitis-affected animals through an observation in the reduction of somatic cell counts and bacterial loads in the milk of treated animals.
Collapse
Affiliation(s)
- Jitendra Kumar
- Molecular Endocrinology, Functional Genomics & Systems Biology Laboratory, Animal Biochemistry Division, ICAR-National Dairy Research Institute, Karnal, Haryana, 132001, India
| | - Suneel Kumar Onteru
- Molecular Endocrinology, Functional Genomics & Systems Biology Laboratory, Animal Biochemistry Division, ICAR-National Dairy Research Institute, Karnal, Haryana, 132001, India
| | - Dheer Singh
- Molecular Endocrinology, Functional Genomics & Systems Biology Laboratory, Animal Biochemistry Division, ICAR-National Dairy Research Institute, Karnal, Haryana, 132001, India
| |
Collapse
|
45
|
K Karunakar K, Cheriyan BV, R K, M G, B A. "Therapeutic advancements in nanomedicine: The multifaceted roles of silver nanoparticles". BIOTECHNOLOGY NOTES (AMSTERDAM, NETHERLANDS) 2024; 5:64-79. [PMID: 39416696 PMCID: PMC11446369 DOI: 10.1016/j.biotno.2024.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/31/2024] [Accepted: 05/31/2024] [Indexed: 10/19/2024]
Abstract
Nanotechnology has the advantages of enhanced bioactivity, reduced toxicity, target specificity, and sustained release and NPs can penetrate cell membranes. The small size of silver nanoparticles, AgNPs, large surface area, and unique physicochemical properties contribute to cell lysis and increased permeability of cell membranes used in the field of biomedicine. Functional precursors integrate with phytochemicals to create distinctive therapeutic properties and the stability of the nanoparticles can be enhanced by Surface coatings and encapsulation methods, The current study explores the various synthesis methods and characterization techniques of silver nanoparticles (AgNPs) and highlights their intrinsic activity in therapeutic applications, Anti-cancer activity noted at a concentration range of 5-50 μg/ml and angiogenesis is mitigated at a dosage range of 10-50 μg/ml, Diabetes is controlled within the same concentration. Wound healing is improved at concentrations of 10-50 μg/ml and with a typical range of 10-08 μg/ml for bacteria with antimicrobial capabilities. Advancement of silver nanoparticles with a focus on the future use of AgNPs-coated wound dressings and medical devices to decrease the risk of infection. Chemotherapeutic drugs can be administered by AgNPs, which reduces adverse effects and an improvement in treatment outcomes. AgNPs have been found to improve cell proliferation and differentiation, making them beneficial for tissue engineering and regenerative medicine. Our study highlights emerging patterns and developments in the field of medicine, inferring potential future paths.
Collapse
Affiliation(s)
- Karthik K Karunakar
- Department of Pharmacy Practice, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Chennai, 602105, TN, India
| | - Binoy Varghese Cheriyan
- Department of Pharmaceutical Chemistry, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Chennai, 602105, TN, India
| | - Krithikeshvaran R
- Department of Pharmacy Practice, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Chennai, 602105, TN, India
| | - Gnanisha M
- Department of Pharmacy Practice, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Chennai, 602105, TN, India
| | - Abinavi B
- Department of Pharmacy Practice, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Chennai, 602105, TN, India
| |
Collapse
|
46
|
Kaplan JB, Horswill AR. Micrococcal nuclease regulates biofilm formation and dispersal in methicillin-resistant Staphylococcus aureus USA300. mSphere 2024; 9:e0012624. [PMID: 38695568 PMCID: PMC11237449 DOI: 10.1128/msphere.00126-24] [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: 02/15/2024] [Accepted: 03/21/2024] [Indexed: 05/14/2024] Open
Abstract
Biofilm formation is an important virulence factor for methicillin-resistant Staphylococcus aureus (MRSA). The extracellular matrix of MRSA biofilms contains significant amounts of double-stranded DNA that hold the biofilm together. MRSA cells secrete micrococcal nuclease (Nuc1), which degrades double-stranded DNA. In this study, we used standard methodologies to investigate the role of Nuc1 in MRSA biofilm formation and dispersal. We quantified biofilm formation and extracellular DNA (eDNA) levels in broth and agar cultures. In some experiments, cultures were supplemented with sub-MIC amoxicillin to induce biofilm formation. Biofilm erosion was quantitated by culturing biofilms on rods and enumerating detached colony-forming units (CFUs), and biofilm sloughing was investigated by perfusing biofilms cultured in glass tubes with fresh broth and measuring the sizes of the detached cell aggregates. We found that an MRSA nuc1- mutant strain produced significantly more biofilm and more eDNA than a wild-type strain, both in the absence and presence of sub-MIC amoxicillin. nuc1- mutant biofilms grown on rods detached significantly less than wild-type biofilms. Detachment was restored by exogenous DNase or complementing the nuc1- mutant. In the sloughing assay, nuc1- mutant biofilms released cell aggregates that were significantly larger than those released by wild-type biofilms. Our results suggest that Nuc1 modulates biofilm formation, biofilm detachment, and the sizes of detached cell aggregates. These processes may play a role in the spread and subsequent survival of MRSA biofilms during biofilm-related infections.IMPORTANCEInfections caused by antibiotic-resistant bacteria known as methicillin-resistant Staphylococcus aureus (MRSA) are a significant problem in hospitals. MRSA forms adherent biofilms on implanted medical devices such as catheters and breathing tubes. Bacteria can detach from biofilms on these devices and spread to other parts of the body such as the blood or lungs, where they can cause life-threatening infections. In this article, researchers show that MRSA secretes an enzyme known as thermonuclease that causes bacteria to detach from the biofilm. This is important because understanding the mechanism by which MRSA detaches from biofilms could lead to the development of procedures to mitigate the problem.
Collapse
Affiliation(s)
| | - Alexander R. Horswill
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
47
|
Wongchai M, Wongkaewkhiaw S, Kanthawong S, Roytrakul S, Aunpad R. Dual-function antimicrobial-antibiofilm peptide hybrid to tackle biofilm-forming Staphylococcus epidermidis. Ann Clin Microbiol Antimicrob 2024; 23:44. [PMID: 38755634 PMCID: PMC11100219 DOI: 10.1186/s12941-024-00701-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 04/28/2024] [Indexed: 05/18/2024] Open
Abstract
BACKGROUND Due to their resistance and difficulty in treatment, biofilm-associated infections are problematic among hospitalized patients globally and account for 60% of all bacterial infections in humans. Antibiofilm peptides have recently emerged as an alternative treatment since they can be effectively designed and exert a different mode of biofilm inhibition and eradication. METHODS A novel antibiofilm peptide, BiF, was designed from the conserved sequence of 18 α-helical antibiofilm peptides by template-assisted technique and its activity was improved by hybridization with a lipid binding motif (KILRR). Novel antibiofilm peptide derivatives were modified by substituting hydrophobic amino acids at positions 5 or 7, and both, with positively charged lysines (L5K, L7K). These peptide derivatives were tested for antibiofilm and antimicrobial activities against biofilm-forming Staphylococcus epidermidis and multiple other microbes using crystal violet and broth microdilution assays, respectively. To assess their impact on mammalian cells, the toxicity of peptides was determined through hemolysis and cytotoxicity assays. The stability of candidate peptide, BiF2_5K7K, was assessed in human serum and its secondary structure in bacterial membrane-like environments was analyzed using circular dichroism. The action of BiF2_5K7K on planktonic S. epidermidis and its effect on biofilm cell viability were assessed via viable counting assays. Its biofilm inhibition mechanism was investigated through confocal laser scanning microscopy and transcription analysis. Additionally, its ability to eradicate mature biofilms was examined using colony counting. Finally, a preliminary evaluation involved coating a catheter with BiF2_5K7K to assess its preventive efficacy against S. epidermidis biofilm formation on the catheter and its surrounding area. RESULTS BiF2_5K7K, the modified antibiofilm peptide, exhibited dose-dependent antibiofilm activity against S. epidermidis. It inhibited biofilm formation at subinhibitory concentrations by altering S. epidermidis extracellular polysaccharide production and quorum-sensing gene expression. Additionally, it exhibited broad-spectrum antimicrobial activity and no significant hemolysis or toxicity against mammalian cell lines was observed. Its activity is retained when exposed to human serum. In bacterial membrane-like environments, this peptide formed an α-helix amphipathic structure. Within 4 h, a reduction in the number of S. epidermidis colonies was observed, demonstrating the fast action of this peptide. As a preliminary test, a BiF2_5K7K-coated catheter was able to prevent the development of S. epidermidis biofilm both on the catheter surface and in its surrounding area. CONCLUSIONS Due to the safety and effectiveness of BiF2_5K7K, we suggest that this peptide be further developed to combat biofilm infections, particularly those of biofilm-forming S. epidermidis.
Collapse
Affiliation(s)
- Mathira Wongchai
- Graduate Program in Biomedical Sciences, Faculty of Allied Health Sciences, Thammasat University, Pathum Thani, Thailand
| | - Saharut Wongkaewkhiaw
- School of Dentistry, King Mongkut's Institute of Technology Ladkrabang, Bangkok, Thailand
| | - Sakawrat Kanthawong
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Sittiruk Roytrakul
- Functional Proteomics Technology Laboratory, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, Thailand
| | - Ratchaneewan Aunpad
- Graduate Program in Biomedical Sciences, Faculty of Allied Health Sciences, Thammasat University, Pathum Thani, Thailand.
| |
Collapse
|
48
|
Aonofriesei F. Surfactants' Interplay with Biofilm Development in Staphylococcus and Candida. Pharmaceutics 2024; 16:657. [PMID: 38794319 PMCID: PMC11125353 DOI: 10.3390/pharmaceutics16050657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/09/2024] [Accepted: 05/14/2024] [Indexed: 05/26/2024] Open
Abstract
The capacity of micro-organisms to form biofilms is a pervasive trait in the microbial realm. For pathogens, biofilm formation serves as a virulence factor facilitating successful host colonization. Simultaneously, infections stemming from biofilm-forming micro-organisms pose significant treatment challenges due to their heightened resistance to antimicrobial agents. Hence, the quest for active compounds capable of impeding microbial biofilm development stands as a pivotal pursuit in biomedical research. This study presents findings concerning the impact of three surfactants, namely, polysorbate 20 (T20), polysorbate 80 (T80), and sodium dodecyl sulfate (SDS), on the initial stage of biofilm development in both Staphylococcus aureus and Candida dubliniensis. In contrast to previous investigations, we conducted a comparative assessment of the biofilm development capacity of these two taxonomically distant groups, predicated on their shared ability to reduce TTC. The common metabolic trait shared by S. aureus and C. dubliniensis in reducing TTC to formazan facilitated a simultaneous evaluation of biofilm development under the influence of surfactants across both groups. Our results revealed that surfactants could impede the development of biofilms in both species by disrupting the initial cell attachment step. The observed effect was contingent upon the concentration and type of compound, with a higher inhibition observed in culture media supplemented with SDS. At maximum concentrations (5%), T20 and T80 significantly curtailed the formation and viability of S. aureus and C. dubliniensis biofilms. Specifically, T20 inhibited biofilm development by 75.36% in S. aureus and 71.18% in C. dubliniensis, while T80 exhibited a slightly lower inhibitory effect, with values ranging between 66.68% (C. dubliniensis) and 65.54% (S. aureus) compared to the controls. Incorporating these two non-toxic surfactants into pharmaceutical formulations could potentially enhance the inhibitory efficacy of selected antimicrobial agents, particularly in external topical applications.
Collapse
Affiliation(s)
- Florin Aonofriesei
- Department of Natural Sciences, Faculty of Natural and Agricultural Sciences, Ovidius University of Constanta, 1, University Street, 900470 Constanța, Romania
| |
Collapse
|
49
|
Malik S, Singh J, Saini K, Chaudhary V, Umar A, Ibrahim AA, Akbar S, Baskoutas S. Paper-based sensors: affordable, versatile, and emerging analyte detection platforms. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2024; 16:2777-2809. [PMID: 38639474 DOI: 10.1039/d3ay02258g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/20/2024]
Abstract
Paper-based sensors, often referred to as paper-based analytical devices (PADs), stand as a transformative technology in the field of analytical chemistry. They offer an affordable, versatile, and accessible solution for diverse analyte detection. These sensors harness the unique properties of paper substrates to provide a cost-effective and adaptable platform for rapid analyte detection, spanning chemical species, biomolecules, and pathogens. This review highlights the key attributes that make paper-based sensors an attractive choice for analyte detection. PADs demonstrate their versatility by accommodating a wide range of analytes, from ions and gases to proteins, nucleic acids, and more, with customizable designs for specific applications. Their user-friendly operation and minimal infrastructure requirements suit point-of-care diagnostics, environmental monitoring, food safety, and more. This review also explores various fabrication methods such as inkjet printing, wax printing, screen printing, dip coating, and photolithography. Incorporating nanomaterials and biorecognition elements promises even more sophisticated and sensitive applications.
Collapse
Affiliation(s)
- Sumit Malik
- Department of Chemistry, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, 133203, Haryana, India.
| | - Joginder Singh
- Department of Chemistry, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, 133203, Haryana, India.
| | - Kajal Saini
- Department of Chemistry, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, 133203, Haryana, India.
| | - Vivek Chaudhary
- Department of Chemistry, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, 133203, Haryana, India.
| | - Ahmad Umar
- Department of Chemistry, Faculty of Science and Arts, Promising Centre for Sensors and Electronic Devices (PCSED), Najran University, Najran-11001, Kingdom of Saudi Arabia.
- Department of Materials Science and Engineering, The Ohio State University, Columbus 43210, OH, USA
- STEM Pioneers Training Lab, Najran University, Najran 11001, Kingdom of Saudi Arabia
| | - Ahmed A Ibrahim
- Department of Chemistry, Faculty of Science and Arts, Promising Centre for Sensors and Electronic Devices (PCSED), Najran University, Najran-11001, Kingdom of Saudi Arabia.
- STEM Pioneers Training Lab, Najran University, Najran 11001, Kingdom of Saudi Arabia
| | - Sheikh Akbar
- Department of Materials Science and Engineering, The Ohio State University, Columbus 43210, OH, USA
| | | |
Collapse
|
50
|
Bouhrour N, Nibbering PH, Bendali F. Medical Device-Associated Biofilm Infections and Multidrug-Resistant Pathogens. Pathogens 2024; 13:393. [PMID: 38787246 PMCID: PMC11124157 DOI: 10.3390/pathogens13050393] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 04/29/2024] [Accepted: 05/04/2024] [Indexed: 05/25/2024] Open
Abstract
Medical devices such as venous catheters (VCs) and urinary catheters (UCs) are widely used in the hospital setting. However, the implantation of these devices is often accompanied by complications. About 60 to 70% of nosocomial infections (NIs) are linked to biofilms. The main complication is the ability of microorganisms to adhere to surfaces and form biofilms which protect them and help them to persist in the host. Indeed, by crossing the skin barrier, the insertion of VC inevitably allows skin flora or accidental environmental contaminants to access the underlying tissues and cause fatal complications like bloodstream infections (BSIs). In fact, 80,000 central venous catheters-BSIs (CVC-BSIs)-mainly occur in intensive care units (ICUs) with a death rate of 12 to 25%. Similarly, catheter-associated urinary tract infections (CA-UTIs) are the most commonlyhospital-acquired infections (HAIs) worldwide.These infections represent up to 40% of NIs.In this review, we present a summary of biofilm formation steps. We provide an overview of two main and important infections in clinical settings linked to medical devices, namely the catheter-asociated bloodstream infections (CA-BSIs) and catheter-associated urinary tract infections (CA-UTIs), and highlight also the most multidrug resistant bacteria implicated in these infections. Furthermore, we draw attention toseveral useful prevention strategies, and advanced antimicrobial and antifouling approaches developed to reduce bacterial colonization on catheter surfaces and the incidence of the catheter-related infections.
Collapse
Affiliation(s)
- Nesrine Bouhrour
- Laboratoire de Microbiologie Appliquée, Faculté des Sciences de la Nature et de la Vie, Université de Bejaia, Bejaia 06000, Algeria;
| | - Peter H. Nibbering
- Department of Infectious Diseases, Leiden University Medical Center, 2300 RC Leiden, The Netherlands;
| | - Farida Bendali
- Laboratoire de Microbiologie Appliquée, Faculté des Sciences de la Nature et de la Vie, Université de Bejaia, Bejaia 06000, Algeria;
| |
Collapse
|