1
|
Wu Y, Wen F, Gou S, Ran Q, Chu Y, Ma W, Zhao K. Multifaceted quorum-sensing inhibiting activity of 3-(Benzo[d][1,3]dioxol-4-yl)oxazolidin-2-one mitigates Pseudomonas aeruginosa virulence. Virulence 2025; 16:2479103. [PMID: 40104940 DOI: 10.1080/21505594.2025.2479103] [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: 11/23/2023] [Revised: 11/10/2024] [Accepted: 03/03/2025] [Indexed: 03/20/2025] Open
Abstract
As antibiotic resistance escalates into a global health crisis, novel therapeutic approaches against infectious diseases are in urgent need. Pseudomonas aeruginosa, an adaptable opportunistic pathogen, poses substantial challenges in treating a range of infections. The quorum-sensing (QS) system plays a pivotal role in orchestrating the production of a large set of virulence factors in a cell density-dependent manner, and the anti-virulence strategy targeting QS may show huge potential. Here, we present a comprehensive investigation into the potential of the synthesized compound 3-(benzo[d][1,3]dioxol-4-yl)oxazolidin-2-one (OZDO, C10H9NO4) as a QS inhibitor to curb the virulence of P. aeruginosa. By employing an integrated approach encompassing in silico screening, in vitro and in vivo functional identification, we elucidated the multifaceted effects of OZDO. Molecular docking predicted that OZDO interfered with three core regulatory proteins of P. aeruginosa QS system. Notably, OZDO exhibited significant inhibition on the production of pyocyanin, rhamnolipid and extracellular proteases, biofilm formation, and cell motilities of P. aeruginosa. Transcriptomic analysis and quantitative real-time PCR displayed the down-regulation of QS-controlled genes in OZDO-treated PAO1, reaffirming the QS-inhibition activity of OZDO. In vivo assessments using a Caenorhabditis elegans-infection model demonstrated OZDO mitigated P. aeruginosa pathogenicity, particularly against the hypervirulent strain PA14. Moreover, OZDO in combination with polymyxin B and aztreonam presented a promising avenue for innovative anti-infective therapy. Our study sheds light on the multifaceted potential of OZDO as an anti-virulence agent and its significance in combating P. aeruginosa-associated infections.
Collapse
Affiliation(s)
- Yi Wu
- Antibiotics Research and Re-Evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, Chengdu, Sichuan, China
| | - Fulong Wen
- Antibiotics Research and Re-Evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, Chengdu, Sichuan, China
| | - Shiyi Gou
- Antibiotics Research and Re-Evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, Chengdu, Sichuan, China
| | - Qiman Ran
- Antibiotics Research and Re-Evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, Chengdu, Sichuan, China
| | - Yiwen Chu
- Antibiotics Research and Re-Evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, Chengdu, Sichuan, China
| | - Wenbo Ma
- Antibiotics Research and Re-Evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, Chengdu, Sichuan, China
| | - Kelei Zhao
- Antibiotics Research and Re-Evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, Chengdu, Sichuan, China
| |
Collapse
|
2
|
Dong Y, Liu X, Xiong S, Cao M, Wu H, Chen L, Zhao M, Zheng Y, Zhang Z, Liu Y, Li Y, Qu Q, Dong C. Guanosine enhances the bactericidal effect of ceftiofur sodium on Streptococcus suis by activating bacterial metabolism. Virulence 2025; 16:2453525. [PMID: 39915976 PMCID: PMC11810099 DOI: 10.1080/21505594.2025.2453525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 12/09/2024] [Accepted: 12/29/2024] [Indexed: 02/12/2025] Open
Abstract
The emergence and rapid development of antibiotic resistance poses a serious threat to global public health. Streptococcus suis (S. suis) is an important zoonotic pathogen, and the development of its antibiotic resistance has made the infections difficult to treat. The combination of non-antibiotic compounds with antibiotics is considered a promising strategy against multidrug-resistant bacteria. However, the mechanism by which metabolites act as antibiotic adjuvant remains unclear. Here, we found that guanosine metabolism was repressed in multidrug-resistant S. suis. Exogenous guanosine promoted the antibacterial effects of ceftiofur sodium (CEF) in vitro and in vivo. Furthermore, we demonstrated that exogenous guanosine promoted the biosynthesis of purine pathway, TCA cycle and bacterial respiration, which make bacteria more sensitive to the killing effect of antibacterial. In addition, the function of the cell membrane is affected by guanosine and the accumulation of antimicrobials in the bacteria increased. Bacterial-oxidative stress and DNA damage induced by guanosine is also one of the mechanisms by which the antibacterial effect is enhanced. These results suggest that guanosine is a promising adjuvant for antibacterial drugs and provide new theoretical basis for the clinical treatment of S. suis infection.
Collapse
Affiliation(s)
- Yue Dong
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Northeast Agricultural University, Harbin, P R China
| | - Xiaona Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Northeast Agricultural University, Harbin, P R China
| | - Shanshan Xiong
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Northeast Agricultural University, Harbin, P R China
| | - Mingyu Cao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Northeast Agricultural University, Harbin, P R China
| | - Haojie Wu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Northeast Agricultural University, Harbin, P R China
| | - Long Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Northeast Agricultural University, Harbin, P R China
| | - Mengmeng Zhao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Northeast Agricultural University, Harbin, P R China
| | - Yadan Zheng
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Northeast Agricultural University, Harbin, P R China
| | - Zhiyun Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Northeast Agricultural University, Harbin, P R China
| | - Yanyan Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Northeast Agricultural University, Harbin, P R China
| | - Yanhua Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Northeast Agricultural University, Harbin, P R China
| | - Qianwei Qu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Northeast Agricultural University, Harbin, P R China
| | - Chunliu Dong
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Northeast Agricultural University, Harbin, P R China
| |
Collapse
|
3
|
Silva E, Ferreira-Santos P, Teixeira JA, Pereira MO, Rocha CM, Sousa AM. Aqueous extracts of Moringa oleifera and Cinnamomum cassia as promising sources of antibiofilm compounds against mucoid and small colony variants of Pseudomonas aeruginosa and Staphylococcus aureus. Biofilm 2025; 9:100250. [PMID: 39877233 PMCID: PMC11772965 DOI: 10.1016/j.bioflm.2024.100250] [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: 09/23/2024] [Revised: 12/20/2024] [Accepted: 12/29/2024] [Indexed: 01/31/2025] Open
Abstract
Bacterial biofilms formed by Staphylococcus aureus and Pseudomonas aeruginosa pose significant challenges in treating cystic fibrosis (CF) airway infections due to their resistance to antibiotics. New therapeutic approaches are urgently needed to treat these chronic infections. This study aimed to investigate the antibiofilm potential of various plant extracts, specifically targeting mucoid and small colony variants of P. aeruginosa and S. aureus and strains. Moreover, it aimed to gain insights into the mechanisms of action and the potential phytochemicals responsible for antibiofilm activity. Solid-liquid extractions were performed on seven biomasses using water and ethanol (70 and 96 %) under controlled conditions, resulting in 21 distinct plant extracts. These extracts were evaluated for extraction yield, antioxidant activity, phenolic content, chemical composition by HPLC-TOF-MS, and antibiofilm activity using a 96-well plate assay, followed by crystal violet staining, bacterial adhesion assessment, and brightfield microscopy. Our findings revealed that aqueous extracts exhibited the highest inhibition of biofilm formation, with cinnamon bark and moringa seeds showing strong antibiofilm activity against both bacterial species. Brightfield microscopy confirmed that these extracts effectively inhibited biofilm formation. Chemical analysis identified key bioactive compounds, including moringin, benzaldehyde, coumarin, and quinic acid, which likely contribute to the observed antibiofilm effects. Recognizing that the antibiofilm properties of moringin, a common compound in both moringa seed and cinnamon bark extracts, remain underexplored, we conducted potential target identification via PharmMapper and molecular docking analyses to provide a foundation for future research. Computational analyses indicated that moringin might inhibit aspartate-semialdehyde dehydrogenase in P. aeruginosa and potentially interact with an unknown target in S. aureus. In conclusion, moringa seed and cinnamon bark extracts demonstrated significant potential for developing new therapies targeting biofilm-associated infections in CF. Further studies are needed to validate the computational predictions, identify the bacterial targets, and elucidate the precise mechanisms behind moringin's antibiofilm activity, which is likely the potential key contributor to the observed activity of the moringa and cinnamon bark extracts.
Collapse
Affiliation(s)
- Eduarda Silva
- Centre of Biological Engineering, LIBRO – Laboratório de Investigação em Biofilmes Rosário Oliveira, University of Minho, Campus de Gualtar, Braga, 4710-057, Portugal
| | - Pedro Ferreira-Santos
- Centre of Biological Engineering, LIBRO – Laboratório de Investigação em Biofilmes Rosário Oliveira, University of Minho, Campus de Gualtar, Braga, 4710-057, Portugal
- Departamento de Enxeñaría Química, Facultade de Ciencias, University of Vigo, As Lagoas, Ourense 32004, Spain
- Instituto de Agroecoloxía e Alimentación (IAA), University of Vigo (Campus Auga), As Lagoas, 32004, Ourense, Spain
| | - José A. Teixeira
- Centre of Biological Engineering, LIBRO – Laboratório de Investigação em Biofilmes Rosário Oliveira, University of Minho, Campus de Gualtar, Braga, 4710-057, Portugal
- LABBELS – Associate Laboratory, Braga/Guimarães, Portugal
| | - Maria Olivia Pereira
- Centre of Biological Engineering, LIBRO – Laboratório de Investigação em Biofilmes Rosário Oliveira, University of Minho, Campus de Gualtar, Braga, 4710-057, Portugal
- LABBELS – Associate Laboratory, Braga/Guimarães, Portugal
| | - Cristina M.R. Rocha
- Centre of Biological Engineering, LIBRO – Laboratório de Investigação em Biofilmes Rosário Oliveira, University of Minho, Campus de Gualtar, Braga, 4710-057, Portugal
- LABBELS – Associate Laboratory, Braga/Guimarães, Portugal
| | - Ana Margarida Sousa
- Centre of Biological Engineering, LIBRO – Laboratório de Investigação em Biofilmes Rosário Oliveira, University of Minho, Campus de Gualtar, Braga, 4710-057, Portugal
- LABBELS – Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
4
|
Alsamhary KE. The effect of quaternary ammonium compounds (QACs) on quorum sensing and resistance of P. aeruginosa in clinical settings. Microb Pathog 2025; 202:107378. [PMID: 40024542 DOI: 10.1016/j.micpath.2025.107378] [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: 11/29/2024] [Revised: 02/07/2025] [Accepted: 02/12/2025] [Indexed: 03/04/2025]
Abstract
Pseudomonas aeruginosa, a formidable opportunistic pathogen, is notorious for its ability to form biofilms and produce virulence factors that favor chronic infections, especially in cystic fibrosis patients. The misuse of disinfectants, combined with environmental leakage and biodegradation, has led to widespread exposure of microorganisms to sub-lethal concentrations of disinfectants, particularly quaternary ammonium compounds (QACs). This study investigates the interaction between QACs, specifically ethylbenzalkyl dimethyl ammonium chloride (EBAC), and the quorum sensing (QS) mechanisms governing P. aeruginosa behavior. The results demonstrate that exposure to sub-minimum inhibitory concentrations (sub-MICs) of EBAC not only enhances the biofilm-forming capability of P. aeruginosa isolates but also modulates the expression of crucial QS-regulated genes. Notably, the bacteria exhibit increased production of biofilm-associated virulence factors such as pyocyanin and elastase, and altered antibiotic susceptibility profiles, indicating a shift towards persistent infection phenotypes. These findings reveal that QAC exposure can significantly increase resistance to antibiotics and external stressors like hydrogen peroxide. These results emphasize the need to reassess the efficacy of QACs in clinical disinfection settings, particularly against P. aeruginosa infections, and highlight the potential for unintended consequences of their use regarding bacterial behavior and virulence. This study provides novel insights into the role of QACs in modulating QS-mediated virulence and antibiotic resistance, offering a new perspective on the risks associated with sub-lethal disinfectant exposure.
Collapse
Affiliation(s)
- Khawla E Alsamhary
- Department of Biology, College of Science and Humanities in Al-Kharj, Prince Sattam Bin Abdulaziz University, Al-Kharj, 11942, Saudi Arabia.
| |
Collapse
|
5
|
Sajeevan A, Andrew DJ, Patra TN, Solomon AP, Dandela R. Novel synthesis and anti-pathogenic properties of ensifentrine and its intermediates against Pseudomonas aeruginosa. RSC Adv 2025; 15:13053-13063. [PMID: 40271403 PMCID: PMC12015936 DOI: 10.1039/d5ra01722j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Accepted: 04/07/2025] [Indexed: 04/25/2025] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a progressive respiratory disorder marked by persistent lung inflammation and airway constriction. It presents a formidable global health challenge owing to its high morbidity and mortality rates. It is often aggravated by infections from pathogens such as Pseudomonas aeruginosa, a predominant pathogen that accelerates lung function deterioration and triggers frequent exacerbations. Ensifentrine (ENF) exhibits strong anti-inflammatory effects and is a selective dual inhibitor of the enzymes PDE3 and PDE4, which have been reported to be beneficial in treating COPD exacerbation. This study examined the anti-pathogenic activity of ENF against P. aeruginosa by adopting an innovative synthetic route. A series of intermediates were synthesized via the novel route, optimizing the yield and integrity of ENF. Further investigations to determine the activity of the compound against P. aeruginosa involved antibacterial and antibiofilm testing and identification of the potential mechanisms of action. Preliminary results demonstrate that ENF and its intermediate ENFA exhibit 50-60% robust biofilm-inhibition and biofilm-eradication effects at remarkably low concentrations of 3.9 μM and 7.9 μM, respectively. Furthermore, ENF disrupts quorum sensing, leading to a 35% reduction in the production of pyoverdine and exopolysaccharide, which are two key virulence factors of P. aeruginosa. Importantly, ENF exhibits synergistic activity with ciprofloxacin, further enhancing its antimicrobial efficacy at a concentration of 0.25 μg mL-1. This study focuses on the innovative synthesis of ENF and its promising anti-pathogenic properties, which may make it an effective adjunctive treatment for COPD caused by P. aeruginosa.
Collapse
Affiliation(s)
- Anusree Sajeevan
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University Thanjavur India
| | - Deepthi Joseph Andrew
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University Thanjavur India
| | - T Nalinikanta Patra
- Department of Industrial and Engineering Chemistry, Institute of Chemical Technology Bhubaneswar Odisha India
| | - Adline Princy Solomon
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University Thanjavur India
| | - Rambabu Dandela
- Department of Industrial and Engineering Chemistry, Institute of Chemical Technology Bhubaneswar Odisha India
| |
Collapse
|
6
|
Rafiee Z, Rezaie M, Choi S. Rapid and sensitive antimicrobial susceptibility testing of biofilm-forming bacteria using scalable paper-based organic transistors. iScience 2025; 28:112312. [PMID: 40264793 PMCID: PMC12013490 DOI: 10.1016/j.isci.2025.112312] [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: 01/03/2025] [Revised: 02/24/2025] [Accepted: 03/25/2025] [Indexed: 04/24/2025] Open
Abstract
A scalable, cost-effective paper-based organic field-effect transistor platform has been developed for rapid antimicrobial susceptibility testing (AST) of biofilm-forming pathogens. Traditional AST methods are costly, labor-intensive, and slow, with a lack of standardized biofilm models. This system directly tracks protons generated by biofilms, which serve as key indicators of bacterial metabolism under antibiotic exposure. A proton-sensitive PEDOT:PSS channel is employed, where metabolic proton activity de-dopes the transistor, reducing conductivity. The engineered paper substrate facilitates rapid, high-quality biofilm formation, improving assay reliability. The platform was validated on three clinically significant pathogens against frontline antibiotics, providing real-time, quantitative antibiotic efficacy profiles. Integrated with a microcontroller and machine learning algorithm, results are displayed on a liquid crystal display (LCD), classifying antibiotic concentration relative to the minimum inhibitory concentration with over 85% accuracy. This clinically translatable system offers a high-throughput, point-of-care solution for efficient infection management and antibiotic stewardship.
Collapse
Affiliation(s)
- Zahra Rafiee
- Bioelectronics & Microsystems Laboratory, Department of Electrical & Computer Engineering, State University of New York at Binghamton, Binghamton, NY 13902, USA
| | - Maryam Rezaie
- Bioelectronics & Microsystems Laboratory, Department of Electrical & Computer Engineering, State University of New York at Binghamton, Binghamton, NY 13902, USA
| | - Seokheun Choi
- Bioelectronics & Microsystems Laboratory, Department of Electrical & Computer Engineering, State University of New York at Binghamton, Binghamton, NY 13902, USA
- Center for Research in Advanced Sensing Technologies & Environmental Sustainability, State University of New York at Binghamton, Binghamton, NY 13902, USA
| |
Collapse
|
7
|
Nahar S, Mahamud AGMSU, Cho AJ, Ashrafudoulla M, Yu J, Park SH, Ha SD. Flavourzyme Suppresses Pseudomonas aeruginosa Biofilms by Targeting Motility, Quorum Sensing, and Virulence Genes. Curr Microbiol 2025; 82:240. [PMID: 40210784 DOI: 10.1007/s00284-025-04200-6] [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: 11/14/2024] [Accepted: 03/19/2025] [Indexed: 04/12/2025]
Abstract
The biofilm-mediated persistence of Pseudomonas aeruginosa in the food and biomedical sectors is currently a global concern. In light of this challenge, this study investigated a preventive approach against P. aeruginosa biofilm formation using Flavourzyme, a food-grade peptidase, considering its antibiofilm potential. The results revealed that a co-culture comprising 300 µL/mL (1 × minimum inhibitory concentration [MIC]) of Flavourzyme could kill P. aeruginosa. On the MBEC™ biofilm-forming device, 0.125 × MIC of Flavourzyme inhibited > 4.5 log CFU/peg of biofilm. Cell motilities and the biosynthesis of quorum sensing (QS) molecules such as N-acyl-homoserine lactones (AHLs), including C4-HSL, decreased significantly at 0.06 × MIC of Flavourzyme and became undetectable at 0.125 × MIC. Interestingly, while 0.03 × MIC of Flavourzyme elicited diverse expressions of QS and virulence-regulating genes, ≥ 0.06 × MIC of Flavourzyme remarkably suppressed the relative genomic expressions. This study proposes Flavourzyme as a potent antibiofilm agent against P. aeruginosa biofilms, recommending specific concentrations for effective use in food preservation.
Collapse
Affiliation(s)
- Shamsun Nahar
- Department of Food Safety and Regulatory Science, Chung-Ang University, Anseong-si, Gyeonggi-do, 17546, Republic of Korea
- GreenTech-Based Food Safety Research Group, Chung-Ang University, BK21 Four, 4726 Seodong-Daero, Anseong-si, Gyeonggi-do, 17546, Republic of Korea
- Department of Obstetrics, Gynecology, and Women's Health, University of Missouri, Columbia, MO, 65211, USA
| | - A G M Sofi Uddin Mahamud
- Department of Food Safety and Regulatory Science, Chung-Ang University, Anseong-si, Gyeonggi-do, 17546, Republic of Korea
- GreenTech-Based Food Safety Research Group, Chung-Ang University, BK21 Four, 4726 Seodong-Daero, Anseong-si, Gyeonggi-do, 17546, Republic of Korea
- Department of Microbiology, University of Georgia, Athens, GA, 30602, USA
| | - Ah Jin Cho
- Department of Food Safety and Regulatory Science, Chung-Ang University, Anseong-si, Gyeonggi-do, 17546, Republic of Korea
- GreenTech-Based Food Safety Research Group, Chung-Ang University, BK21 Four, 4726 Seodong-Daero, Anseong-si, Gyeonggi-do, 17546, Republic of Korea
| | - Md Ashrafudoulla
- Department of Food Safety and Regulatory Science, Chung-Ang University, Anseong-si, Gyeonggi-do, 17546, Republic of Korea
- GreenTech-Based Food Safety Research Group, Chung-Ang University, BK21 Four, 4726 Seodong-Daero, Anseong-si, Gyeonggi-do, 17546, Republic of Korea
- National Institutes of Health, 9000 Rockville Pike, Bethesda, MD, 20892, USA
| | - Jisu Yu
- Lotte R&D Center, 201, Magokjungang-ro, Gangseo-gu, Seoul, 07594, Republic of Korea
| | - Si Hong Park
- Department of Food Science & Technology, Oregon State University, Corvallis, OR, 97331, USA
| | - Sang-Do Ha
- Department of Food Safety and Regulatory Science, Chung-Ang University, Anseong-si, Gyeonggi-do, 17546, Republic of Korea.
- GreenTech-Based Food Safety Research Group, Chung-Ang University, BK21 Four, 4726 Seodong-Daero, Anseong-si, Gyeonggi-do, 17546, Republic of Korea.
| |
Collapse
|
8
|
Nazari MJ, Anwary MT, Ghazanfar K, Amiri ME, Hafid SY, Jawad MJ, Mosawi SH. Inhibition of acyl-homoserine-lactone synthase in Pseudomonas aeruginosa biofilms by 7-O-methyl-aromadendrin by using molecular docking and molecular dynamics simulation. In Silico Pharmacol 2025; 13:56. [PMID: 40226106 PMCID: PMC11982000 DOI: 10.1007/s40203-025-00350-4] [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: 01/08/2025] [Accepted: 03/31/2025] [Indexed: 04/15/2025] Open
Abstract
This study investigates the potential of 7-O-methyl aromadendrin (7-OMA), a naturally occurring flavonoid-glycoside, as an inhibitor of acyl-homoserine-lactone (AHL) synthase in Pseudomonas aeruginosa, a key enzyme in quorum sensing and biofilm formation. Using molecular docking and molecular dynamics simulations, we evaluated the binding interactions and inhibitory effects of 7-OMA on AHL synthase. Molecular docking revealed a suitable binding affinity (-6.66 kcal/mol) between 7-OMA and the enzyme, with interactions at critical active site residues. Molecular dynamics simulations demonstrated that 7-OMA stabilizes the enzyme through hydrogen bonds and van der Waals interactions while enhancing its structural flexibility. The average RMSD of AHL synthase increased slightly in the presence of 7-OMA, indicating partial instability of the enzyme. Additionally, the average Rg value increased, suggesting that 7-OMA may expand the enzyme structure or reduce its compactness. MM-PBSA analysis confirmed the strength of these interactions, with favorable van der Waals and electrostatic contributions to the binding energy. These results suggest that 7-OMA disrupts the structural dynamics of AHL synthase, potentially inhibiting biofilm formation and reducing the virulence of Pseudomonas aeruginosa. The findings highlight the therapeutic potential of 7-OMA as a novel inhibitor of AHL synthase, offering a promising strategy to combat biofilm-associated infections. Future studies should focus on evaluating the bioavailability, in vivo efficacy, and clinical applicability of 7-OMA, as well as its broader activity against other multidrug-resistant pathogens. Graphical abstract
Collapse
Affiliation(s)
- Mohammad Jalal Nazari
- Department of Internal Medicine, Faculty of Medicine, Khatam Al-Nabieen University, Kabul, Afghanistan
- Medical Research and Technology Center, Khatam Al-Nabieen University, Kabul, Afghanistan
| | - Mohammad Tariq Anwary
- Department of Internal Medicine, Faculty of Medicine, Khatam Al-Nabieen University, Kabul, Afghanistan
- Medical Research and Technology Center, Khatam Al-Nabieen University, Kabul, Afghanistan
| | - Khanbaba Ghazanfar
- Department of Internal Medicine, Faculty of Medicine, Khatam Al-Nabieen University, Kabul, Afghanistan
- Medical Research and Technology Center, Khatam Al-Nabieen University, Kabul, Afghanistan
| | - Mohammad Edris Amiri
- Department of Internal Medicine, Faculty of Medicine, Khatam Al-Nabieen University, Kabul, Afghanistan
- Medical Research and Technology Center, Khatam Al-Nabieen University, Kabul, Afghanistan
| | - Sayed Yahya Hafid
- Department of Internal Medicine, Faculty of Medicine, Khatam Al-Nabieen University, Kabul, Afghanistan
| | - Mohammad Jawad Jawad
- Department of Internal Medicine, Faculty of Medicine, Khatam Al-Nabieen University, Kabul, Afghanistan
| | | |
Collapse
|
9
|
Hassan WHB, Ghani AEA, Taema EA, Yahya G, El-Sadek ME, Mansour B, Abdel-Halim MS, Arafa AM. Chemical profile, virtual screening, and virulence-inhibiting properties of Sphagneticola trilobata L. essential oils against Pseudomonas aeruginosa. Sci Rep 2025; 15:11964. [PMID: 40199892 PMCID: PMC11978798 DOI: 10.1038/s41598-025-94486-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 03/13/2025] [Indexed: 04/10/2025] Open
Abstract
The escalating threat of microbial resistance underscores the urgent need for innovative solutions, including natural agents capable of attenuating virulence. This study explores the antimicrobial and anti-virulence potential of essential oils (EOs) derived from Sphagneticola trilobata against Pseudomonas aeruginosa. Through GC/MS analysis, volatile metabolites from the flower heads and leaves/stems of Egyptian S. trilobata were identified, revealing 43 and 62 components, respectively. Key compounds included α-phellandrene, α-pinene, D-limonene, and α-thujene. The Minimum Inhibitory Concentrations (MIC) of flower head and leaf/stem EOs against P. aeruginosa were 1.17% and 1.75% v/v, respectively. At sub-MIC doses (1/8th of the MIC), the EOs exhibited significant anti-virulence properties, including complete inhibition of protease activity and disruption of biofilm formation, which are crucial for bacterial survival and pathogenicity. Additionally, they effectively suppressed the expression of quorum sensing genes, which are essential for bacterial communication and virulence. Virtual screening of four major EO components (+)-(R)-limonene, (±)-α-pinene, α-phellandrene, and α-thujene against five critical protein targets involved in biofilm formation, quorum sensing, virulence, and protease activity in P. aeruginosa further supported their anti-virulence and antibiofilm actions, showing high affinity for these targets. These findings suggest that the EOs of S. trilobata hold great potential as natural virulence attenuating agents, particularly against biofilm-forming pathogens like P. aeruginosa.
Collapse
Affiliation(s)
- Wafaa H B Hassan
- Department of Pharmacognosy, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| | - Afaf E Abdel Ghani
- Department of Pharmacognosy, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| | - Esraa A Taema
- Department of Pharmacognosy, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| | - Galal Yahya
- Department of Microbiology and Immunology, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| | - Mohamed E El-Sadek
- Department of Medicinal Chemistry, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| | - Basem Mansour
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Delta University for Science and Technology, International Coastal Road, Gamasa City, 35712, Egypt
| | - Mahmoud Saad Abdel-Halim
- Department of Microbiology and Immunology, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| | - Asmaa M Arafa
- Department of Pharmacognosy, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt.
| |
Collapse
|
10
|
Zou J, Wang J, Gao L, Xue W, Zhu J, Zhang Y, Gou S, Liu H, Zhong C, Ni J. Ultra-short lipopeptides containing d-amino acid exhibiting excellent stability and antibacterial activity against gram-positive bacteria. Eur J Med Chem 2025; 287:117341. [PMID: 39908797 DOI: 10.1016/j.ejmech.2025.117341] [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: 12/11/2024] [Revised: 01/21/2025] [Accepted: 01/29/2025] [Indexed: 02/07/2025]
Abstract
As novel antibacterial agents, antimicrobial peptides (AMPs) possess broad-spectrum antibacterial activity and low drug resistance, holding significant development potential. Nevertheless, the stability of AMPs significantly restricts their application. In light of this, we synthesized a series of ultra-short lipopeptides using d-amino acid substitution to enhance the stability of ultra-short lipopeptide C12-RRW-NH2 that was selected from our previous research while maintaining its antibacterial activity against gram-positive bacteria. Amongst, the ultra-short lipopeptide Lip7 (C12-rrw-NH2) with full d-amino acid demonstrated outstanding stability in protease, serum, and salt ion environments. It exerted excellent antibacterial activity against gram-positive bacteria, especially against methicillin-resistant Staphylococcus aureus (MRSA). Meanwhile, Lip7 presented a low propensity to develop bacterial resistance with potential for combination therapy with conventional antibiotics. Studies on its antibacterial mechanism revealed that Lip7 could rapidly depolarize the bacterial cytoplasmic membrane, disrupt the integrity of the bacterial membrane, lead to leakage of nucleic acid and protein, promote the generation of reactive oxygen species, and ultimately result in bacterial death. Additionally, Lip7 also exhibited therapeutic potential in both local and systemic MRSA-infected mice models with better safety in vivo. These findings highlighted that Lip7 is an ideal novel antibacterial alternative to offer guiding schemes for developing high-stability antimicrobial peptides to fight multidrug-resistant gram-bacteria.
Collapse
Affiliation(s)
- Jing Zou
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China
| | - Jiahui Wang
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China
| | - Luyang Gao
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China
| | - Wenjing Xue
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China
| | - Jingyi Zhu
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China
| | - Yun Zhang
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China; State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China
| | - Sanhu Gou
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China; State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China
| | - Hui Liu
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China; State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China
| | - Chao Zhong
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China; State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China.
| | - Jingman Ni
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China; State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China.
| |
Collapse
|
11
|
Delgado-Nungaray JA, Figueroa-Yáñez LJ, Reynaga-Delgado E, García-Ramírez MA, Aguilar-Corona KE, Gonzalez-Reynoso O. Influence of Amino Acids on Quorum Sensing-Related Pathways in Pseudomonas aeruginosa PAO1: Insights from the GEM iJD1249. Metabolites 2025; 15:236. [PMID: 40278365 DOI: 10.3390/metabo15040236] [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: 03/07/2025] [Revised: 03/20/2025] [Accepted: 03/26/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND/OBJECTIVES Amino acids (AAs) play a critical role in diseases such as cystic fibrosis where Pseudomonas aeruginosa PAO1 adapts its metabolism in response to host-derived nutrients. The adaptation influences virulence and complicates antibiotic treatment mainly for the antimicrobial resistance context. D- and L-AAs have been analyzed for their impact on quorum sensing (QS), a mechanism that regulates virulence factors. This research aimed to reconstruct the genome-scale metabolic model (GEM) of P. aeruginosa PAO1 to investigate the metabolic roles of D- and L-AAs in QS-related pathways. METHODS The updated GEM, iJD1249, was reconstructed by using protocols to integrate data from previous models and refined with well-standardized in silico media (LB, M9, and SCFM) to improve flux balance analysis accuracy. The model was used to explore the metabolic impact of D-Met, D-Ala, D-Glu, D-Ser, L-His, L-Glu, L-Arg, and L-Ornithine (L-Orn) at 5 and 50 mM in QS-related pathways, focusing on the effects on bacterial growth and carbon flux distributions. RESULTS Among the tested AAs, D-Met was the only one that did not enhance the growth rate of P. aeruginosa PAO1, while L-Arg and L-Orn increased fluxes in the L-methionine biosynthesis pathway, influencing the metH gene. These findings suggest a differential metabolic role for D-and L-AAs in QS-related pathways. CONCLUSIONS Our results shed some light on the metabolic impact of AAs on QS-related pathways and their potential role in P. aeruginosa virulence. Future studies should assess D-Met as a potential adjuvant in antimicrobial strategies, optimizing the concentration in combination with antibiotics to maximize its therapeutic effectiveness.
Collapse
Affiliation(s)
- Javier Alejandro Delgado-Nungaray
- Chemical Engineering Department, University Center for Exact and Engineering Sciences, University of Guadalajara, Blvd. M. García Barragán # 1451, Guadalajara 44430, Mexico
| | - Luis Joel Figueroa-Yáñez
- Industrial Biotechnology Unit, Center for Research and Assistance in Technology and Design of the State of Jalisco, A.C. (CIATEJ), Zapopan 45019, Mexico
| | - Eire Reynaga-Delgado
- Pharmacobiology Department, University Center for Exact and Engineering Sciences, University of Guadalajara, Blvd. M. García Barragán # 1451, Guadalajara 44430, Mexico
| | - Mario Alberto García-Ramírez
- Electronics Department, University Center for Exact and Engineering Sciences, University of Guadalajara, Blvd. M. García Barragán # 1451, Guadalajara 44430, Mexico
| | - Karla Esperanza Aguilar-Corona
- Food Engineering and Biotechnology, University Center for Exact and Engineering Sciences, University of Guadalajara, Blvd. M. García Barragán # 1451, Guadalajara 44430, Mexico
| | - Orfil Gonzalez-Reynoso
- Chemical Engineering Department, University Center for Exact and Engineering Sciences, University of Guadalajara, Blvd. M. García Barragán # 1451, Guadalajara 44430, Mexico
| |
Collapse
|
12
|
Mahmud MN, Momoshed M, Ahamed Talukder MF, Ferdous J, Koly FA, Islam S. Exploring the occurrence of Pseudomonas aeruginosa and comprehensive whole genome analysis of the bcsir_p4_s20 strain from municipal wastewater in Chattogram. World J Microbiol Biotechnol 2025; 41:112. [PMID: 40148700 DOI: 10.1007/s11274-025-04328-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Accepted: 03/17/2025] [Indexed: 03/29/2025]
Abstract
Various studies reported the existence of multidrug-resistant (MDR) Pseudomonas aeruginosa in environmental samples, including hospital wastewater, municipal wastewater, and surface water. In this study, we investigated the impact of untreated municipal wastewater transmitting antibiotic-resistant P. aeruginosa strains in wastewater networks of Chattogram City, Bangladesh, through antibiotic susceptibility profiles and whole-genome sequencing (WGS) of the MDR P. aeruginosa bcsir_p4_s20. Forty-two P. aeruginosa isolates were identified from eight locations using polymerase chain reaction (PCR), targeting the oprI and oprL genes, and antibiotic susceptibility was determined against 11 antibiotics by the disc diffusion method. Resistant isolates were identified at all locations, with the highest resistance frequency displayed towards meropenem, cefepime, and colistin. The WGS of bcsir_p4_s20 was performed using the NextSeq 2000 platform. Several bioinformatics tools, like FastQC, Trimmomatic, SPAdes, and Prokka, were used for quality evaluation, low-quality read and adapter filtration, de novo assembly, and functional annotation. Comprehensive Antibiotic Resistance Database (CARD), AMRFinderPlus, and virulence factor database (VFDB) were employed to determine resistance genes and virulence factors. The strain belongs to the O7 serogroup and sequence type ST357. The analysis identified antibiotic resistance genes (blaPDC-11, sul1, and others) that cause resistance through efflux pump and inactivation mechanisms, and virulent genes responsible for adherence (flagella, type IV pili), enzyme (phospholipase C), iron uptake (pyoverdine), secretion system (exoT, exoU), and toxin (toxA) secretion. Therefore, municipal wastewater is a potential reservoir for MDR P. aeruginosa, and establishing wastewater treatment plants (WWTPs) at the primary source points before discharging it to the wastewater network is suggested to mitigate the risk of outbreaks.
Collapse
Affiliation(s)
- Md Nuruddin Mahmud
- Department of Microbiology, University of Chittagong, Chattogram, Bangladesh
| | - Momthahena Momoshed
- Department of Microbiology, University of Chittagong, Chattogram, Bangladesh
| | | | - Jannatul Ferdous
- Industrial Microbiology Research Division, BCSIR Chattogram Laboratories, Bangladesh Council of Scientific and Industrial Research (BCSIR), Chattogram, 4220, Bangladesh
| | - Farjana Akter Koly
- Industrial Microbiology Research Division, BCSIR Chattogram Laboratories, Bangladesh Council of Scientific and Industrial Research (BCSIR), Chattogram, 4220, Bangladesh
| | - Saiful Islam
- Industrial Microbiology Research Division, BCSIR Chattogram Laboratories, Bangladesh Council of Scientific and Industrial Research (BCSIR), Chattogram, 4220, Bangladesh.
| |
Collapse
|
13
|
Liu Z, Yang Y, Xie X, Li R, You J, Zhao X, Wang Y, Guo J. Development and Characterization of LasR Immobilized Monolithic Column for Screening Active Ingredients as Quorum Sensing Inhibitors Against P. aeruginosa in Natural Products. Drug Des Devel Ther 2025; 19:2051-2064. [PMID: 40124560 PMCID: PMC11929421 DOI: 10.2147/dddt.s501621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 03/01/2025] [Indexed: 03/25/2025] Open
Abstract
Background and Aim The enzyme/protein immobilized monolithic capillary combined with liquid chromatography-mass spectrometry is an efficient screening strategy for the corresponding agonist/antagonist. LasR is the potential therapeutic target since it plays a vital role in the colonization and invasion of Pseudomonas aeruginosa (P. aeruginosa). Therefore, reagents that inhibit LasR may be effective against P. aeruginosa. To screen and find LasR inhibitors rapidly, a LasR-immobilized monolithic capillary column was prepared and characterized. Methods Firstly, the recombinant LasR protein was prepared in E. coli. Then, the LasR protein was immobilized to the surface of poly (glycidyl methacrylate-co-poly(ethylene glycol)diacrylate)-ethylenediamine monolithic column. The affinity and stability of prepared column was also evaluated. Furthermore, the prepared column was applied to fishing LasR inhibitor in Scutellaria baicalensis Georgi extract. The interaction of the screening compound to LasR was confirmed through molecular docking. Results The recombinant active LasR protein was prepared in E. coli. After purification and validation, a comparative ligand fishing monolithic column was prepared through immobilizing LasR to the surface of the poly (glycidyl methacrylate-co-poly(ethylene glycol)diacrylate)-ethylenediamine through amidation reaction. The LasR was successfully immobilized to the monolithic column characterizing by Fourier transform infrared spectroscopy and scanning electron microscopy. The activity of immobilized LasR was reserved as it has affinity to the nature ligand 3-oxo-C12-HSL and stablied within 24 h at 4 °C. In the Scutellaria baicalensis Georgi extract, baicalein was screened as a potential LasR inhibitor. The molecular docking results and the in vivo evaluation confirmed the activity of baicalein. Conclusion The proposed LasR immobilized monolithic column is a viable strategy in screening LasR inhibitors. It can be considered as a possible alternative to traditional methods for screening LasR inhibitors as drug candidates against P. aeruginosa.
Collapse
Affiliation(s)
- Zheng Liu
- School of Medicine, Foshan University, Foshan, 528000, People’s Republic of China
| | - Yue Yang
- School of Medicine, Foshan University, Foshan, 528000, People’s Republic of China
| | - Xiaoyuan Xie
- School of Medicine, Foshan University, Foshan, 528000, People’s Republic of China
| | - Rui Li
- School of Medicine, Foshan University, Foshan, 528000, People’s Republic of China
| | - Jifeng You
- School of Medicine, Foshan University, Foshan, 528000, People’s Republic of China
| | - Xianglong Zhao
- School of Medicine, Foshan University, Foshan, 528000, People’s Republic of China
- College of Pharmacy, Jinan University, Guangzhou, 510632, People’s Republic of China
| | - Yuanyuan Wang
- School of Medicine, Foshan University, Foshan, 528000, People’s Republic of China
| | - Jialiang Guo
- School of Medicine, Foshan University, Foshan, 528000, People’s Republic of China
- College of Pharmacy, Jinan University, Guangzhou, 510632, People’s Republic of China
- Guangdong Provincial Engineering Technology Research Center of Whole Process Quality Control and Analysis of Lingnan Traditional Chinese Medicine, Foshan, 528225, People’s Republic of China
| |
Collapse
|
14
|
Tuytschaever T, Raes K, Sampers I. Biofilm detection in the food industry: Challenges in identifying biofilm eps markers and analytical techniques with insights for Listeria monocytogenes. Int J Food Microbiol 2025; 432:111091. [PMID: 39923351 DOI: 10.1016/j.ijfoodmicro.2025.111091] [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: 10/17/2024] [Revised: 01/30/2025] [Accepted: 01/31/2025] [Indexed: 02/11/2025]
Abstract
Extracellular polymeric substances (EPS) in biofilms are promising targets for eradicating biofilms and monitoring their presence, especially in the food industry. For this understanding, the composition of the EPS matrix is crucial. Ideally, a biofilm marker is found serving both purposes, but such a compound has not yet been discovered. This review aims to identify general biofilm EPS markers distinct from planktonic cells, focusing on macromolecules in the EPS matrix. It also evaluates the feasibility of this goal across different bacterial groups and environmental conditions and discusses EPS analysis methods. This review digs deeper into the EPS matrix starting with an introduction to the EPS matrix itself and describing some of its influencing factors. Next, a brief description of cell-to-cell communication within biofilms is provided, as these interactions significantly influence the EPS matrix. The main part of this review describes the macromolecules inside the EPS matrix and attempts to find biofilm EPS markers applied to bacteria in general and specifically to Listeria monocytogenes as biofilms are a major contributor to its persistence. The last part of the review focuses on the analytical techniques available to characterize the EPS matrix. The review revealed that although multiple candidates showed great potential as biofilm markers, none were unique but ubiquitous in all bacteria tested. To achieve easy biofilm detection with current techniques, it's necessary to identify markers specific to the environmental conditions and common bacterial groups within each food category, sector, or facility, due to the lack of standardization in these techniques. This tailored approach ensures more accurate and effective biofilm monitoring. Moreover, the lack of standardized analytical techniques, including quantification techniques, complexifies studying the EPS matrix and developing monitoring and intervention strategies. Optimizing analytical techniques is crucial for this tailored approach, as it requires refined methods for detection, characterization, and quantification. This ensures the accurate identification of biofilm markers specific to environmental conditions and bacterial groups within each food sector.
Collapse
Affiliation(s)
- Tessa Tuytschaever
- Research Unit VEG-i-TEC, Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, Campus Kortrijk, Sint-Martens-Latemlaan 2B, 8500 Kortrijk, Belgium.
| | - Katleen Raes
- Research Unit VEG-i-TEC, Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, Campus Kortrijk, Sint-Martens-Latemlaan 2B, 8500 Kortrijk, Belgium.
| | - Imca Sampers
- Research Unit VEG-i-TEC, Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, Campus Kortrijk, Sint-Martens-Latemlaan 2B, 8500 Kortrijk, Belgium.
| |
Collapse
|
15
|
Li H, Zhang S, Li Q, Shu Y, Li S, Wu B, Xu Z. The Role of Yinqiao Powder in Modulating Pseudomonas aeruginosa Biofilm and Virulence Factors. Infect Drug Resist 2025; 18:1405-1414. [PMID: 40098712 PMCID: PMC11911820 DOI: 10.2147/idr.s507257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 03/04/2025] [Indexed: 03/19/2025] Open
Abstract
Purpose It is now understood that the primary challenges in treating Pseudomonas aeruginosa (P. aeruginosa) infections are the secretion of multiple virulence factors, the formation of biofilm, and the emergence of multi-drug resistance. Small regulatory RNAs (sRNAs) and quorum sensing (QS) play an important role in regulating bacterial biofilms and multiple virulence factors, presenting potential targets for novel anti-P. aeruginosa therapies. Yinqiao Powder has demonstrated inhibitory activity against various bacteria and viruses. The objective of this study was to elucidate the precise mechanism of Yinqiao Powder's impact on P. aeruginosa virulence and to ascertain its clinical utility. Methods First, the effects of Yinqiao Powder on various virulence factors of P. aeruginosa were assessed through virulence phenotype experiments, including biofilm formation assay, pyocyanin production assay, rhamnolipid assay, and motility assay. Then, a cytotoxicity assay was used to evaluate the effect of P. aeruginosa treated by Yinqiao Powder on cells. Finally, an RT-qPCR assay was used to detect the effects of Yinqiao Powder on QS system and virulence-related gene expression. Results This study revealed that sub-minimum inhibitory concentration (sub-MIC) levels of Yinqiao Powder significantly inhibit biofilm formation, swarming motility, pyocyanin and rhamnolipid production in a dose-dependent manner. The cytotoxicity assay also confirmed that Yinqiao Powder weakened the cytotoxicity of P. aeruginosa. Furthermore, Yinqiao Powder was found to modulate the P. aeruginosa sRNA-QS-virulence network. Specifically, it repressed the lasI, the rhlI, and sRNA P27 while upregulating sRNA PhrD. Additionally, the phzA and pqsA genes, associated with pyocyanin and rhamnolipid/biofilm regulation, respectively, were repressed by Yinqiao Powder. Conclusion Yinqiao Powder effectively inhibits QS system-related regulatory genes, sRNAs, biofilm formation, swarming motility, pyocyanin and rhamnolipid production at specific concentrations. These results support the potential of Yinqiao Powder as a quorum-sensing inhibitor.
Collapse
Affiliation(s)
- Honglin Li
- Department of Clinical Laboratory, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, 621000, People's Republic of China
| | - Shebin Zhang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, People's Republic of China
| | - Qiwei Li
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, People's Republic of China
| | - Yurong Shu
- Department of Clinical Laboratory, South China Agricultural University Hospital, Guangzhou, 510000, People's Republic of China
| | - Song Li
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, People's Republic of China
| | - Bo Wu
- Department of Transfusion, The Fourth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511300, People's Republic of China
| | - Zhenjie Xu
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, People's Republic of China
| |
Collapse
|
16
|
Pérez-Ropero G, Dolcemascolo R, Pérez-Ràfols A, Andersson K, Danielson UH, Rodrigo G, Buijs J. Regulatory Effects of RNA-Protein Interactions Revealed by Reporter Assays of Bacteria Grown on Solid Media. BIOSENSORS 2025; 15:175. [PMID: 40136972 PMCID: PMC11940492 DOI: 10.3390/bios15030175] [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: 01/23/2025] [Revised: 03/03/2025] [Accepted: 03/06/2025] [Indexed: 03/27/2025]
Abstract
Reporter systems are widely used to study biomolecular interactions and processes in vivo, representing one of the basic tools used to characterize synthetic regulatory circuits. Here, we developed a method that enables the monitoring of RNA-protein interactions through a reporter system in bacteria with high temporal resolution. For this, we used a Real-Time Protein Expression Assay (RT-PEA) technology for real-time monitoring of a fluorescent reporter protein, while having bacteria growing on solid media. Experimental results were analyzed by fitting a three-variable Gompertz growth model. To validate the method, the interactions between a set of RNA sequences and the RNA-binding protein (RBP) Musashi-1 (MSI1) were evaluated, as well as the allosteric modulation of the interaction by a small molecule (oleic acid). This new approach proved to be suitable to quantitatively characterize RNA-RBP interactions, thereby expanding the toolbox to study molecular interactions in living bacteria, including allosteric modulation, with special relevance for systems that are not suitable to be studied in liquid media.
Collapse
Affiliation(s)
- Guillermo Pérez-Ropero
- Ridgeview Instruments AB, 75237 Uppsala, Sweden (J.B.)
- Department of Chemistry—BMC, Uppsala University, 75123 Uppsala, Sweden
| | - Roswitha Dolcemascolo
- Institute for Integrative Systems Biology (I2SysBio), Centro Superior de Investigaciones Científicas (CSIC)—University of Valencia, 46980 Paterna, Spain
- Department of Biotechnology, Polytechnic University of Valencia, 46022 Valencia, Spain
| | - Anna Pérez-Ràfols
- Giotto Biotech SRL, 50019 Sesto Fiorentino, Italy
- Magnetic Resonance Center (CERM), Department of Chemistry Ugo Schiff, Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine (CIRMMP), University of Florence, 50019 Sesto Fiorentino, Italy
| | - Karl Andersson
- Ridgeview Instruments AB, 75237 Uppsala, Sweden (J.B.)
- Department of Immunology, Genetics, and Pathology, Uppsala University, 75185 Uppsala, Sweden
| | - U. Helena Danielson
- Department of Chemistry—BMC, Uppsala University, 75123 Uppsala, Sweden
- Science for Life Laboratory, Drug Discovery & Development Platform, Uppsala University, 75123 Uppsala, Sweden
| | - Guillermo Rodrigo
- Institute for Integrative Systems Biology (I2SysBio), Centro Superior de Investigaciones Científicas (CSIC)—University of Valencia, 46980 Paterna, Spain
| | - Jos Buijs
- Ridgeview Instruments AB, 75237 Uppsala, Sweden (J.B.)
- Department of Immunology, Genetics, and Pathology, Uppsala University, 75185 Uppsala, Sweden
| |
Collapse
|
17
|
Zhang N, Li X, Liu X, Cheng P, Li L, Chai Y, Cao M, Yang Y. Aspirin enhances the antibacterial activity of colistin against multidrug-resistant Pseudomonas aeruginosa. Eur J Pharmacol 2025; 997:177480. [PMID: 40057155 DOI: 10.1016/j.ejphar.2025.177480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 03/05/2025] [Accepted: 03/05/2025] [Indexed: 03/14/2025]
Abstract
Multidrug-resistant (MDR) Pseudomonas aeruginosa (PSA), recently reclassified by the World Health Organization (WHO) as a high-priority antimicrobial-resistant pathogen, continues to impose a substantial global health burden due to escalating resistance and stagnant therapeutic innovation. Colistin retains critical clinical utility against MDR P. aeruginosa infections; however, its dose-limiting nephrotoxicity and neurotoxicity necessitate strategies to optimise therapeutic indices. This study investigated the molecular mechanism underlying the synergistic activity of aspirin in potentiating colistin efficacy against MDR P. aeruginosa. In vitro analyses revealed marked synergistic bactericidal activity (FIC index ≤0.5), with metabolomic profiling demonstrating suppression of key metabolic pathways integral to bacterial membrane biogenesis, including glycerophospholipid metabolism and fatty acid biosynthesis. Ultrastructural imaging confirmed irreversible disruption of membrane integrity via combined treatment. In a rat model of P. aeruginosa-induced pneumonia, colistin-aspirin co-administration demonstrated superior efficacy to monotherapy, significantly reducing pulmonary bacterial load (3 to 4-log CFU/g reduction vs colistin alone; p < 0.01), attenuating histopathological injury, and suppressing pro-inflammatory cytokine levels (IL-6, IL-8, TNF-α) by 30-47%. Critically, this synergy enabled a reduction of colistin dosing to one-sixteenth while maintaining bactericidal potency. These findings provide mechanistic insights into aspirin-mediated colistin sensitisation and evidence supporting combinatorial regimens to circumvent colistin toxicity barriers. This work establishes a rational foundation for clinical translation of repurposed aspirin-colistin therapy against MDR P. aeruginosa infections.
Collapse
Affiliation(s)
- Ning Zhang
- Guizhou University of Traditional Chinese Medicine, 550025, Guiyang, China; College of Acupuncture and Massage, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China; First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, 550000, Guizhou, China
| | - Xue Li
- Guizhou University of Traditional Chinese Medicine, 550025, Guiyang, China; Department of the Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, 550003, Guiyang, China
| | - Xin Liu
- Guizhou University of Traditional Chinese Medicine, 550025, Guiyang, China; School of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China
| | - Ping Cheng
- Animal-Derived Food Safety Innovation Team, College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, Anhui, China
| | - Lailai Li
- Guizhou University of Traditional Chinese Medicine, 550025, Guiyang, China; School of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China
| | - Yihui Chai
- Guizhou University of Traditional Chinese Medicine, 550025, Guiyang, China; School of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China
| | - Mingle Cao
- People's Hospital of Duyun City, Duyun, 558000, Guizhou, China
| | - Yuqi Yang
- Guizhou University of Traditional Chinese Medicine, 550025, Guiyang, China; School of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China.
| |
Collapse
|
18
|
de Oliveira Silva YR, Contreras-Martel C, Rodrigues de Melo R, Zanphorlin LM, Trindade DM, Dessen A. Architecture of an embracing lipase-foldase complex of the type II secretion system of Acinetobacter baumannii. Structure 2025; 33:601-612.e4. [PMID: 39904335 DOI: 10.1016/j.str.2024.12.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 10/23/2024] [Accepted: 12/28/2024] [Indexed: 02/06/2025]
Abstract
Acinetobacter baumannii is a major human pathogen responsible for a growing number of multi-antibiotic-resistant infections, and of critical priority for the World Health Organization (WHO). A. baumannii employs a type II secretion system (T2SS) to secrete toxins extracellularly to enable cytotoxicity and colonization. Lipase LipA, secreted by the A. baumannii T2SS, is required for virulence and fitness, and in the periplasm is maintained in an active state by its essential foldase, LipB. Here we report that LipA is able to recognize lipids of different chain lengths at extremes of pH and temperature, thanks to its stabilization by LipB through an extended, highly helical "embrace." A vast bioinformatic analysis indicates that LipB-like foldases are widespread over numerous proteobacteria, and thus the extended foldase architecture shown here could be widespread. These results provide new insight into A. baumannii's adaptability as a pathogen in different environments and could facilitate the development of novel antibacterials.
Collapse
Affiliation(s)
- Yuri Rafael de Oliveira Silva
- Brazilian Biosciences National Laboratory (LNBio), CNPEM, Campinas São Paulo 13084-971, Brazil; Departamento de Genética, Evolução, Microbiologia e Imunologia, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), Campinas, São Paulo 13083-970, Brazil
| | - Carlos Contreras-Martel
- University Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale (IBS), 38044 Grenoble, France
| | | | | | - Daniel Maragno Trindade
- Brazilian Biosciences National Laboratory (LNBio), CNPEM, Campinas São Paulo 13084-971, Brazil.
| | - Andréa Dessen
- University Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale (IBS), 38044 Grenoble, France.
| |
Collapse
|
19
|
Sui B, Li X, Li N, Tao Y, Wang L, Xu Y, Hou Y, Hu B, Tan D. Synergistic action of mucoactive drugs and phages against Pseudomonas aeruginosa and Klebsiella pneumoniae. Microbiol Spectr 2025; 13:e0160124. [PMID: 39912676 DOI: 10.1128/spectrum.01601-24] [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: 07/02/2024] [Accepted: 01/06/2025] [Indexed: 02/07/2025] Open
Abstract
N-acetylcysteine (NAC) and ambroxol hydrochloride (AMB) are commonly prescribed alongside antibiotics to alleviate sputum retention in lower respiratory tract infections, which are often caused by bacterial pathogens. With the rising threat of antibiotic resistance, phage therapy has emerged as a promising alternative alongside. However, no studies have explored the potential interactions between phages and these mucoactive agents despite their frequent concurrent use during phage therapy. Therefore, investigating the potential synergy and its subsequent impact on phage infection dynamics could enhance clinical strategies for treating bacterial infections with phages. Our study utilized Pseudomonas aeruginosa strain ZS-PA-35 and Klebsiella pneumoniae strain Kp36, alongside their respective phages, to investigate their interactions in the presence of NAC or AMB. Our findings indicate that, under specific conditions, these mucoactive agents can function as adjuvants to lytic phages, enhancing bacterial susceptibility to phage infection and facilitating subsequent phage proliferation. Our study revealed that these synergistic interactions are strongly influenced by the physiological characteristics of the phages, the surrounding microenvironments, and the physiology of host tissues, as varying outcomes of phage-host interactions were observed among different phages and across distinct media. Taken together, our results emphasize the complexity of interactions between phages and NAC or AMB, underscoring the need for caution when using combination treatments.IMPORTANCEN-acetylcysteine (NAC) and ambroxol hydrochloride (AMB) are used in medical treatment of patients with acute and chronic bronchitis. Often, the choice of NAC or AMB is empirically determined by physicians. However, the potential impact of combining NAC or AMB with phage therapy remains unclear. To address this gap, a comprehensive understanding of their interplay is crucial to determine any potential synergistic effects. This study aims to elucidate how NAC or AMB influence phages targeting different receptors, thereby affecting their antibacterial activity against Pseudomonas aeruginosa and Klebsiella pneumoniae. Our results suggest that, under certain conditions, NAC or AMB provides an adjuvant effect by rendering the cells more susceptible to phage infection. These results contribute to advancing our understanding of the clinical combination of mucoactive agents and phage therapy, offering insights for optimizing treatment efficacy.
Collapse
Affiliation(s)
- Bingrui Sui
- MOE Key Laboratory of Bio-Intelligent Manufacturing, School of Bioengineering, Dalian University of Technology, Dalian, China
| | - Xiaoyu Li
- MOE Key Laboratory of Bio-Intelligent Manufacturing, School of Bioengineering, Dalian University of Technology, Dalian, China
| | - Na Li
- Department of Infectious Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yang Tao
- MOE Key Laboratory of Bio-Intelligent Manufacturing, School of Bioengineering, Dalian University of Technology, Dalian, China
| | - Lili Wang
- MOE Key Laboratory of Bio-Intelligent Manufacturing, School of Bioengineering, Dalian University of Technology, Dalian, China
| | - Yongping Xu
- MOE Key Laboratory of Bio-Intelligent Manufacturing, School of Bioengineering, Dalian University of Technology, Dalian, China
| | - Yumin Hou
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Bijie Hu
- Department of Infectious Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Demeng Tan
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| |
Collapse
|
20
|
Ran Q, Yuan Y, Wu Y, Gan X, Deng J, Chu Y, Ji Q, Wang X, Zhao K. Two amino-substituted diphenyl fumaramide derivatives inhibit the virulence regulated by quorum sensing system of Pseudomonas aeruginosa. J Appl Microbiol 2025; 136:lxaf038. [PMID: 39971733 DOI: 10.1093/jambio/lxaf038] [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: 09/09/2024] [Revised: 02/06/2025] [Accepted: 02/18/2025] [Indexed: 02/21/2025]
Abstract
AIM Pseudomonas aeruginosa employs the quorum sensing (QS) system, a sophisticated cell-to-cell communication mechanism, to modulate the synthesis and secretion of a range of virulence factors, which contribute to the establishment of acute or chronic infections in hosts. This study seeks to attenuate the virulence of P. aeruginosa by inhibiting the QS system, thereby reducing its pathogenicity as a promising alternative to traditional antibiotics. METHODS AND RESULTS Two compounds with an amino-substituted diphenyl fumaramide core, N1-(4-bromophenyl)-N4-(4'-oxo-3',4'-dihydro-1'H-spiro [cyclopentane-1,2'-quinazolin]-6'-yl) fumaramide (10D) and N1-(3-chloro-4-fluorophenyl)-N4-(4-oxo-3,4,4',5'-tetrahydro-1H,2'H-spiro [quinazoline-2,3'-thiophen]-6-yl) fumaramide (12A), were identified through in-silico screening. The QS inhibitory potential of both compounds was explored in vitro and in vivo. In in vitro experiments, neither compound exhibited bactericidal effects but significantly inhibited the production of QS-regulated extracellular protease and pyocyanin. Quantitative PCR analysis revealed that QS-activated genes and downstream virulence genes were transcriptionally suppressed by 10D or 12A. Molecular docking and molecular dynamics simulations predicted stable interactions between these compounds and the key QS regulators LasR and PqsR. When combined with polymyxin B, kanamycin, and levofloxacin, 10D and 12A exhibited synergistic antibacterial activity. Furthermore, compounds 10D and 12A significantly improved the survival of mice challenged with P. aeruginosa and effectively reduced the bacterial load in the lungs. CONCLUSION This study indicates that 10D and 12A possess considerable QS inhibitory potential, effectively attenuating the pathogenicity of P. aeruginosa. Moreover, the study offers structural insights and methodological guidance for the advancement of anti-virulence drug development.
Collapse
Affiliation(s)
- Qiman Ran
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, Chengdu 610106 Sichuan, China
| | - Yang Yuan
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, Chengdu 610106 Sichuan, China
| | - Yi Wu
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, Chengdu 610106 Sichuan, China
| | - Xiongyao Gan
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, Chengdu 610106 Sichuan, China
| | - Junfeng Deng
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, Chengdu 610106 Sichuan, China
| | - Yiwen Chu
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, Chengdu 610106 Sichuan, China
| | - Qinggang Ji
- School of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, China
| | - Xinrong Wang
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, Chengdu 610106 Sichuan, China
| | - Kelei Zhao
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, Chengdu 610106 Sichuan, China
| |
Collapse
|
21
|
Wang J, Zhang L, Fu L, Pang Z. Kaempferol Mitigates Pseudomonas aeruginosa-Induced Acute Lung Inflammation Through Suppressing GSK3β/JNK/c-Jun Signaling Pathway and NF-κB Activation. Pharmaceuticals (Basel) 2025; 18:322. [PMID: 40143103 PMCID: PMC11944347 DOI: 10.3390/ph18030322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/16/2025] [Accepted: 02/11/2025] [Indexed: 03/28/2025] Open
Abstract
Background: Pseudomonas aeruginosa, one of the common bacterial pathogens causing nosocomial pneumonia, is characterized as highly pathogenic and multidrug-resistant. Kaempferol (KP), a natural flavonoid, has been shown to exhibit effectiveness in treating infection-induced lung injury. Methods: We applied network pharmacology to explore the underlying mechanisms of KP in treating P. aeruginosa pneumonia and further validated them through a mouse model of acute bacterial lung infection and an in vitro macrophage infection model. Results: The in vivo studies demonstrated that treatment with KP suppressed the production of proinflammatory cytokines, including TNF, IL-1β, IL-6, and MIP-2, and attenuated the neutrophil infiltration and lesions in lungs, leading to an increased survival rate of mice. Further studies revealed that KP treatment enhanced the phosphorylation of GSK3β at Ser9 and diminished the phosphorylation of JNK, c-Jun, and NF-κB p65 in lungs in comparison to the mice without drug treatment. Consistently, the in vitro studies showed that pretreatment with KP reduced the activation of GSK3β, JNK, c-Jun, and NF-κB p65 and decreased the levels of the proinflammatory cytokines in macrophages during P. aeruginosa infection. Conclusions: KP reduced the production of proinflammatory cytokines by inhibiting GSK3β/JNK/c-Jun signaling pathways and NF-κB activation, which effectively mitigated the P. aeruginosa-induced acute lung inflammation and injury, and elevated the survival rates of mice.
Collapse
Affiliation(s)
- Jue Wang
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China;
| | - Linlin Zhang
- Institute of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China;
| | - Lu Fu
- School of Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China;
| | - Zheng Pang
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China;
| |
Collapse
|
22
|
Drenkard E, Godfrey C, Hopke A, Thundivalappil SR, Li MC, Irimia D, Hurley BP. Pseudomonas aeruginosa aggregates elicit neutrophil swarming. iScience 2025; 28:111805. [PMID: 39967870 PMCID: PMC11834114 DOI: 10.1016/j.isci.2025.111805] [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: 07/29/2024] [Revised: 09/03/2024] [Accepted: 01/10/2025] [Indexed: 02/20/2025] Open
Abstract
Pseudomonas aeruginosa, a gram-negative multidrug-resistant (MDR) opportunist, belongs to the ESKAPE group of pathogens associated with the highest risk of mortality. Neutrophil swarming is a host defense strategy triggered by larger threats, where neutrophil swarms contain and clear damage/infection. Current ex vivo models designed to study neutrophil-pathogen interactions largely focus on individual neutrophil engagement with bacteria and fail to capture neutrophil swarming. Here, we report an ex vivo model that reproducibly elicits neutrophil swarming in response to bacterial aggregates. A rapid and robust swarming response follows engagement with pathogenic targets. Components of the type III secretion system (T3SS), a critical P. aeruginosa virulence determinant, are involved in swarm interaction. This ex vivo approach for studying neutrophil swarming in response to large pathogen targets constitutes a valuable tool for elucidating host-pathogen interaction mechanisms and for evaluating novel therapeutics to combat MDR infections.
Collapse
Affiliation(s)
- Eliana Drenkard
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston MA 02114, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Christian Godfrey
- Harvard Medical School, Boston, MA 02115, USA
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Boston, MA 02129, USA
- Shriners Hospital for Children, Boston, MA 02114, USA
| | - Alex Hopke
- Harvard Medical School, Boston, MA 02115, USA
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Boston, MA 02129, USA
- Shriners Hospital for Children, Boston, MA 02114, USA
| | - Sujatha Rajeev Thundivalappil
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston MA 02114, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Michael Chen Li
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston MA 02114, USA
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Boston, MA 02129, USA
- Shriners Hospital for Children, Boston, MA 02114, USA
| | - Daniel Irimia
- Harvard Medical School, Boston, MA 02115, USA
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Boston, MA 02129, USA
- Shriners Hospital for Children, Boston, MA 02114, USA
| | - Bryan P. Hurley
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston MA 02114, USA
- Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
23
|
Bouzada FM, Mestre B, Vaquer A, Tejada S, de la Rica R. Detecting Respiratory Pathogens for Diagnosing Lower Respiratory Tract Infections at the Point of Care: Challenges and Opportunities. BIOSENSORS 2025; 15:129. [PMID: 40136926 PMCID: PMC11940763 DOI: 10.3390/bios15030129] [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: 01/06/2025] [Revised: 02/17/2025] [Accepted: 02/19/2025] [Indexed: 03/27/2025]
Abstract
Lower respiratory tract infections (LRTIs) are a leading cause of mortality worldwide, claiming millions of lives each year and imposing significant healthcare costs. Accurate detection of respiratory pathogens is essential for the effective management of LRTIs. However, this process often relies on sputum analysis, which requires extensive pretreatment steps. The viscous nature and complex composition of sputum present additional challenges, especially in settings where a rapid diagnosis at the point of care is essential. In this review, we describe the main types of LRTI, highlighting different patient care pathway and points of care. We review current methods for liquefying sputum samples and provide an overview of current commercially available diagnostic tools used in hospitals for LRTI detection. Furthermore, we critically review recent advancements in the literature focused on detecting respiratory pathogens and mechanisms of antimicrobial resistance in sputum, including nucleic acid amplification tests, immunoassays and other innovative approaches. Throughout the paper, we highlight challenges and opportunities associated with developing new biosensor technologies tailored for detecting respiratory pathogens in lower respiratory specimens. By shedding light on these pressing issues, we aim to inspire scientific community to create innovative diagnostic tools to address the urgent healthcare burden of lung diseases.
Collapse
Affiliation(s)
- Francisco M. Bouzada
- Multidisciplinary Sepsis Group, Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain; (F.M.B.); (B.M.); (R.d.l.R.)
| | - Bartomeu Mestre
- Multidisciplinary Sepsis Group, Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain; (F.M.B.); (B.M.); (R.d.l.R.)
| | - Andreu Vaquer
- Multidisciplinary Sepsis Group, Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain; (F.M.B.); (B.M.); (R.d.l.R.)
- Department of Chemistry, University of the Balearic Islands, 07122 Palma de Mallorca, Spain
| | - Sofía Tejada
- Multidisciplinary Sepsis Group, Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain; (F.M.B.); (B.M.); (R.d.l.R.)
| | - Roberto de la Rica
- Multidisciplinary Sepsis Group, Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain; (F.M.B.); (B.M.); (R.d.l.R.)
- (CIBERINFEC)—Centro de Investigación Biomédica en Red de Enfermedades Infecciosas, Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
24
|
Nketia PB, Manu P, Osei-Poku P, Kwarteng A. Phenazine Scaffolds as a Potential Allosteric Inhibitor of LasR Protein in Pseudomonas aeruginosa. Bioinform Biol Insights 2025; 19:11779322251319594. [PMID: 39991110 PMCID: PMC11843726 DOI: 10.1177/11779322251319594] [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: 09/15/2024] [Accepted: 01/24/2025] [Indexed: 02/25/2025] Open
Abstract
Millions of individuals suffer from chronic infections caused by bacterial biofilms, resulting in significant loss of life. Pseudomonas aeruginosa stands out as a major culprit in causing such chronic infections, largely due to its antibiotic resistance. This pathogen poses a considerable threat in healthcare settings, particularly to critically ill and immunocompromised patients. The persistence of chronic and recurrent bacterial infections is often attributed to bacterial biofilms. Therefore, there is an urgent need to discover novel small molecules capable of efficiently eliminating biofilms independent of bacterial growth. In this project, an in silico drug discovery approach was employed to identify nine halogenated-phenazine compounds as allosteric inhibitors of the LasR protein. The LasR is a key transcription factor that triggers other quorum-sensing systems and plays a crucial role in biofilm formation and activation of virulence genes. By inhibiting LasR, specifically targeting its allosteric site, the dimerization of LasR and subsequent biofilm formation could be prevented. Molecular docking and simulations, coupled with binding energy calculations, identified five compounds with potential as anti-biofilm agents. These compounds exhibited higher binding affinities to the distal site, suggesting their structural capability to interact with allosteric site residues of the LasR protein. Based on these findings, it is proposed that these compounds could serve as promising leads for the treatment of biofilm and quorum-sensing-related infections.
Collapse
Affiliation(s)
- Prisca Baah Nketia
- Department of Chemistry, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
- Kumasi Centre for Collaborative Research in Tropical Medicine, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Prince Manu
- Department of Chemistry, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
- Kumasi Centre for Collaborative Research in Tropical Medicine, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Priscilla Osei-Poku
- Kumasi Centre for Collaborative Research in Tropical Medicine, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
- Department of Biochemistry and Biotechnology, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Alexander Kwarteng
- Kumasi Centre for Collaborative Research in Tropical Medicine, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
- Department of Biochemistry and Biotechnology, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| |
Collapse
|
25
|
Nortje NQ, Aribisala JO, Pillay C, Sabiu S. Molecular modelling and experimental validation of mangiferin and its related compounds as quorum sensing modulators of Pseudomonas aeruginosa. Arch Microbiol 2025; 207:53. [PMID: 39921728 PMCID: PMC11807064 DOI: 10.1007/s00203-025-04240-3] [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] [Received: 11/18/2024] [Revised: 12/29/2024] [Accepted: 01/07/2025] [Indexed: 02/10/2025]
Abstract
The LasR quorum sensing system regulates the virulence factors of Pseudomonas aeruginosa, a multi-drug resistant pathogen. Mangiferin and related compounds have been found to modulate this system as determined by in silico and in vitro experimental procedures. ZINCPharmer was used to compile a library of over 1000 metabolites that were screened to the top five based on shared pharmacophores and drug-like properties with mangiferin. Molecular docking and molecular dynamics simulation (140 ns) showed that ZINC E (- 55.64 ± 2.93 kcal/mol) and ZINC D (- 54.51 ± 2.82 kcal/mol) had significantly lower binding free energy compared to mangiferin-LasR (- 42.24 ± 3.94 kcal/mol) and the reference standard (azithromycin-LasR (- 40.01 ± 6.15 kcal/mol). ZINC D (95.16%) competed favorably with mangiferin (95.77%) as potential QS modulators at sub-minimum inhibitory concentrations relative to ZINC E (85.07%) and azithromycin (85.79%). These observations suggest mangiferin and related lead compounds as potential drug candidates for P. aeruginosa infection management.
Collapse
Affiliation(s)
- Nicolas Quinn Nortje
- Department of Biotechnology and Food Science, Faculty of Applied Sciences, Durban University of Technology, P.O. Box 1334, Durban, 4000, South Africa
| | - Jamiu Olaseni Aribisala
- Department of Biotechnology and Food Science, Faculty of Applied Sciences, Durban University of Technology, P.O. Box 1334, Durban, 4000, South Africa
| | - Charlene Pillay
- Department of Biotechnology and Food Science, Faculty of Applied Sciences, Durban University of Technology, P.O. Box 1334, Durban, 4000, South Africa.
| | - Saheed Sabiu
- Department of Biotechnology and Food Science, Faculty of Applied Sciences, Durban University of Technology, P.O. Box 1334, Durban, 4000, South Africa
| |
Collapse
|
26
|
Al-Daghistani HI, Matalqah SM, Shadid KA, Abu-Niaaj LF, Zein S, Abo-Ali RM. Quorum Quenching of P. aeruginosa by Portulaca oleracea Methanolic Extract and Its Phytochemical Profile. Pathogens 2025; 14:163. [PMID: 40005538 PMCID: PMC11858189 DOI: 10.3390/pathogens14020163] [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: 12/27/2024] [Revised: 01/19/2025] [Accepted: 01/24/2025] [Indexed: 02/27/2025] Open
Abstract
Quorum sensing (QS) is a molecular communication mechanism among bacterial cells. It is critical in regulating virulence factors, motility, antibiotic resistance, and biofilm formation. Pseudomonas aeruginosa is a Gram-negative opportunistic pathogen linked to healthcare-associated infections, food poisoning, and biofilm formation. Treating infections caused by pathogenic bacteria has become a challenge due to the development of multi-antibiotic resistance upon continuous exposure of bacteria to antibiotics. An alternative strategy to conventional antimicrobials to decrease the bacterial pathogenicity is QS inhibition, also known as quorum quenching. Using plant-derived compounds is an environmentally friendly strategy to block the bacterial QS and inhibit bacterial growth. Portulaca oleracea is a popular plant in different countries and is also used in traditional medicine. It is widely consumed raw in salads and as garnishes, though it can be cooked as a vegetarian dish. This study evaluates the antimicrobial activity of the methanolic extract of P. oleracea and its effectiveness in blocking or attenuating the QS of P. aeruginosa. The agar well diffusion method used for screening the antibacterial activity showed a significant growth inhibition of P. aeruginosa by the extract at 500 mg/mL with a minimum inhibitory concentration of 31.25 mg/mL. A bioindicator bacterium, Chromobacterium violaceum CV026, was used to determine the effect of the methanolic extract on the QS of P. aeruginosa. The results indicated a significant reduction in biofilm formation, pyocyanin production, and LasA staphylolytic activity. The phytochemical analysis by Gas Chromatography-Mass Spectrometry showed that the methanolic extract contained several phenols, alkaloids, esters, and other compounds previously reported to have antibacterial and antioxidant effects. These findings highlight the effectiveness of P. oleracea methanolic extract in attenuating the QS and virulence factors of P. aeruginosa. This study suggests that P. oleracea is an important source of natural antimicrobials and its use would be beneficial in food and pharmaceutical applications.
Collapse
Affiliation(s)
- Hala I. Al-Daghistani
- Department of Medical Laboratory Sciences, Faculty of Allied Medical Sciences, Al-Ahliyya Amman University, Amman 19328, Jordan;
| | - Sina M. Matalqah
- Pharmacological and Diagnostic Research Centre, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman 19328, Jordan; (S.M.M.); (K.A.S.)
| | - Khalid A. Shadid
- Pharmacological and Diagnostic Research Centre, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman 19328, Jordan; (S.M.M.); (K.A.S.)
| | - Lubna F. Abu-Niaaj
- Department of Agricultural and Life Sciences, College of Engineering, Science, Technology and Agriculture, Central State University, Wilberforce, OH 45384, USA
| | - Sima Zein
- Department of Pharmaceutical Biotechnology, Faculty of Allied Medical Sciences, Al-Ahliyya Amman University, Amman 19328, Jordan;
| | - Raeda M. Abo-Ali
- Faculty of Nursing, Al-Balqa Applied University, Amman 19117, Jordan;
| |
Collapse
|
27
|
Shehu K, Schneider M, Kraegeloh A. Menadione as Antibiotic Adjuvant Against P. aeruginosa: Mechanism of Action, Efficacy and Safety. Antibiotics (Basel) 2025; 14:163. [PMID: 40001407 PMCID: PMC11851977 DOI: 10.3390/antibiotics14020163] [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: 01/11/2025] [Revised: 01/29/2025] [Accepted: 02/04/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Antibiotic resistance in chronic lung infections caused by Pseudomonas aeruginosa requires alternative approaches to improve antibiotic efficacy. One promising approach is the use of adjuvant compounds that complement antibiotic therapy. This study explores the potential of menadione as an adjuvant to azithromycin against planktonic cells and biofilms of P. aeruginosa, focusing on its mechanisms of action and cytotoxicity in pulmonary cell models. Methods: The effect of menadione in improving the antibacterial and antibiofilm potency of azithromycin was tested against P. aeruginosa. Mechanistic studies in P. aeruginosa and AZMr-E. coli DH5α were performed to probe reactive oxygen species (ROS) production and bacterial membrane disruption. Cytotoxicity of antibacterial concentrations of menadione was assessed by measuring ROS levels and membrane integrity in Calu-3 and A549 lung epithelial cells. Results: Adding 0.5 µg/mL menadione to azithromycin reduced the minimum inhibitory concentration (MIC) by four-fold and the minimum biofilm eradication concentration (MBEC) by two-fold against P. aeruginosa. Adjuvant mechanisms of menadione involved ROS production and disruption of bacterial membranes. Cytotoxicity tests revealed that antibacterial concentrations of menadione (≤64 µg/mL) did not affect ROS levels or membrane integrity in lung cell lines. Conclusions: Menadione enhanced the efficacy of azithromycin against P. aeruginosa while exhibiting a favorable safety profile in lung epithelial cells at antibacterial concentrations. These findings suggest that menadione is a promising antibiotic adjuvant. However, as relevant data on the toxicity of menadione is sparse, further toxicity studies are required to ensure its safe use in complementing antibiotic therapy.
Collapse
Affiliation(s)
- Kristela Shehu
- Department of Pharmacy, Biopharmaceutics & Pharmaceutical Technology, Saarland University, 66123 Saarbrücken, Germany;
- INM—Leibniz Institute for New Materials, 66123 Saarbrücken, Germany
| | - Marc Schneider
- Department of Pharmacy, Biopharmaceutics & Pharmaceutical Technology, Saarland University, 66123 Saarbrücken, Germany;
| | | |
Collapse
|
28
|
Upadhyay A, Jaiswal N, Kumar A. Biofilm battle: New transformative tactics to tackle the bacterial biofilm infections. Microb Pathog 2025; 199:107277. [PMID: 39756524 DOI: 10.1016/j.micpath.2025.107277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/28/2024] [Accepted: 01/02/2025] [Indexed: 01/07/2025]
Abstract
Bacterial biofilm infections are the root cause of persistent infections and the prevalence of resistance to specific or multiple antibiotics. Biofilms have unique features that provide a protective environment for bacteria under various stress conditions and contribute significantly to the pathogenesis of chronic infections. They cover bacterial cells with a self-produced extracellular polymeric matrix, effectively hiding the bacterial cells and their targets. Conventional therapies cannot effectively treat and control bacterial biofilm infections. Therefore, advanced therapeutic means like microneedles, targeted tissue therapy, phage therapy, nanodrug therapy, combination drug therapy, microbial therapy, and immune cell hijacking therapy are needed to tackle the complex issue. These advanced therapies have shown promising results not only in bacterial biofilm infections but also in diseases such as cancer and genetic disorders. Due to their unique features and mechanisms, they significantly contribute to preventing bacterial infections by disrupting biofilm. This article aims to serve as a comprehensive overview of the ongoing battle against biofilms with transformative therapies. This article compiles advancements in new therapies that have demonstrated effective roles in the disruption of bacterial biofilms. We also discuss the current developments and Food and Drug Administration-approved status of these therapies. Additionally, this article summarizes the limitations and future steps needed for these therapies in the field of bacterial biofilm prevention. Thus, these therapies represent the future of preventing bacterial biofilm infections and could be also effective in the reversal of resistance.
Collapse
Affiliation(s)
- Aditya Upadhyay
- Department of Biotechnology, National Institute of Technology, Raipur, 492010, (CG), India
| | - Neha Jaiswal
- Department of Biotechnology, National Institute of Technology, Raipur, 492010, (CG), India
| | - Awanish Kumar
- Department of Biotechnology, National Institute of Technology, Raipur, 492010, (CG), India.
| |
Collapse
|
29
|
Ezzariga N, Zouhari O, Rhars A, Lemkhente Z, Aghrouch M. Biofilm and Antibiotic Resistance Study of Bacteria Involved in Nosocomial Infections. Cureus 2025; 17:e78673. [PMID: 39926624 PMCID: PMC11804273 DOI: 10.7759/cureus.78673] [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] [Accepted: 02/07/2025] [Indexed: 02/11/2025] Open
Abstract
Nosocomial infections are increasingly problematic due to growing bacterial resistance. Biofilms play a key role in the persistence of these infections, leading to treatment failures and poor patient outcomes. Addressing antibiotic resistance within biofilms is especially critical in hospitals, making it essential to develop new strategies to manage biofilm-related infections and curb bacterial resistance. The study, conducted at the regional hospital center in Agadir, Morocco, analyzed 75 bacteria (37 antibiotic-sensitive and 38 resistant). Seven bacteria were isolated from catheters, and others from preserved samples. Biofilm formation was assessed using the tissue culture plate (TCP) method, involving strain recovery; culture on cystine, lactose, electrolyte-deficient (CLED) medium; microplate inoculation; staining with crystal violet; and optical density (OD) measurement. The results showed that 77.33% of the bacteria formed biofilms. All catheter-isolated bacteria showed biofilm formation. Strong biofilm production was observed in 66.67% of Acinetobacter baumannii and in most Pseudomonas aeruginosa strains. Enterobacteriaceae also demonstrated significant biofilm formation. Notably, 70% of carbapenem-resistant bacteria showed strong biofilm production. Most nosocomial bacteria form biofilms, with a higher prevalence in antibiotic-resistant strains. Sensitive bacteria also form biofilms but less frequently. Bacterial conjugation may facilitate the acquisition of carbapenem resistance within biofilms.
Collapse
Affiliation(s)
- Nihal Ezzariga
- Bacteriology, Faculty of Medicine and Pharmacy, Ibn Zohr University, Agadir, MAR
| | - Oumaima Zouhari
- Nursing, Higher Institute of Nursing Professions and Health Techniques, Agadir, MAR
| | - Amal Rhars
- Parasitology and Mycology, Faculty of Medicine and Pharmacy, Ibn Zohr University, Agadir, MAR
| | - Zohra Lemkhente
- Parasitology and Mycology, Faculty of Medicine and Pharmacy, Ibn Zohr University, Agadir, MAR
| | - Mohamed Aghrouch
- Medical Biology, Centre Hospitalier Régional Hassan II, Agadir, MAR
| |
Collapse
|
30
|
Li DD, Wang Y, Li H, Niu WX, Hong J, Jung JH, Lee JH. Multifaceted Antipathogenic Activity of Two Novel Natural Products, Chermesiterpenoid B and Chermesiterpenoid B Seco Acid Methyl Ester, Against Pseudomonas aeruginosa. Microb Biotechnol 2025; 18:e70101. [PMID: 39936740 DOI: 10.1111/1751-7915.70101] [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: 07/07/2024] [Revised: 01/22/2025] [Accepted: 01/22/2025] [Indexed: 02/13/2025] Open
Abstract
Pseudomonas aeruginosa is an opportunistic human pathogen that causes both acute and chronic infections due to its virulence factors, biofilm formation and the ability to suppress the host immune system. Quorum sensing (QS) plays a key role in regulating these pathogenic traits and also downregulates the expression of peroxisome proliferator-activated receptor-γ (PPAR-γ) in host cells. In this study, we isolated two novel natural products from the jellyfish-derived fungus Penicillium chermesinum, chermesiterpenoid B (Che B) seco acid methyl ester (Che B ester) and Che B. Both compounds act as partial agonists of PPAR-γ and exhibit anti-QS activity. Che B ester and Che B were found to inhibit biofilm formation, reduce the production of proteases and decrease the infectivity of P. aeruginosa, all without affecting bacterial growth. In host cells, Che B ester and Che B reduced P. aeruginosa-induced inflammation by activating PPAR-γ. This multifaceted function makes these compounds promising candidates for developing new antipathogenic agents against bacterial infections with few side effects.
Collapse
Affiliation(s)
- Dan-Dan Li
- College of Pharmacy, Pusan National University, Busan, Republic of Korea
- Research Institute for Drug Development, Pusan National University, Busan, Republic of Korea
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Ying Wang
- College of Pharmacy, Pusan National University, Busan, Republic of Korea
| | - Huiyan Li
- College of Pharmacy, Pusan National University, Busan, Republic of Korea
- Research Institute for Drug Development, Pusan National University, Busan, Republic of Korea
| | - Wen-Xin Niu
- College of Pharmacy, Pusan National University, Busan, Republic of Korea
- Research Institute for Drug Development, Pusan National University, Busan, Republic of Korea
| | - Jongki Hong
- College of Pharmacy, Kyung Hee University, Seoul, Republic of Korea
| | - Jee H Jung
- College of Pharmacy, Pusan National University, Busan, Republic of Korea
- Research Institute for Drug Development, Pusan National University, Busan, Republic of Korea
| | - Joon-Hee Lee
- College of Pharmacy, Pusan National University, Busan, Republic of Korea
- Research Institute for Drug Development, Pusan National University, Busan, Republic of Korea
| |
Collapse
|
31
|
Zhang K, Huang Y, Jiang Y, Liu T, Kong J, Cai S, Wen Z, Chen Y. Effect of Candida albicans' supernatant on biofilm formation and virulence factors of Pseudomonas aeruginosa through las/rhl System. BMC Microbiol 2025; 25:60. [PMID: 39893414 PMCID: PMC11786564 DOI: 10.1186/s12866-024-03604-x] [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: 01/19/2024] [Accepted: 10/24/2024] [Indexed: 02/04/2025] Open
Abstract
Pseudomonas aeruginosa (P. aeruginosa) and Candida albicans (C. albicans) are opportunistic pathogens whose mixed infections can exacerbate microbial dissemination and drug resistance, contributing to high mortality and morbidity rates among infected individuals. Few studies have explored the impact of C. albicans supernatant on P. aeruginosa, and the underlying mechanisms of such mixed infections remain unclear. In this study, we investigated the effects of C. albicans supernatant on biofilm formation and virulence factor activity in wild-type P. aeruginosa PAO1 and its quorum sensing-deficient mutants, ΔlasIrhlI and ΔlasRrhlR. Our results demonstrated that the biofilm formation capability and virulence were significantly higher in the PAO1 group compared to the ΔlasIrhlI and ΔlasRrhlR groups. Furthermore, exposure to C. albicans supernatant significantly enhanced both the biofilm formation and virulence of PAO1, whereas no significant changes were observed in the ΔlasIrhlI and ΔlasRrhlR mutants relative to their respective controls. These findings suggest that C. albicans supernatant may modulate P. aeruginosa biofilm formation and virulence via the las/rhl quorum sensing system.
Collapse
Affiliation(s)
- Ke Zhang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Qingxiu District, Nanning City, Guangxi Zhuang Autonomous Region, 530021, China
| | - Yingying Huang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Qingxiu District, Nanning City, Guangxi Zhuang Autonomous Region, 530021, China
| | - Yuting Jiang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Qingxiu District, Nanning City, Guangxi Zhuang Autonomous Region, 530021, China
| | - Tangjuan Liu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Qingxiu District, Nanning City, Guangxi Zhuang Autonomous Region, 530021, China
| | - Jinliang Kong
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Qingxiu District, Nanning City, Guangxi Zhuang Autonomous Region, 530021, China
| | - Shuangqi Cai
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Qingxiu District, Nanning City, Guangxi Zhuang Autonomous Region, 530021, China
| | - Zhongwei Wen
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Qingxiu District, Nanning City, Guangxi Zhuang Autonomous Region, 530021, China
| | - Yiqiang Chen
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Qingxiu District, Nanning City, Guangxi Zhuang Autonomous Region, 530021, China.
| |
Collapse
|
32
|
Golan N, Parizat A, Tabachnikov O, Barnea E, Olsen WP, Otzen DE, Landau M. Resilience and charge-dependent fibrillation of functional amyloid: Interactions of Pseudomonas biofilm-associated FapB and FapC amyloids. J Biol Chem 2025; 301:108096. [PMID: 39706277 PMCID: PMC11787515 DOI: 10.1016/j.jbc.2024.108096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 11/21/2024] [Accepted: 12/09/2024] [Indexed: 12/23/2024] Open
Abstract
FapC and FapB are biofilm-associated amyloids involved in the virulence of Pseudomonas and other bacteria. We herein demonstrate their exceptional thermal and chemical resilience, suggesting that their biofilm structures might withstand standard sterilization, thereby contributing to the persistence of Pseudomonas aeruginosa infections. Our findings also underscore the impact of environmental factors on functional amyloid in Pseudomonas (Fap) proteins, suggesting that orthologs in different Pseudomonas strains adapt to specific environments and roles. Challenging previous assumptions about a simple nucleation role for FapB in promoting FapC aggregation, the study shows a significant influence of FapC on FapB aggregation. The interaction between these FapB and FapC is intricate: FapB stabilizes FapC fibrils, while FapC slows down FapB fibrillation but can still serve as a cross-seeding template. This complex interplay is the key to understanding their roles in bacterial biofilms. Furthermore, the study highlights distinct differences between Fap and Escherichia coli's CsgA (curli) amyloid, where CsgB assumes a simple unidirectional role in nucleating CsgA fibrillation, emphasizing the importance of a comprehensive understanding of various amyloid systems. This knowledge is vital for developing effective intervention strategies against bacterial infections and leveraging the unique properties of these amyloids in technological applications such as novel bionanomaterials or protective coatings.
Collapse
Affiliation(s)
- Nimrod Golan
- Department of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Amit Parizat
- Department of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Orly Tabachnikov
- Department of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Eilon Barnea
- Department of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - William P Olsen
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus C, Denmark
| | - Daniel E Otzen
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus C, Denmark
| | - Meytal Landau
- Department of Biology, Technion-Israel Institute of Technology, Haifa, Israel; CSSB Centre for Structural Systems Biology, Deutsches Elektronen-Synchrotron DESY, Hamburg, Germany; The Center for Experimental Medicine, Universitätsklinikum Hamburg-Eppendorf (UKE), Hamburg, Germany; European Molecular Biology Laboratory (EMBL), Hamburg, Germany.
| |
Collapse
|
33
|
Wu J, Thompson TP, O'Connell NH, McCracken K, Powell J, Gilmore BF, Dunne CP, Kelly SA. Extended-spectrum β-lactamase-producing bacteria from hospital wastewater pipes: isolation, characterization and biofilm control using common disinfectants. J Hosp Infect 2025; 156:34-49. [PMID: 39586542 DOI: 10.1016/j.jhin.2024.11.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/17/2024] [Accepted: 11/09/2024] [Indexed: 11/27/2024]
Abstract
BACKGROUND Hospital wastewater systems have been identified as reservoirs for antibiotic-resistant bacteria, with biofilms harbouring extended-spectrum β-lactamase (ESBL)-producing micro-organisms posing significant infection risk. AIM To study the antimicrobial susceptibility and biofilm control of ESBL-producing bacteria from wastewater pipes from a tertiary care teaching hospital in Ireland, which had experienced endemic infection outbreaks caused by ESBL-producing bacteria. METHODS Following isolation of ESBL producers on selective agar, antibiotic susceptibility profiles were determined for a number of antibiotics assessed for their ability to form biofilms. Biofilm eradication studies using the commercially available disinfectants bleach, Optizan™, Virkon™ and Clinell™ were performed on selected isolates. FINDINGS ESBL-producing bacteria (N = 39 isolates) showed a high degree of resistance to β-lactams. Biofilm-forming ability ranged from non-adherent to strongly adherent and appeared to be source dependent, suggesting that the characteristics of the pipe environment played an important role in biofilm formation. All disinfectants showed effective biofilm eradication under suggested working conditions. Effectiveness was significantly reduced following reductions in concentration and contact time, with only Clinell™ showing significant biofilm reduction against all isolates at all concentrations and contact times tested. Of the chlorine-based formulations, Optizan™ frequently outperformed bleach at lower concentrations and treatment times. Biofilm eradication was strain dependent, with varying disinfectant response profiles observed from biofilms from different Stenotrophomonas maltophilia isolates. CONCLUSIONS This study highlights the high degree of ESBL-producing bacteria recovery from patient-facing hospital wastewater apparatus. Their ability to form resident biofilms and act as potential reservoirs of infection emphasizes the need for rigorous and effective infection control practices.
Collapse
Affiliation(s)
- J Wu
- School of Pharmacy, Queen's University Belfast, Belfast, UK
| | - T P Thompson
- School of Pharmacy, Queen's University Belfast, Belfast, UK
| | - N H O'Connell
- Microbiology Department, University Hospital Limerick, Limerick, Ireland; School of Medicine and Centre for Interventions in Infection, Inflammation, and Immunity (4i), University of Limerick, Limerick, Ireland
| | - K McCracken
- Keith McCracken Consulting Limited, Greencastle, Co. Donegal, Ireland
| | - J Powell
- Microbiology Department, University Hospital Limerick, Limerick, Ireland; School of Medicine and Centre for Interventions in Infection, Inflammation, and Immunity (4i), University of Limerick, Limerick, Ireland
| | - B F Gilmore
- School of Pharmacy, Queen's University Belfast, Belfast, UK; School of Medicine and Centre for Interventions in Infection, Inflammation, and Immunity (4i), University of Limerick, Limerick, Ireland
| | - C P Dunne
- School of Pharmacy, Queen's University Belfast, Belfast, UK; School of Medicine and Centre for Interventions in Infection, Inflammation, and Immunity (4i), University of Limerick, Limerick, Ireland
| | - S A Kelly
- School of Pharmacy, Queen's University Belfast, Belfast, UK; School of Medicine and Centre for Interventions in Infection, Inflammation, and Immunity (4i), University of Limerick, Limerick, Ireland.
| |
Collapse
|
34
|
Felton SM, Akula N, Kolling GL, Azadi P, Black I, Kumar A, Heiss C, Capobianco J, Uknalis J, Papin JA, Berger BW. Applying a polysaccharide lyase from Stenotrophomonas maltophilia to disrupt alginate exopolysaccharide produced by Pseudomonas aeruginosa clinical isolates. Appl Environ Microbiol 2025; 91:e0185324. [PMID: 39670718 PMCID: PMC11784403 DOI: 10.1128/aem.01853-24] [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: 09/18/2024] [Accepted: 11/11/2024] [Indexed: 12/14/2024] Open
Abstract
Pseudomonas aeruginosa is considered one of the most challenging, drug-resistant, opportunistic pathogens partly due to its ability to synthesize robust biofilms. Biofilm is a mixture of extracellular polymeric substances (EPS) that encapsulates microbial cells, leading to immune evasion, antibiotic resistance, and thus higher risk of infection. In the cystic fibrosis lung environment, P. aeruginosa undergoes a mucoid transition, defined by overproduction of the exopolysaccharide alginate. Alginate encapsulation results in bacterial resistance to antibiotics and the host immune system. Given its role in airway inflammation and chronic infection, alginate is an obvious target to improve treatment for P. aeruginosa infection. Previously, we demonstrated polysaccharide lyase Smlt1473 from Stenotrophomonas maltophilia strain k279a can catalyze the degradation of multiple polyuronides in vitro, including D-mannuronic acid (poly-ManA). Poly-ManA is a major constituent of P. aeruginosa alginate, suggesting that Smlt1473 could have potential application against multidrug-resistant P. aeruginosa and perhaps other microbes with related biofilm composition. In this study, we demonstrate that Smlt1473 can inhibit and degrade alginate from P. aeruginosa. Additionally, we show that tested P. aeruginosa strains are dominant in acetylated alginate and that all but one have similar M-to-G ratios. These results indicate that variation in enzyme efficacy among the isolates is not primarily due to differences in total EPS or alginate chemical composition. Overall, these results demonstrate Smlt1473 can inhibit and degrade P. aeruginosa alginate and suggest that other factors including rate of EPS production, alginate sequence/chain length, or non-EPS components may explain differences in enzyme efficacy. IMPORTANCE Pseudomonas aeruginosa is a major opportunistic human pathogen in part due to its ability to synthesize biofilms that confer antibiotic resistance. Biofilm is a mixture of polysaccharides, DNA, and proteins that encapsulate cells, protecting them from antibiotics, disinfectants, and other cleaning agents. Due to its ability to increase antibiotic and immune resistance, the exopolysaccharide alginate plays a large role in airway inflammation and chronic P. aeruginosa infection. As a result, colonization with P. aeruginosa is the leading cause of morbidity and mortality in CF patients. Thus, it is an obvious target to improve the treatment regimen for P. aeruginosa infection. In this study, we demonstrate that polysaccharide lyase, Smlt1473, inhibits alginate secretion and degrades established alginate from a variety of mucoid P. aeruginosa clinical isolates. Additionally, Smlt1473 differs from other alginate lyases in that it is active against acetylated alginate, which is secreted during chronic lung infection. These results suggest that Smlt1473 may be useful in treating infections associated with alginate-producing P. aeruginosa, as well as have the potential to reduce P. aeruginosa EPS in non-clinical settings.
Collapse
Affiliation(s)
- Samantha M. Felton
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Nikki Akula
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Glynis L. Kolling
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Parastoo Azadi
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - Ian Black
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - Ambrish Kumar
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - Christian Heiss
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - Joseph Capobianco
- United States Department of Agriculture (USDA), Agricultural Research Service (ARS), Eastern Regional Research Center, Wyndmoor, Pennsylvania, USA
| | - Joseph Uknalis
- United States Department of Agriculture (USDA), Agricultural Research Service (ARS), Eastern Regional Research Center, Wyndmoor, Pennsylvania, USA
| | - Jason A. Papin
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Bryan W. Berger
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
- Department of Chemical Engineering, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
35
|
Gu X, Wu Q, Chai Y, Huang X, Zhou X, Han M, Wu T, Zhang X, Zhong F. Epidemiological and molecular characteristics of extraintestinal pathogenic escherichia coli isolated from diseased cattle and sheep in Xinjiang, China from 2015 to 2019. BMC Vet Res 2025; 21:42. [PMID: 39885526 PMCID: PMC11783789 DOI: 10.1186/s12917-025-04502-8] [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: 10/20/2024] [Accepted: 01/16/2025] [Indexed: 02/01/2025] Open
Abstract
Escherichia coli has become a common causative agent of infections in animals, inflicting serious economic losses on livestock production and posing a threat to public health. Escherichia coli infection is common and tends to be complex in Xinjiang, a major region of cattle and sheep breeding in China. This study aims to explore the current status and molecular characteristics of Escherichia coli infection in cattle and sheep in Xinjiang, as part of the disease prevention and control strategy. Herein we isolated Extraintestinal pathogenic Escherichia coli (ExPEC) from the liver, spleen, lung, heart, and lymph nodes of infected cattle and sheep (Xinjiang, China), and phylogenetic grouping, serotyping, and multilocus sequence typing were performed to determine epidemic and molecular characteristics. We also assessed their biofilm formation ability. A total of 132 strains of ExPEC were identified from diseased cattle and sheep, belonging to 7 phylogenetic groups. A and B1 are advantageous groups. Further, 22 serogroups were found, with O101 (26/132), O154 (14/132), and O65 (8/132) being the predominant ones. Among the seven sequence types identified by multilocus sequence typing, ST10 was the most common, followed by ST23 and ST457. Of 132, 105 (79.5%) strains were able to form biofilms: 15 strains (11.4%) were strong, 28 (21.2%) were medium, and 62 (47%) were weak biofilm producers. These findings will contribute to a better understanding of the molecular epidemiology of ExPEC in Xinjiang, China, and can be applied to the development, prevention, and disease control of future diagnostic tools and vaccine.
Collapse
Affiliation(s)
- Xiaoxiao Gu
- College of Animal Science and Technology, Shihezi University, Shihezi, 832003, Xinjiang, China
| | - Qin Wu
- College of Animal Science and Technology, Shihezi University, Shihezi, 832003, Xinjiang, China
| | - Yingjin Chai
- College of Animal Science and Technology, Shihezi University, Shihezi, 832003, Xinjiang, China
| | - Xin Huang
- State Key Laboratory for Sheep Genetic Improvement and Healthy Production, Xinjiang Academy of Agricultural and Reclamation Science, Shihezi, 832000, Xinjiang, China.
| | - Xia Zhou
- College of Animal Science and Technology, Shihezi University, Shihezi, 832003, Xinjiang, China.
| | - Mengli Han
- State Key Laboratory for Sheep Genetic Improvement and Healthy Production, Xinjiang Academy of Agricultural and Reclamation Science, Shihezi, 832000, Xinjiang, China
| | - Tongzhong Wu
- State Key Laboratory for Sheep Genetic Improvement and Healthy Production, Xinjiang Academy of Agricultural and Reclamation Science, Shihezi, 832000, Xinjiang, China
| | - Xingxing Zhang
- State Key Laboratory for Sheep Genetic Improvement and Healthy Production, Xinjiang Academy of Agricultural and Reclamation Science, Shihezi, 832000, Xinjiang, China
| | - Fagang Zhong
- State Key Laboratory for Sheep Genetic Improvement and Healthy Production, Xinjiang Academy of Agricultural and Reclamation Science, Shihezi, 832000, Xinjiang, China
| |
Collapse
|
36
|
Hernández-Benítez JA, Santos-Ocampo BN, Rosas-Ramírez DG, Bautista-Hernández LA, Bautista-de Lucio VM, Pérez NO, Rodríguez-Tovar AV. The Effect of Temperature over the Growth and Biofilm Formation of the Thermotolerant Aspergillus flavus. J Fungi (Basel) 2025; 11:53. [PMID: 39852472 PMCID: PMC11766932 DOI: 10.3390/jof11010053] [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: 11/21/2024] [Revised: 01/04/2025] [Accepted: 01/07/2025] [Indexed: 01/26/2025] Open
Abstract
Aspergillus flavus is a medically relevant fungus, particularly in tropical regions. Although its aflatoxin production and thermotolerance are well documented, its biofilm-forming ability has received less attention, despite being a key factor in the virulence of A. flavus as an opportunistic pathogen, which can significantly impact therapeutic outcomes. To investigate the influence of temperature on the growth and biofilm formation of an A. flavus isolate, we compared it on solid media with the reference strain A. flavus ATCC 22546 and documented morphological changes during conidial germination. We examined biofilm formation in both strains across different temperatures and evaluated the susceptibility of this A. flavus isolate to antifungal agents in both planktonic and biofilm form. Our results showed that the temperature can promote conidiation on solid media. Radial growth was highest at 28 °C, while the conidial count and density were favored at higher temperatures. Moreover, we determined that 37 °C was the optimal temperature for conidial germination and biofilm formation. We described four distinct phases in A. flavus biofilm development-initiation (0-12 h), consolidation (12-24 h), maturation (24-48 h), and dispersion (48-72 h)-with the notable presence of conidial heads at 42 °C. Carbohydrates and proteins constitute the primary components of the extracellular matrix. We observed an abundance of lipid droplets within the hyphae of the MMe18 strain biofilm. The mature biofilms demonstrated reduced susceptibility to amphotericin B and itraconazole, requiring higher inhibitory concentrations for both antifungals compared with their planktonic counterparts.
Collapse
Affiliation(s)
- José Alejandro Hernández-Benítez
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prol. Carpio y Plan de Ayala s/n Col. Casco de Santo Tomás, Alcaldia Miguel Hidalgo, Mexico City C.P. 11340, Mexico; (J.A.H.-B.); (B.N.S.-O.)
| | - Brenda Nallely Santos-Ocampo
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prol. Carpio y Plan de Ayala s/n Col. Casco de Santo Tomás, Alcaldia Miguel Hidalgo, Mexico City C.P. 11340, Mexico; (J.A.H.-B.); (B.N.S.-O.)
| | - Daniel Genaro Rosas-Ramírez
- Departamento de Química de Biomacromoléculas, Instituto de Química, Universidad Nacional Autónoma de México, Av. Universidad 3000, Circuito Exterior s/n, Ciudad Universitaria, Alcaldía Coyoacán, Mexico City C.P. 04510, Mexico;
| | - Luis Antonio Bautista-Hernández
- Unidad de Investigación del Instituto de Oftalmología, Fundación de Asistencia Privada Conde de Valenciana I.A.P., Chimalpopoca 14, Col. Obrera, Alcaldía Cuahutémoc, Mexico City C.P. 06800, Mexico; (L.A.B.-H.); (V.M.B.-d.L.)
| | - Víctor Manuel Bautista-de Lucio
- Unidad de Investigación del Instituto de Oftalmología, Fundación de Asistencia Privada Conde de Valenciana I.A.P., Chimalpopoca 14, Col. Obrera, Alcaldía Cuahutémoc, Mexico City C.P. 06800, Mexico; (L.A.B.-H.); (V.M.B.-d.L.)
| | - Néstor Octavio Pérez
- Departamento de Investigación y Desarrollo, Probiomed, S.A. de C.V., Cruce de Carreteras Acatzingo-Zumahuacan s/n, Tenancingo C.P. 52400, State of Mexico, Mexico
| | - Aída Verónica Rodríguez-Tovar
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prol. Carpio y Plan de Ayala s/n Col. Casco de Santo Tomás, Alcaldia Miguel Hidalgo, Mexico City C.P. 11340, Mexico; (J.A.H.-B.); (B.N.S.-O.)
| |
Collapse
|
37
|
Tahmasebi H, Arjmand N, Monemi M, Babaeizad A, Alibabaei F, Alibabaei N, Bahar A, Oksenych V, Eslami M. From Cure to Crisis: Understanding the Evolution of Antibiotic-Resistant Bacteria in Human Microbiota. Biomolecules 2025; 15:93. [PMID: 39858487 PMCID: PMC11764262 DOI: 10.3390/biom15010093] [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: 12/08/2024] [Revised: 01/01/2025] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
The growing prevalence of antibiotic-resistant bacteria within the human microbiome has become a pressing global health crisis. While antibiotics have revolutionized medicine by significantly reducing mortality and enabling advanced medical interventions, their misuse and overuse have led to the emergence of resistant bacterial strains. Key resistance mechanisms include genetic mutations, horizontal gene transfer, and biofilm formation, with the human microbiota acting as a reservoir for antibiotic resistance genes (ARGs). Industrialization and environmental factors have exacerbated this issue, contributing to a rise in infections with multidrug-resistant (MDR) bacteria, such as methicillin-resistant Staphylococcus aureus (MRSA) and carbapenem-resistant Enterobacteriaceae. These resistant pathogens compromise the effectiveness of essential treatments like surgical prophylaxis and chemotherapy, increase healthcare costs, and prolong hospital stays. This crisis highlights the need for a global One-Health approach, particularly in regions with weak regulatory frameworks. Innovative strategies, including next-generation sequencing (NGS) technologies, offer promising avenues for mitigating resistance. Addressing this challenge requires coordinated efforts, encompassing research, policymaking, public education, and antibiotic stewardship, to safeguard current antibiotics and foster the development of new therapeutic solutions. An integrated, multidimensional strategy is essential to tackle this escalating problem and ensure the sustainability of effective antimicrobial treatments.
Collapse
Affiliation(s)
- Hamed Tahmasebi
- School of Medicine, Shahroud University of Medical Sciences, Shahroud 36147-73943, Iran
| | - Neda Arjmand
- Department of Obstetrics and Gynecology, Tehran Medical University, Tehran 14167-53955, Iran
| | - Marzieh Monemi
- Department of Basic Science, Faculty of Pharmacy and Pharmaceutical Science, Tehran Medical Science, Islamic Azad University, Tehran 19395-1495, Iran
| | - Ali Babaeizad
- Student Research Committee, Semnan University of Medical Sciences, Semnan 35147-99442, Iran
| | - Farnaz Alibabaei
- Student Research Committee, Semnan University of Medical Sciences, Semnan 35147-99442, Iran
| | - Negar Alibabaei
- Student Research Committee, Mazandaran University of Medical Sciences, Sari 48157-33971, Iran
| | - Aisa Bahar
- Department of Biochemistry, Semnan University of Medical Sciences, Semnan 35147-99442, Iran
| | | | - Majid Eslami
- Cancer Research Center, Semnan University of Medical Sciences, Semnan 35147-99442, Iran
- Department of Bacteriology and Virology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan 35147-99442, Iran
| |
Collapse
|
38
|
Maiga A, Ampomah-Wireko M, Li H, Fan Z, Lin Z, Zhen H, Kpekura S, Wu C. Multidrug-resistant bacteria quorum-sensing inhibitors: A particular focus on Pseudomonasaeruginosa. Eur J Med Chem 2025; 281:117008. [PMID: 39500066 DOI: 10.1016/j.ejmech.2024.117008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 10/17/2024] [Accepted: 10/27/2024] [Indexed: 12/02/2024]
Abstract
Many widely used conventional antibiotics have failed to show clinical efficacy against Pseudomonas aeruginosa (P. aeruginosa) due to the strain's rising resistance to most clinically relevant antimicrobials. P. aeruginosa uses quorum sensing to regulate its virulence and biofilm development, which contributes to its pathogenicity and drug resistance. This mechanism is responsible for the resurgence and persistence of infections. Therefore, targeting the virulence and pathogenicity of P. aeruginosa through quorum sensing (QS) is regarded as a potential target for anti-infective therapy. However, a number of natural and synthetic compounds have been demonstrated to interfere with quorum sensing, resulting in potential antibacterial agents. In this review, we discuss the mechanisms of P. aeruginosa QS and recent advances in the development of quorum sensing inhibitors (both synthetic and natural) that have the potential to become effective antibiotics.
Collapse
Affiliation(s)
- Aichata Maiga
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Maxwell Ampomah-Wireko
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Hongteng Li
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Zhengmin Fan
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Ziwei Lin
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Haojie Zhen
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Stephen Kpekura
- School of Nursing and Health, Ministry of Education of China, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Chunli Wu
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou, 450001, PR China; Zhengzhou Key Laboratory of New Veterinary Drug Preparation Innovation, Zhengzhou, 450001, PR China; Henan Qunbo Pharmaceutical Research Institute Co. LTD, PR China.
| |
Collapse
|
39
|
Pajaro-Castro N, Diaz-Morales E, Hoyos K, Ibañez-Bersinger C. Whole-Genome Sequencing of Resistance, Virulence and Regulation Genes in Extremely Resistant Strains of Pseudomonas aeruginosa. Med Sci (Basel) 2025; 13:6. [PMID: 39846701 PMCID: PMC11755660 DOI: 10.3390/medsci13010006] [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: 11/09/2024] [Revised: 12/05/2024] [Accepted: 12/23/2024] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND/OBJECTIVES Pseudomonas aeruginosa is a clinically significant opportunistic pathogen, renowned for its ability to acquire and develop diverse mechanisms of antibiotic resistance. This study examines the resistance, virulence, and regulatory mechanisms in extensively drug-resistant clinical strains of P. aeruginosa. METHODS Antibiotic susceptibility was assessed using the Minimum Inhibitory Concentration (MIC) method, and whole-genome sequencing (WGS) was performed on the Illumina NovaSeq platform. RESULTS The analysis demonstrated a higher prevalence of virulence genes compared to resistance and regulatory genes. Key virulence factors identified included secretion systems, motility, adhesion, and biofilm formation. Resistance mechanisms observed comprised efflux pumps and beta-lactamases, while regulatory systems involved two-component systems, transcriptional regulators, and sigma factors. Additionally, phenotypic profiles were found to correlate with resistance genes identified through genotypic analysis. CONCLUSIONS This study underscores the significant resistance and virulence of the clinical P. aeruginosa strains analyzed, highlighting the urgent need for alternative strategies to address infections caused by extensively drug-resistant bacteria.
Collapse
Affiliation(s)
- Nerlis Pajaro-Castro
- Medical and Pharmaceutical Sciences Group, Faculty of Health Sciences, University of Sucre, Sincelejo 700001, Sucre, Colombia;
- Salud Social Clinic, Sincelejo 700001, Sucre, Colombia; (E.D.-M.); (K.H.)
| | - Erick Diaz-Morales
- Salud Social Clinic, Sincelejo 700001, Sucre, Colombia; (E.D.-M.); (K.H.)
| | - Kenia Hoyos
- Salud Social Clinic, Sincelejo 700001, Sucre, Colombia; (E.D.-M.); (K.H.)
| | - Cristhian Ibañez-Bersinger
- Medical and Pharmaceutical Sciences Group, Faculty of Health Sciences, University of Sucre, Sincelejo 700001, Sucre, Colombia;
| |
Collapse
|
40
|
Azeem K, Fatima S, Ali A, Ubaid A, Husain FM, Abid M. Biochemistry of Bacterial Biofilm: Insights into Antibiotic Resistance Mechanisms and Therapeutic Intervention. Life (Basel) 2025; 15:49. [PMID: 39859989 PMCID: PMC11767195 DOI: 10.3390/life15010049] [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: 12/01/2024] [Revised: 12/30/2024] [Accepted: 12/30/2024] [Indexed: 01/27/2025] Open
Abstract
Biofilms, composed of structured communities of bacteria embedded in a self-produced extracellular matrix, pose a significant challenge due to their heightened resistance to antibiotics and immune responses. This review highlights the mechanisms underpinning antibiotic resistance within bacterial biofilms, elucidating the adaptive strategies employed by microorganisms to withstand conventional antimicrobial agents. This encompasses the role of the extracellular matrix, altered gene expression, and the formation of persister cells, contributing to the recalcitrance of biofilms to eradication. A comprehensive understanding of these resistance mechanisms provides a for exploring innovative therapeutic interventions. This study explores promising avenues for future research, emphasizing the necessity of uncovering the specific genetic and phenotypic adaptations occurring within biofilms. The identification of vulnerabilities in biofilm architecture and the elucidation of key biofilm-specific targets emerge as crucial focal points for the development of targeted therapeutic strategies. In addressing the limitations of traditional antibiotics, this review discusses innovative therapeutic approaches. Nanomaterials with inherent antimicrobial properties, quorum-sensing inhibitors disrupting bacterial communication, and bacteriophages as biofilm-specific viral agents are highlighted as potential alternatives. The exploration of combination therapies, involving antimicrobial agents, biofilm-disrupting enzymes, and immunomodulators, is emphasized to enhance the efficacy of existing treatments and overcome biofilm resilience.
Collapse
Affiliation(s)
- Kashish Azeem
- Medicinal Chemistry Laboratory, Department of Biosciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (K.A.); (S.F.); (A.A.); (A.U.)
| | - Sadaf Fatima
- Medicinal Chemistry Laboratory, Department of Biosciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (K.A.); (S.F.); (A.A.); (A.U.)
| | - Asghar Ali
- Medicinal Chemistry Laboratory, Department of Biosciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (K.A.); (S.F.); (A.A.); (A.U.)
- Clinical Biochemistry Laboratory, Department of Biochemistry, School of Chemical and Life Science, Jamia Hamdard, New Delhi 110062, India
| | - Ayesha Ubaid
- Medicinal Chemistry Laboratory, Department of Biosciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (K.A.); (S.F.); (A.A.); (A.U.)
| | - Fohad Mabood Husain
- Department of Food Science and Nutrition, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammad Abid
- Medicinal Chemistry Laboratory, Department of Biosciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (K.A.); (S.F.); (A.A.); (A.U.)
| |
Collapse
|
41
|
Mounayer N, Shoshani S, Afrimzon E, Iline-Vul T, Topaz M, Banin E, Margel S. Encapsulation of Hydrogen Peroxide in PVA/PVP Hydrogels for Medical Applications. Gels 2025; 11:31. [PMID: 39852002 PMCID: PMC11765405 DOI: 10.3390/gels11010031] [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: 12/11/2024] [Revised: 12/26/2024] [Accepted: 12/31/2024] [Indexed: 01/26/2025] Open
Abstract
Researchers have been investigating the physical and morphological properties of biodegradable polymer and copolymer films, blending them with other chemicals to solve challenges in medical, industrial, and eco-environmental fields. The present study introduces a novel, straightforward method for preparing biodegradable hydrogels based on polyvinyl alcohol (PVA) and polyvinyl pyrrolidone (PVP) for medical applications. The resulting PVA/PVP-based hydrogel uniquely combines the water absorbency, biocompatibility, and biodegradability of the polymer composite. For hygiene products and medical uses, such as wound healing, hydrogen peroxide (HP) was encapsulated in the PVA/PVP hydrogels for controlled release application. Incorporating PVP into PVA significantly enhances the hydrogel water absorbency and improves the mechanical properties. However, to mitigate the disadvantage of high water absorbency which could result in undesired early dissolution, efforts were made to increase the water resistance and the mechanical characteristics of these hydrogels using freeze-thaw (F/T) cycles and chemical crosslinking PVA chains with trisodium trimetaphosphate (STMP). The resulting hydrogels serve as environmentally friendly bio-based polymer blends, broadening their applications in medical and industrial products. The structural and morphological properties of the hydrogel were characterized using Fourier transform infrared spectroscopy (FTIR), environmental scanning electron microscope analysis (E-SEM), and water-swelling tests. The HP controlled release rate was evaluated through kinetic release experiments using the ex vivo skin model. The antibacterial activity of the hydrogel films was examined on four medically relevant bacteria: Staphylococcus aureus, Enterococcus faecalis, Escherichia coli, and Pseudomonas aeruginosa, with an adapted disk diffusion assay. Using this assay, we also evaluated the antibacterial effect of the hydrogel films over the course of days, demonstrating the HP controlled release from these hydrogels. These findings support further in vivo investigation into controlled HP release systems for improved wound-healing outcomes.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Shlomo Margel
- Institute of Nanotechnology and Advanced Materials, Department of Chemistry, Bar-Ilan University, Ramat-Gan 5290002, Israel; (N.M.); (S.S.); (E.A.); (T.I.-V.); (M.T.); (E.B.)
| |
Collapse
|
42
|
Al-Alousy NW, Al-Nasiri FS. Bacterial infections associated with cutaneous leishmaniasis in Salah Al-Din province, Iraq. Microb Pathog 2025; 198:107144. [PMID: 39579944 DOI: 10.1016/j.micpath.2024.107144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 11/20/2024] [Accepted: 11/21/2024] [Indexed: 11/25/2024]
Abstract
Bacterial co-infection with cutaneous leishmaniasis (CL) can effect on the clinical appearance of lesions and delay the healing process. The pattern of bacterial pathogens involved has rarely been investigated in Iraq. The aim of present study was to identify the bacterial agents contaminating CL and their susceptibility to commonly used antibiotics. Four hundred cases of CL were diagnosed in Salah El-Din General Hospital, Iraq. A total of 424 ulcer samples obtained from 400 patients of CL were cultured, and all isolates were diagnosed based on phenotypic characteristics of colonies, bacterial cells and using biochemical tests. No bacterial growth appeared in 124 cases (29.25 %) of the total number of examined ulcer samples, whereas 300 cases (70.75 %) were contaminated. Both Gram-positive (Staphylococcus aureus, Staphylococcus epidermidis, Streptococcus pyogenes) and Gram-negative bacterial species (Escherichi coli, Klebsiella spp., Pseudomonas aeruginosa) were present. The infection with S. aureus represented the highest percentage (26.42 %), while Ps. aeruginosa had the lowest percentage (2.83 %) compared with other bacterial infections. The sensitivity of the isolated bacteria associated with CL was tested against a number of antibiotics (Amoxicillin, Ampicillin, Cefixime, Chloramphenlcol, Doxycycline, Tetracycline). Resistance to Amoxicillin, Ampicillin, and Cefixime was generally high. While, Chloramphenicol showed absolute effectiveness against isolated bacteria. The results of the current study show that bacterial infections should be considered in diagnosing and treating CL lesions, with Chloramphenicol demonstrating the highest efficiency in treating such bacterial infections. The present study also suggests that hygiene, use of suitable disinfectants, controlling of antibiotic administration and prescription in hospitals and pharmacies must be ensured. In addition, regular surveillance in the endemic area will help control bacterial co-infection and hamper the occurrence of drug-resistant pathogens. The lesion care and management of secondary bacterial infection are essential and anti -leishmanial therapy in CL may be more effective when combined with antibiotics. Future molecular studies are needed to identify the species of Leishmania causing CL in Iraq to gain a better understanding of their clinical manifestations (dry or moist ulcers) and their associated bacteria.
Collapse
|
43
|
Taslimi Eshkalak M, Mazloumi Jourkouyeh E, Faezi Ghasemi M, Zamani H, Zahmatkesh H, Rasti B. ZnO-Rutin nanostructure as a potent antibiofilm agent against Pseudomonasaeruginosa. Microb Pathog 2025; 198:107156. [PMID: 39608510 DOI: 10.1016/j.micpath.2024.107156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 11/03/2024] [Accepted: 11/22/2024] [Indexed: 11/30/2024]
Abstract
Pseudomonas aeruginosa is a common human pathogen that is resistant to multiple antibiotics due to its ability to form biofilms. Developing novel nanoformulations capable of inhibiting and removing biofilms offers a promising solution for controlling biofilm-related infections. In this study, we investigated the anti-biofilm activity of rutin-conjugated ZnO nanoparticles (ZnO-Rutin NPs) in pathogenic strains of P. aeruginosa. The synthesized ZnO-Rutin NPs had amorphous shapes with sizes ranging from 14 to 100 nm. The broth microdilution assay revealed that ZnO-Rutin NPs, with an MIC value of 2 mg/mL, exhibit greater antimicrobial activity than ZnO NPs and rutin alone. Based on crystal violet staining, the biofilm inhibition rate by ½ MIC of the conjugated nanoparticles was recorded at above 90 %. The significant reduction in exopolysaccharide (62.75-66.37 %) and alginate (38.3-57.61 %) levels, as well as the formation of thin biofilms in the ZnO-Rutin NP-treated group, confirmed the anti-biofilm potential of these nanoparticles. Additionally, a significant decrease in the metabolic activity and viable cells of mature biofilms was observed after exposure to the conjugated nanoparticles. Furthermore, ZnO-Rutin NPs considerably attenuated the expression of the Las-Rhl quorum-sensing transcriptional regulator genes (lasR and rhlR) in P. aeruginosa by 0.39-0.40 and 0.25-0.42 folds, respectively. This work demonstrated that ZnO-Rutin NPs are remarkably capable of inhibiting the initial stage of biofilm formation and eradicating mature biofilms, suggesting they could be a useful agent for treating P. aeruginosa biofilm-related infections.
Collapse
Affiliation(s)
- Mahya Taslimi Eshkalak
- Department of Microbiology, Faculty of Basic Sciences, Lahijan Branch, Islamic Azad University, Lahijan, Iran
| | - Edris Mazloumi Jourkouyeh
- Department of Microbiology, Faculty of Basic Sciences, Lahijan Branch, Islamic Azad University, Lahijan, Iran
| | - Mohammad Faezi Ghasemi
- Department of Microbiology, Faculty of Basic Sciences, Lahijan Branch, Islamic Azad University, Lahijan, Iran
| | | | - Hossein Zahmatkesh
- Department of Microbiology, Faculty of Basic Sciences, Lahijan Branch, Islamic Azad University, Lahijan, Iran.
| | - Behnam Rasti
- Department of Microbiology, Faculty of Basic Sciences, Lahijan Branch, Islamic Azad University, Lahijan, Iran.
| |
Collapse
|
44
|
Wang G, Zhang C, Huang Z, Chen J, Chen H, Lin T, Zhou Z, Gu N, Huang P. Transcytosable and Ultrasound-Activated Liposome Enables Deep Penetration of Biofilm for Surgical Site Infection Management. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2411092. [PMID: 39463041 DOI: 10.1002/adma.202411092] [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: 07/29/2024] [Revised: 09/22/2024] [Indexed: 10/29/2024]
Abstract
Biofilm-associated surgical site infection (BSSI) is a common and grievous postoperative complication lacking effective remedies, mainly due to the poor drug accumulation and penetration in the biofilms featured by dense extracellular polymeric substances (EPSs). Here, it is found that the vascular cell adhesion molecule-1 (VCAM1) is highly overexpressed in the vascular cells of BSSI. It is proposed that the combination of VCAM1-mediated transcytosis and ultrasonic cavitation can consecutively overcome the biological barriers of vascular endothelial cells and EPS for biofilm eradication. To demonstrate the feasibility, a VCAM1-targeted and ultrasound (US)-activated liposome (LPCOTML) loaded with a reactive-oxygen-species (ROS)-responsive lipoid prodrug of oleoyl meropenem, sonosensitizer of lipoid Ce6, and perfluoropentane is developed. LPCOTML can recognize the receptors on vascular cells, and initiate receptor-mediated transcytosis for transendothelial transport into the BSSI periphery. LPCOTML subsequently transforms from nanoparticle into microbubble via liquid-gas phase transition under US irradiation, triggering strong ultrasonic cavitation to blow up the EPS and deeply penetrate the biofilms. The sonosensitizer Ce6 induces ROS production under US irradiation and triggers the release of meropenem to induce potent antibacterial effect in a BSSI model. This study presents an effective strategy to tackle the biological barriers in BSSI via combining receptor-mediated transcytosis and ultrasonic cavitation.
Collapse
Affiliation(s)
- Guowei Wang
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Chengyue Zhang
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Zixuan Huang
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310030, China
| | - Jifan Chen
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Hongjian Chen
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, Hangzhou, 310009, China
| | - Tao Lin
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Zhuxian Zhou
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Ning Gu
- Medical School of Nanjing University, Nanjing University, Nanjing, 210093, China
| | - Pintong Huang
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, Hangzhou, 310009, China
| |
Collapse
|
45
|
Priya, Gaur PK, Kumar S. Nanocarrier-Mediated Dermal Drug Delivery System of Antimicrobial Agents for Targeting Skin and Soft Tissue Infections. Assay Drug Dev Technol 2025; 23:2-28. [PMID: 39587945 DOI: 10.1089/adt.2024.060] [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: 11/27/2024] Open
Abstract
Antimicrobial resistance in disease-causing microbes is seen as a severe problem that affects the entire world, makes therapy less effective, and raises mortality rates. Dermal antimicrobial therapy becomes a desirable choice in the management of infectious disorders since the rising resistance to systemic antimicrobial treatment frequently necessitates the use of more toxic drugs. Nanoparticulate systems such as nanobactericides, which have built-in antibacterial activity, and nanocarriers, which function as drug delivery systems for conventional antimicrobials, are just two examples of the treatment methods made feasible by nanotechnology. Silver nanoparticles, zinc oxide nanoparticles, and titanium dioxide nanoparticles are examples of inorganic nanoparticles that are efficient on sensitive and multidrug-resistant bacterial strains both as nanobactericides and nanocarriers. To stop the growth of microorganisms that are resistant to standard antimicrobials, various antimicrobials for dermal application are widely used. This review covers the most prevalent microbes responsible for skin and soft tissue infections, techniques to deliver dermal antimicrobials, topical antimicrobial safety concerns, current issues, challenges, and potential future developments. A thorough and methodical search of databases, such as Google Scholar, PubMed, Science Direct, and others, using specified keyword combinations, such as "antimicrobials," "dermal," "nanocarriers," and numerous others, was used to gather relevant literature for this work.
Collapse
Affiliation(s)
- Priya
- Department of Pharmaceutical Technology, Meerut Institute of Engineering & Technology, Meerut, Uttar Pradesh, India
| | - Praveen Kumar Gaur
- Department of Pharmaceutics, Metro College of Health Sciences & Research, Greater Noida, Uttar Pradesh, India
| | - Shobhit Kumar
- Department of Pharmaceutical Technology, Meerut Institute of Engineering & Technology, Meerut, Uttar Pradesh, India
| |
Collapse
|
46
|
Lock GDA, Helfer VE, Dias BB, Barreto F, Dalla Costa T, de Araújo BV. Can distinct Gram-negative biofilm-forming bacteria have different impacts on ciprofloxacin lung penetration? Microb Pathog 2025; 198:107092. [PMID: 39515546 DOI: 10.1016/j.micpath.2024.107092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/04/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
Literature have shown that Gram-negative bacteria release endotoxins which alter drug membrane transporters and could potentially influence antimicrobials distribution to the infection site depending on the infecting bacteria. Previously, a population pharmacokinetic (popPK) model describing ciprofloxacin (CIP) concentrations in healthy, and Pseudomonas aeruginosa pneumonic rats showed that the chronic stage of the infection significantly reduced the drug lung penetration. In this study, CIP lung penetration in Klebsiella pneumoniae chronically (14 d) infected rats following CIP 20 mg/kg i.v. bolus dosing was investigated and the popPK model developed previously was used to evaluate CIP lung exposure. Drug plasma exposure was similar for both bacteria and higher than observed in healthy animals. Probability of target attainment analysis using plasma data following current dosing regimen (20 mg q8h equivalent to 400 mg q8h in humans) showed that CIP PK/PD index (ƒAUC0-24/MIC ≥90) is achieved for the most prevalent MIC's of both bacteria. However, CIP free lung concentrations were reduced in infected animals by 46.8 % (P. aeruginosa) and 68.4 % (K. pneumoniae) in comparison to healthy animals. The higher lung clearance observed (0.306 L/h/kg) in K. pneumoniae infected animals lead to a lower free CIP lung exposure in comparison to the P. aeruginosa group (0.105 L/h/kg). In summary, although plasma PK/PD index is achieved by the current regimen, chronic pneumonia by biofilm-forming bacteria decreases lung exposure to CIP and this decrease is dependent on the infecting bacteria. The clinical relevance of this finding needs to be determined.
Collapse
Affiliation(s)
- Graziela de Araújo Lock
- Pharmacokinetics and PK/PD Modeling Lab, Pharmaceutical Sciences Graduate Program, College of Pharmacy, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Victória Etges Helfer
- Pharmacokinetics and PK/PD Modeling Lab, Pharmaceutical Sciences Graduate Program, College of Pharmacy, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Bruna Bernar Dias
- Pharmacokinetics and PK/PD Modeling Lab, Pharmaceutical Sciences Graduate Program, College of Pharmacy, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Fabiano Barreto
- Federal Laboratory of Animal and Plant Health and Inspection - LFDA, Porto Alegre, RS, Brazil
| | - Teresa Dalla Costa
- Pharmacokinetics and PK/PD Modeling Lab, Pharmaceutical Sciences Graduate Program, College of Pharmacy, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Bibiana Verlindo de Araújo
- Pharmacokinetics and PK/PD Modeling Lab, Pharmaceutical Sciences Graduate Program, College of Pharmacy, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil.
| |
Collapse
|
47
|
Benaissa A, Wafaa B, Ngenge Tamfu A, Ammara B, Kucukaydin S, Latti N, Khadir A, Bendahou M, Anouar EH, Ceylan O. Inhibition of Clinical Multidrug-Resistant Pseudomonas aeruginosa Biofilms by Cinnamaldehyde and Eugenol From Essential Oils: In Vitro and In Silico Analysis. Chem Biodivers 2024:e202402693. [PMID: 39740034 DOI: 10.1002/cbdv.202402693] [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: 10/21/2024] [Revised: 12/27/2024] [Accepted: 12/28/2024] [Indexed: 01/02/2025]
Abstract
Pseudomonas aeruginosa causes nosocomial infections and chronic diseases. Cinnamomum cassia and Syzygium aromaticum are used natural antimicrobials. Essential oil (EO) from C. cassia (CCEO) and S. aromaticum (CEO) was characterized using GC-MS analysis. Eugenol (82.31%), eugenol acetate (10.57%), and β-caryophyllene (3.41%) were major constituents in CEO while cinnamaldehyde (88.18%), cinnamyl acetate (2.85%) and 2-methoxy cinnamaldehyde (1.77%) were main components in CCEO. The EOs and major constituents exhibited good antimicrobial activity against clinical strains of P. aeruginosa. Cinnamaldehyde exhibited the best antimicrobial effect with minimal inhibitory concentration (MIC) as low as 0.031% ± 0.07% (v/v) and inhibition zones reaching 30 ± 0.5 mm diameter. Test samples showed antibiofilm activities against two culture types and seven clinical strains of P. aeruginosa at concentrations of 2MIC to MIC/4. CCEO and its major constituent cinnamaldehyde were more active, compared to CEO and its major constituent eugenol. Scanning electron microscopy images showed untreated colonies with well-developed biofilms while there was significant reduction of biofilms with distorted architecture and cell shrinkage upon treatment with test samples. In silico studies indicated great interactions between the major compounds, eugenol and cinnamaldehyde, with the receptor proteins of P. aeruginosa revealed by negative binding energies. Eugenol and cinnamaldehyde exhibited appreciable druglikeness.
Collapse
Affiliation(s)
- Asma Benaissa
- Laboratory of Applied Microbiology in Food, Biomedical, and Environment (LAMAABE), Department of Biology, Faculty of Sciences of Nature, Life, Earth, and Universe, Abou Bekr Belkaïd University of Tlemcen, Tlemcen, Algeria
| | - Bouali Wafaa
- Laboratory Antifungal, Antibiotic, Physico-chemical, Synthesis and Biological Activity, Department of Biology, Faculty of Natural Sciences and Life, Sciences of the Earth and the Universe, University Abou Bekr Belkaid Tlemcen, Tlemcen, Algeria
| | - Alfred Ngenge Tamfu
- Department of Chemical Engineering, School of Chemical Engineering and Mineral Industries, University of Ngaoundere, Ngaoundere, Cameroon
- Food Quality Control and Analysis Program, Ula Ali Kocman Vocational School, Mugla Sitki Kocman University, Mugla, Turkey
| | - Bousselham Ammara
- Microbiology Laboratory, University Hospital Center of Tlemcen, Tlemcen, Algeria
| | - Selcuk Kucukaydin
- Department of Medical Services and Techniques, Koycegiz Vocational School of Health Services, Mugla Sitki Kocman University, Mugla, Turkey
| | - Nawel Latti
- Laboratory of Applied Microbiology in Food, Biomedical, and Environment (LAMAABE), Department of Biology, Faculty of Sciences of Nature, Life, Earth, and Universe, Abou Bekr Belkaïd University of Tlemcen, Tlemcen, Algeria
| | - Abdelmounaim Khadir
- Laboratory of Applied Microbiology in Food, Biomedical, and Environment (LAMAABE), Department of Biology, Faculty of Sciences of Nature, Life, Earth, and Universe, Abou Bekr Belkaïd University of Tlemcen, Tlemcen, Algeria
- Department of Biology, Oran University, Oran, Algeria
| | - Mourad Bendahou
- Laboratory of Applied Microbiology in Food, Biomedical, and Environment (LAMAABE), Department of Biology, Faculty of Sciences of Nature, Life, Earth, and Universe, Abou Bekr Belkaïd University of Tlemcen, Tlemcen, Algeria
| | - El Hassane Anouar
- Department of Chemistry, College of Sciences and Humanities in Al-Kharj, Prince Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Ozgur Ceylan
- Food Quality Control and Analysis Program, Ula Ali Kocman Vocational School, Mugla Sitki Kocman University, Mugla, Turkey
| |
Collapse
|
48
|
Olana MD, Asrat D, Swedberg G. Antimicrobial resistance profile, biofilm forming capacity and associated factors of multidrug resistance in Pseudomonas aeruginosa among patients admitted at Tikur Anbessa Specialized Hospital and Yekatit 12 Hospital Medical College in Addis Ababa, Ethiopia. BMC Infect Dis 2024; 24:1472. [PMID: 39732630 DOI: 10.1186/s12879-024-10359-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 12/16/2024] [Indexed: 12/30/2024] Open
Abstract
BACKGROUND Pseudomonas aeruginosa is one of the leading causes of nosocomial infections and the most common multidrug-resistant pathogen. This study aimed to determine antimicrobial resistance patterns, biofilm-forming capacity, and associated factors of multidrug resistance in P. aeruginosa isolates at two hospitals in Addis Ababa, Ethiopia. METHODS A cross-sectional study was conducted from August 2022 to August 2023 at Tikur Anbessa Specialized Hospital and Yekatit 12 Hospital Medical College. Culture and identification of P. aeruginosa were done using standard microbiological methods. An antimicrobial susceptibility test was done by Kirby-Bauer disk diffusion according to CLSI recommendations. The microtiter plate assay method was used to determine biofilm-forming capacity. SPSS version 25 was used for data analysis. Bivariate and multivariable logistic regression were used to assess factors associated with multidrug resistance in P. aeruginosa. The Spearman correlation coefficient (rs = 0.266)) was performed to evaluate the relationship between biofilm formation and drug resistance. RESULTS The overall prevalence of P. aeruginosa was 19.6%. High levels of resistance were observed for ciprofloxacin (51.8%), ceftazidime (50.6%), and cefepime (48.2%). The level of multidrug-resistance was 56.6%. The isolates showed better susceptibility to ceftazidime-avibactam (95.2%) and imipenem (79.5%). Overall, 95.2% of P. aeruginosa were biofilm-producing isolates, and 27.7% and 39.8% of isolates were strong and moderate biofilm producers, respectively. A positive correlation and statistically significant relationship was observed between resistance to multiple drugs and the level of biofilm formation (rs = 0.266; p-value = 0.015). Previous history of exposure to ciprofloxacin (OR, 5.1; CI, 1.12-24.7, p-value, 0.032) was identified as an independent associated factor for multidrug resistance in P. aeruginosa. CONCLUSION The present study indicates an association between multidrug resistance in P. aeruginosa and its biofilm formation capabilities. Additionally, over half of the isolates were resistant to multiple drugs, with prior use of ciprofloxacin linked to the development of multidrug-resistance. These findings suggest that antibiotic stewardship programs in hospital settings may be beneficial in addressing resistance.
Collapse
Affiliation(s)
- Matifan Dereje Olana
- Department of Medical Laboratory Sciences, Collage of Medicine and Health Sciences, Ambo University, Ambo, Ethiopia.
- Department of Microbiology, Immunology and Parasitology, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia.
- Department of Medical Biochemistry and Microbiology, Biomedical Centre, Uppsala University, Uppsala, Sweden.
| | - Daniel Asrat
- Department of Microbiology, Immunology and Parasitology, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | - Göte Swedberg
- Department of Medical Biochemistry and Microbiology, Biomedical Centre, Uppsala University, Uppsala, Sweden
| |
Collapse
|
49
|
Lange A, Kutwin M, Zawadzka K, Ostrowska A, Strojny-Cieślak B, Nasiłowska B, Bombalska A, Jaworski S. Impaired Biofilm Development on Graphene Oxide-Metal Nanoparticle Composites. Nanotechnol Sci Appl 2024; 17:303-320. [PMID: 39734361 PMCID: PMC11681909 DOI: 10.2147/nsa.s485841] [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: 07/06/2024] [Accepted: 12/10/2024] [Indexed: 12/31/2024] Open
Abstract
Purpose Biofilms are one of the main threats related to bacteria. Owing to their complex structure, in which bacteria are embedded in the extracellular matrix, they are extremely challenging to eradicate, especially since they can inhabit both biotic and abiotic surfaces. This study aimed to create an effective antibiofilm nanofilm based on graphene oxide-metal nanoparticles (GOM-NPs). Methods To create nanofilms, physicochemical analysis was performed, including zeta potential (Zp) (and the nanocomposites stability in time) and size distribution measurements, scanning transmission electron microscopy (STEM), energy dispersive X-ray analysis (EDX), and atomic force microscopy (AFM) of the nanofilm surfaces. During biological analysis, reactive oxygen species (ROS) and antioxidant capacity were measured in planktonic cells treated with the nanocomposites. Thereafter, biofilm formation was checked via crystal violet staining, biofilm thickness was assessed by confocal microscopy using double fluorescent staining, and biofilm structure was analyzed by scanning electron microscopy. Results The results showed that two of the three nanocomposites were effective in reducing biofilm formation (GOAg and GOZnO), although the nanofilms were characterized by the roughest surface, indicating that high surface roughness is unfavorable for biofilm formation by the tested bacterial species (Staphylococcus aureus (ATCC 25923), Salmonella enterica (ATCC 13076), Pseudomonas aeruginosa (ATCC 27853)). Conclusion The performed analysis indicated that graphene oxide may be a platform for metal nanoparticles that enhances their properties (eg colloidal stability, which is maintained over time). Nanocomposites based on graphene oxide with silver nanoparticles and other types of nanocomposites with zinc oxide were effective against biofilms, contributing to changes throughout the biofilm structure, causing a significant reduction in the thickness of the structure, and affecting cell distribution. A nanocomposite consisting of graphene oxide with copper nanoparticles inhibited the biofilm, but to a lesser extent.
Collapse
Affiliation(s)
- Agata Lange
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, Warsaw, Poland
| | - Marta Kutwin
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, Warsaw, Poland
| | - Katarzyna Zawadzka
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, Warsaw, Poland
| | - Agnieszka Ostrowska
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, Warsaw, Poland
| | - Barbara Strojny-Cieślak
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, Warsaw, Poland
| | - Barbara Nasiłowska
- Center for Biomedical Engineering, Institute of Optoelectronics, Military University of Technology, Warsaw, Poland
| | - Aneta Bombalska
- Department of Optoelectronic Technologies, Institute of Optoelectronics, Military University of Technology, Warsaw, Poland
| | - Sławomir Jaworski
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, Warsaw, Poland
| |
Collapse
|
50
|
Koujalagi T, Ruhal R. Mitigating Health Risks Through Environmental Tracking of Pseudomonas aeruginosa. Curr Microbiol 2024; 82:57. [PMID: 39718600 DOI: 10.1007/s00284-024-04036-6] [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: 09/24/2024] [Accepted: 12/12/2024] [Indexed: 12/25/2024]
Abstract
Pseudomonas aeruginosa is a prevalent nosocomial pathogen and a significant reservoir of antimicrobial resistance genes in residential and built environments. It is also widespread in various indoor and outdoor settings, including sewage, surface waters, soil, recreational waters (both treated and untreated), and industrial effluents. Surveillance efforts for P. aeruginosa are primarily focused on hospitals rather than built environments. However, evidence links multidrug-resistant P. aeruginosa of human origin with activity in built environments and hospital settings. Consequently, tracking this pathogen across all environments is crucial for understanding the mechanisms of reverse transmission from built environments to humans. This review explores public health hygiene by examining the prevalence of P. aeruginosa in various environments, its sequence types, the factors contributing to multidrug resistance, and the identification methods through global surveillance. Whole-genome sequencing with sequence typing and real-time quantitative PCR are widely used to identify and study antimicrobial-resistant strains worldwide. Additionally, advanced techniques such as functional metagenomics, next-generation sequencing, MALDI-TOF, and biosensors are being extensively employed to detect antimicrobial-resistant strains and mitigate the ongoing evolution of bacterial resistance to antibiotics. Our review strongly underscores the importance of environmental monitoring of P. aeruginosa in preventing human infections. Furthermore, strategic planning in built environments is essential for effective epidemiological surveillance of P. aeruginosa and the development of comprehensive risk assessment models.
Collapse
Affiliation(s)
- Tushar Koujalagi
- School of Bio Science and Technology, VIT University, Vellore, Tamil Nadu, 632014, India
| | - Rohit Ruhal
- School of Bio Science and Technology, VIT University, Vellore, Tamil Nadu, 632014, India.
| |
Collapse
|