1
|
Ferreira RL, Parente Rocha JA, Leite VRMC, Moraes D, Graziani D, Pranchevicius MCDS, Soares CMDA. Proteomic profile of multidrug-resistant Serratia marcescens under meropenem challenge. Microb Pathog 2025; 204:107570. [PMID: 40222567 DOI: 10.1016/j.micpath.2025.107570] [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: 11/06/2024] [Revised: 01/13/2025] [Accepted: 04/09/2025] [Indexed: 04/15/2025]
Abstract
Serratia marcescens is an opportunistic bacterium implicated in the prevalence of serious nosocomial infections and increased outbreaks in Intensive Care Units (ICUs) and Neonatal Intensive Care Units (NICUs). S. marcescens strains are resistant to several antimicrobial classes and express numerous virulence factors that promote pathogenicity. In the present study, the proteomic profile of the multidrug-resistant (MDR) S. marcescens clinical isolate challenged with the antimicrobial meropenem was evaluated. The proteins obtained were analyzed using liquid chromatography coupled with tandem mass spectrometry (LC-MSE). A total of 199 induced proteins were identified revealing that multidrug-resistant S. marcescens promotes increasing of proteins related to energy metabolism and efflux pump and decreases synthesis of proteins related to oxidative stress response and cell mobility upon meropenem challenge, shedding some light on the relationship between expressed proteins and bacterial pathogenicity after antimicrobial induction.
Collapse
Affiliation(s)
- Roumayne Lopes Ferreira
- Laboratório de Biologia Molecular, Instituto de Ciências Biológicas, ICB II, Campus II, Universidade Federal de Goiás, 74001-970, Goiânia, GO, Brazil.
| | - Juliana Alves Parente Rocha
- Laboratório de Biologia Molecular, Instituto de Ciências Biológicas, ICB II, Campus II, Universidade Federal de Goiás, 74001-970, Goiânia, GO, Brazil.
| | - Vanessa Rafaela Milhomem Cruz Leite
- Laboratório de Biologia Molecular, Instituto de Ciências Biológicas, ICB II, Campus II, Universidade Federal de Goiás, 74001-970, Goiânia, GO, Brazil.
| | - Dayane Moraes
- Laboratório de Biologia Molecular, Instituto de Ciências Biológicas, ICB II, Campus II, Universidade Federal de Goiás, 74001-970, Goiânia, GO, Brazil.
| | - Daniel Graziani
- Laboratório de Avaliação de Moléculas, Células e Tecidos, Escola de Veterinária e Zootecnia, Universidade Federal de Goiás, Goiânia, GO, Brazil.
| | | | - Célia Maria de Almeida Soares
- Laboratório de Biologia Molecular, Instituto de Ciências Biológicas, ICB II, Campus II, Universidade Federal de Goiás, 74001-970, Goiânia, GO, Brazil.
| |
Collapse
|
2
|
Moulick S, Bera R, Roy DN. Bactericidal action of ginger (Zingiber officinale Roscoe) extract against Escherichia coli through synergistic modulation of the AcrAB-TolC efflux pump and inhibition of peptidoglycan synthesis: In vitro and in silico approaches. Microb Pathog 2025; 204:107624. [PMID: 40268150 DOI: 10.1016/j.micpath.2025.107624] [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: 11/13/2024] [Revised: 04/16/2025] [Accepted: 04/21/2025] [Indexed: 04/25/2025]
Abstract
The emergence of multidrug-resistant Escherichia coli is considered a severe threat to global health, largely attributed to the bacterium's ability to expel antibiotics via efflux pump systems. This study explores the antibacterial efficacy of a methanol extract derived from Zingiber officinale R. (ginger), a traditional medicinal spice, against an E. coli strain overexpressing the AcrAB-TolC efflux system. To evaluate the extract's efficacy, three E. coli strains were tested: AG100 (AcrAB-TolC+), AG100A (ΔAcrAB), and D22 (lpxC mutant). The ginger extract exhibited antibacterial activity against E. coli AG100A and D22, with minimal inhibitory concentrations (MIC) and minimal bactericidal concentrations (MBC) of 625 μg/mL. However, the extract showed no inhibitory effect against E. coli AG100, even at 10 mg/mL, suggesting the AcrAB-TolC system plays a key role in resistance. Notably, combining the extract with an efflux pump inhibitor (EPI) strongly enhanced its bactericidal effect, reducing the MBC for AG100 to 313 μg/mL. EtBr accumulation assays confirmed that the ginger extract, in combination with EPI, amplified intracellular drug retention, peaking fluorescence within 30 min and sustaining elevated levels over 60 min. Molecular docking further revealed that bioactive compounds such as 6-shogaol strongly bind within the binding domain of AcrB homotrimer, inhibiting pump function. Additionally, cell wall biosynthesis assays demonstrated 69-75 % inhibition when the ginger extract was used at 2-fold-4-fold its MIC in the presence of EPI, further intensifying bactericidal effects. These results underscore ginger's dual-action mechanisms, highlighting its potential as an effective natural antimicrobial agent against drug-resistant E. coli.
Collapse
Affiliation(s)
- Soumitra Moulick
- TCG Lifesciences Private Limited, Kolkata, West Bengal, India; Department of Biotechnology, National Institute of Technology - Raipur, Chhattisgarh, India
| | - Rammohan Bera
- TCG Lifesciences Private Limited, Kolkata, West Bengal, India
| | - Dijendra Nath Roy
- Department of Biotechnology, National Institute of Technology - Raipur, Chhattisgarh, India.
| |
Collapse
|
3
|
Jiang C, Zhou L, Wang M, Shen S, Cheng W, Zhao Q, Cui K, He L. Sensitivity determination and resistance mechanism of Sclerotium rolfsii to difenoconazole. PEST MANAGEMENT SCIENCE 2025; 81:2734-2741. [PMID: 39726070 DOI: 10.1002/ps.8624] [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: 07/12/2024] [Revised: 12/09/2024] [Accepted: 12/12/2024] [Indexed: 12/28/2024]
Abstract
BACKGROUND Peanut stem rot, caused by Sclerotium rolfsii, has become increasingly prevalent in China, leading to significant yield losses in peanut production. To effectively manage peanut stem rot, we assessed the potential application of difenoconazole against peanut stem rot. RESULTS Difenoconazole has a good inhibitory effect on the mycelial growth of S. rolfsii, with half maximal effective concentration (EC50) values ranging from 0.10 to 1.58 μg/mL and an average of 0.33 ± 0.02 μg/mL. Nonetheless, a small percentage of wild-type isolates exhibiting low resistance to difenoconazole were identified in the field. The primary reason for S. rolfsii resistance to difenoconazole was found to be attributed to the overexpression of CYP51. In addition, a small number of resistant isolates also exhibited multidrug resistance through the overexpression of efflux pump genes atrB and atrD. Pot experiments revealed that difenoconazole demonstrated superior protective efficacy against peanut stem rot, with mist spray treatment exhibiting better control efficacy compared to root drench treatment. Specifically, at a concentration of 100 μg/mL, the protective efficacy of difenoconazole mist spray against peanut stem rot reached 65.24%, which was statistically similar to that of tebuconazole. Furthermore, no significant correlation was observed between sensitivity to difenoconazole and mefentrifluconazole, benzovindiflupyr, boscalid, thifluzamide, carboxin, or picoxystrobin. CONCLUSION To delay the emergence of resistant populations, we recommend early-stage application of difenoconazole via spraying for peanut stem rot management, alongside the judicious use of fungicides with no cross-resistance like thifluzamide and boscalid for optimal control. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Chaofan Jiang
- College of Plant Protection, Henan Agricultural University, Zhengzhou, China
- Henan Key Laboratory of Creation and Application of New Pesticide, Henan Agricultural University, Zhengzhou, China
- Henan Research Center of Green Pesticide Engineering and Technology, Henan Agricultural University, Zhengzhou, China
| | - Lin Zhou
- College of Plant Protection, Henan Agricultural University, Zhengzhou, China
- Henan Key Laboratory of Creation and Application of New Pesticide, Henan Agricultural University, Zhengzhou, China
- Henan Research Center of Green Pesticide Engineering and Technology, Henan Agricultural University, Zhengzhou, China
| | - Mengke Wang
- College of Plant Protection, Henan Agricultural University, Zhengzhou, China
- Henan Key Laboratory of Creation and Application of New Pesticide, Henan Agricultural University, Zhengzhou, China
- Henan Research Center of Green Pesticide Engineering and Technology, Henan Agricultural University, Zhengzhou, China
| | - Sirui Shen
- College of Plant Protection, Henan Agricultural University, Zhengzhou, China
- Henan Key Laboratory of Creation and Application of New Pesticide, Henan Agricultural University, Zhengzhou, China
- Henan Research Center of Green Pesticide Engineering and Technology, Henan Agricultural University, Zhengzhou, China
| | - Weifeng Cheng
- College of Plant Protection, Henan Agricultural University, Zhengzhou, China
- Henan Key Laboratory of Creation and Application of New Pesticide, Henan Agricultural University, Zhengzhou, China
- Henan Research Center of Green Pesticide Engineering and Technology, Henan Agricultural University, Zhengzhou, China
| | - Qingchen Zhao
- College of Plant Protection, Henan Agricultural University, Zhengzhou, China
- Henan Key Laboratory of Creation and Application of New Pesticide, Henan Agricultural University, Zhengzhou, China
- Henan Research Center of Green Pesticide Engineering and Technology, Henan Agricultural University, Zhengzhou, China
| | - Kaidi Cui
- College of Plant Protection, Henan Agricultural University, Zhengzhou, China
- Henan Key Laboratory of Creation and Application of New Pesticide, Henan Agricultural University, Zhengzhou, China
- Henan Research Center of Green Pesticide Engineering and Technology, Henan Agricultural University, Zhengzhou, China
| | - Leiming He
- College of Plant Protection, Henan Agricultural University, Zhengzhou, China
- Henan Key Laboratory of Creation and Application of New Pesticide, Henan Agricultural University, Zhengzhou, China
- Henan Research Center of Green Pesticide Engineering and Technology, Henan Agricultural University, Zhengzhou, China
| |
Collapse
|
4
|
Zou H, Huang X, Xiao W, He H, Liu S, Zeng H. Recent advancements in bacterial anti-phage strategies and the underlying mechanisms altering susceptibility to antibiotics. Microbiol Res 2025; 295:128107. [PMID: 40023108 DOI: 10.1016/j.micres.2025.128107] [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: 11/12/2024] [Revised: 02/12/2025] [Accepted: 02/16/2025] [Indexed: 03/04/2025]
Abstract
The rapid spread of multidrug-resistant bacteria and the challenges in developing new antibiotics have brought renewed international attention to phage therapy. However, in bacteria-phage co-evolution, the rapid development of bacterial resistance to phage has limited its clinical application. This review consolidates the latest advancements in research on anti-phage mechanisms, encompassing strategies such as systems associated with reduced nicotinamide adenine dinucleotide (NAD+) to halt the propagation of the phage, symbiotic bacteria episymbiont-mediated modulation of gene expression in host bacteria to resist phage infection, and defence-related reverse transcriptase (DRT) encoded by bacteria to curb phage infections. We conduct an in-depth analysis of the underlying mechanisms by which bacteria undergo alterations in antibiotic susceptibility after developing phage resistance. We also discuss the remaining challenges and promising directions for phage-based therapy in the future.
Collapse
Affiliation(s)
- Huanhuan Zou
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Xiaoyi Huang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Wenyue Xiao
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Haoxuan He
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Shenshen Liu
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Haiyan Zeng
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China.
| |
Collapse
|
5
|
Choe D, Lee E, Kim K, Hwang S, Jeong KJ, Palsson BO, Cho BK, Cho S. Rapid identification of key antibiotic resistance genes in E. coli using high-resolution genome-scale CRISPRi screening. iScience 2025; 28:112435. [PMID: 40352728 PMCID: PMC12063145 DOI: 10.1016/j.isci.2025.112435] [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: 02/06/2025] [Accepted: 04/10/2025] [Indexed: 05/14/2025] Open
Abstract
Bacteria possess a vast repertoire of genes to adapt to environmental challenges. Understanding the gene fitness landscape under antibiotic stress is crucial for elucidating bacterial resistance mechanisms and antibiotic action. To explore this, we conducted a genome-scale CRISPRi screen using a high-density sgRNA library in Escherichia coli exposed to various antibiotics. This screen identified essential genes under antibiotic-induced stress and offered insights into the molecular mechanisms underlying bacterial responses. We uncovered previously unrecognized genes involved in antibiotic resistance, including essential membrane proteins. The screen also underscored the importance of transcriptional modulation of essential genes in antibiotic tolerance. Our findings emphasize the utility of genome-wide CRISPRi screening in mapping the genetic landscape of antibiotic resistance. This study provides a valuable resource for identifying potential targets for antibiotics or antimicrobial strategies. Moreover, it offers a framework for exploring transcriptional regulatory networks and resistance mechanisms in E. coli and other bacterial pathogens.
Collapse
Affiliation(s)
- Donghui Choe
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Eunju Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
- KI for the BioCentury, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Kangsan Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
- KI for the BioCentury, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Soonkyu Hwang
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
- KI for the BioCentury, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Ki Jun Jeong
- KI for the BioCentury, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
- Graduate School of Engineering Biology, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Bernhard O. Palsson
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Byung-Kwan Cho
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
- KI for the BioCentury, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
- Graduate School of Engineering Biology, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Suhyung Cho
- KI for the BioCentury, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| |
Collapse
|
6
|
Yan X, Tao R, Zhou H, Zhang Y, Chen D, Ma L, Bai Y. Sublethal sanitizers exposure differentially affects biofilm formation in three adapted Salmonella strains: A phenotypic-transcriptomic analysis of increased biofilm formed by ATCC 14028. Int J Food Microbiol 2025; 436:111189. [PMID: 40222328 DOI: 10.1016/j.ijfoodmicro.2025.111189] [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: 03/27/2025] [Accepted: 04/02/2025] [Indexed: 04/15/2025]
Abstract
PURPOSE Using sanitizer in food industry is an important mean of sterilization and biofilm eradication, but inappropriate operation may lead to resistance, posing a concealed risk to food safety. The purpose of this study was to assess the impact of sub-lethal sanitizers on the biofilm formed by adaptive Salmonella and to explore the variations in transcription within adaptive Salmonella biofilms when co-incubated with sublethal concentrations of sanitizers. METHODS The microbroth dilution method was determined to measure the MIC of three sanitizers on Salmonella, and adaptation induction was conducted with steadily increasing sanitizer concentrations. The effect of sub-MIC sanitizers on the biofilm of Salmonella was investigated by crystal violet method, confocal laser scanning microscopy and transcriptomics. RESULTS The results indicated that the maximum growth concentration of the adapted strains was 1.69-43.25 times that of the original MIC, and the number of bacteria and matrix content were increased when re-exposed to sub-MIC benzalkonium chloride (BZK), and the expression of regulatory factors and various amino acid biosynthesis and metabolism-related genes showed an up-regulation trend. SIGNIFICANCE This will be beneficial to clarify the correlation and mechanism between the sanitizer adaptation of salmonellae caused by improper sanitization and increased biofilm formation resulting from this adaptation. And it helps to adjust the appropriate dosage of sanitizer and optimize sanitation standard operating procedures (SSOP) in the foodstuff industry, thereby effectively promoting the bactericidal effect and eliminating foodborne pathogens' biofilm.
Collapse
Affiliation(s)
- Xiaoxue Yan
- College of Food Science, Southwest University, 2 Tiansheng Rd, Beibei, Chongqing 400715, PR China.
| | - Rongfeng Tao
- College of Food Science, Southwest University, 2 Tiansheng Rd, Beibei, Chongqing 400715, PR China.
| | - Hongyuan Zhou
- College of Food Science, Southwest University, 2 Tiansheng Rd, Beibei, Chongqing 400715, PR China; Chongqing Key Laboratory of Specialty Food Co-Built by Sichuan and Chongqing, Chongqing 400715, PR China.
| | - Yuhao Zhang
- College of Food Science, Southwest University, 2 Tiansheng Rd, Beibei, Chongqing 400715, PR China; Chongqing Key Laboratory of Specialty Food Co-Built by Sichuan and Chongqing, Chongqing 400715, PR China; Key Laboratory of Condiment Supervision Technology for State Market Regulation, Chongqing 401121, PR China; Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, Chongqing 400715, PR China.
| | - Dong Chen
- College of Food Science, Southwest University, 2 Tiansheng Rd, Beibei, Chongqing 400715, PR China.
| | - Liang Ma
- College of Food Science, Southwest University, 2 Tiansheng Rd, Beibei, Chongqing 400715, PR China; Chongqing Key Laboratory of Specialty Food Co-Built by Sichuan and Chongqing, Chongqing 400715, PR China; Key Laboratory of Condiment Supervision Technology for State Market Regulation, Chongqing 401121, PR China.
| | - Yamin Bai
- Key Laboratory of Condiment Supervision Technology for State Market Regulation, Chongqing 401121, PR China; Chongqing Institute for Food and Drug Control, Chongqing 401121, PR China
| |
Collapse
|
7
|
Thakur RK, Aggarwal K, Sood N, Kumar A, Joshi S, Jindal P, Maurya R, Patel P, Kurmi BD. Harnessing advances in mechanisms, detection, and strategies to combat antimicrobial resistance. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 982:179641. [PMID: 40373688 DOI: 10.1016/j.scitotenv.2025.179641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 04/28/2025] [Accepted: 05/08/2025] [Indexed: 05/17/2025]
Abstract
Antimicrobial resistance (AMR) is a growing global health crisis, threatening the effectiveness of antibiotics and other antimicrobial agents, leading to increased morbidity, mortality, and economic burdens. This review article provides a comprehensive analysis of AMR, beginning with a timeline of antibiotics discovery and the year of first observed resistance. Main mechanisms of AMR in bacteria, fungi, viruses, and parasites are summarized, and the main mechanisms of bacteria are given in detail. Additionally, we discussed in detail methods for detecting AMR, including phenotypic, genotypic, and advanced methods, which are crucial for identifying and monitoring AMR. In addressing AMR mitigation, we explore innovative interventions such as CRISPR-Cas systems, nanotechnology, antibody therapy, artificial intelligence (AI), and the One Health approach. Moreover, we discussed both finished and ongoing clinical trials for AMR. This review emphasizes the urgent need for global action and highlights promising technologies that could shape the future of AMR surveillance and treatment. By integrating interdisciplinary research and emerging clinical insights, this study aims to guide individuals toward impactful solutions in the battle against AMR.
Collapse
Affiliation(s)
- Ritik Kumar Thakur
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga 142001, Punjab, India
| | - Kaushal Aggarwal
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga 142001, Punjab, India
| | - Nayan Sood
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga 142001, Punjab, India
| | - Aman Kumar
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga 142001, Punjab, India
| | - Sachin Joshi
- Department of Pharmaceutical Quality Assurance, ISF College of Pharmacy, GT Road, Moga 142001, Punjab, India
| | - Priya Jindal
- Department of Pharmaceutical Quality Assurance, ISF College of Pharmacy, GT Road, Moga 142001, Punjab, India
| | - Rashmi Maurya
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga 142001, Punjab, India
| | - Preeti Patel
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, GT Road, Moga 142001, Punjab, India.
| | - Balak Das Kurmi
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga 142001, Punjab, India.
| |
Collapse
|
8
|
Qiu L, Guo X, Shim H, Hao T, Liang Z, Wang S, Lu Z, Lu Q, He Z. Unveiling triclosan biodegradation: Novel metabolic pathways, genomic insights, and global environmental adaptability of Pseudomonas sp. strain W03. JOURNAL OF HAZARDOUS MATERIALS 2025; 488:137313. [PMID: 39862779 DOI: 10.1016/j.jhazmat.2025.137313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 01/12/2025] [Accepted: 01/20/2025] [Indexed: 01/27/2025]
Abstract
The polychlorinated aromatic antimicrobial agent triclosan (TCS) is widely used to indiscriminately and rapidly kill microorganisms. The global use of TCS has led to widespread environmental contamination, posing significant threats to ecosystem and human health. Here we reported a newly isolated Pseudomonas sp. W03 for degrading TCS metabolically at concentrations up to 10 mg/L. This strain exhibited optimal degradation activity at 30°C and pH 7.0, and retained substantial activity at pH 4.0, although it was sensitive to alkaline conditions. Genomic analysis of strain W03 revealed a circular chromosome comprising 6075,907 bp with a GC content of 65.08 %. A novel TCS degradation pathway, involving dechlorination, oxidation, ether bond fission, and reoxidation processes, was identified. Also, the study mapped the global distribution of analogous Pseudomonas using 16S rRNA gene sequences, revealing their widespread presence in diverse aquatic environments, with a significant abundance in wastewater systems. These findings indicated that these bacteria play a critical ecological role in both natural and engineered environments, particularly in the degradation of organic pollutants. This study enhances our understanding of microbial degradation of emerging contaminants and presents a promising candidate for bioremediation strategies aimed at mitigating TCS-related water pollution.
Collapse
Affiliation(s)
- Lan Qiu
- Marine Synthetic Ecology Research Center, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), School of Marine Science, Sun Yat-sen University, Zhuhai 519080, China; Department of Civil and Environmental Engineering, Faculty of Science and Technology, University of Macau, 999078, Macao
| | - Xiaoyuan Guo
- Department of Civil and Environmental Engineering, Faculty of Science and Technology, University of Macau, 999078, Macao
| | - Hojae Shim
- Department of Civil and Environmental Engineering, Faculty of Science and Technology, University of Macau, 999078, Macao
| | - Tianwei Hao
- Department of Civil and Environmental Engineering, Faculty of Science and Technology, University of Macau, 999078, Macao
| | - Zhiwei Liang
- Department of Environmental Science and Engineering, Sun Yat-sen University, Guangzhou 510006, China
| | - Shanquan Wang
- Department of Environmental Science and Engineering, Sun Yat-sen University, Guangzhou 510006, China
| | - Zhenmei Lu
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qihong Lu
- Department of Environmental Science and Engineering, Sun Yat-sen University, Guangzhou 510006, China.
| | - Zhili He
- Marine Synthetic Ecology Research Center, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), School of Marine Science, Sun Yat-sen University, Zhuhai 519080, China.
| |
Collapse
|
9
|
Gao H, Wang B, Li M, Zhou P, Wu C, Wan C, Shen L, Fu J, Han W, Zhou Y, Yu F. Emergence and dissemination of multidrug-resistant Klebsiella pneumoniae harboring the novel tmexCD-toprJ RND efflux pump operon. Front Cell Infect Microbiol 2025; 15:1579880. [PMID: 40370409 PMCID: PMC12075523 DOI: 10.3389/fcimb.2025.1579880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Accepted: 04/07/2025] [Indexed: 05/16/2025] Open
Abstract
The global emergence of multidrug-resistant (MDR) Klebsiella pneumoniae, particularly carbapenem-resistant K. pneumoniae (CRKP), presents a severe public health threat, limiting available treatment options. Tigecycline and eravacycline, have been considered a last-resort therapeutic against MDR Enterobacteriaceae. However, strains were resistant to these antibiotics increased recently. The tmexCD-toprJ, a plasmid-encoded resistance-nodulation-division (RND)-type efflux pump, has emerged as a critical factor conferring resistance to tigecycline and eravacycline. In this study, we reported the emergence of 11 CRKP isolates harboring tmexCD-toprJ, isolated from two lung transplant patients in a tertiary hospital in eastern China. Most of the isolates (82%) exhibited high-level resistance to tigecycline and eravacycline, along with other common antibiotics. Whole-genome sequencing (WGS) and phylogenetic analysis indicated these strains are not clonal, and resistance phenotypes were associated with the tmexCD-toprJ operon and other crucial resistance elements. We also found the tmexCD-toprJ operon was located on a conjugative plasmid, sharing high sequence similarity with the operon identified in Pseudomonas aeruginosa. Our results showed that the tmexCD-toprJ-harboring plasmid is efficiently transferable, which contributes to the dissemination of tigecycline and eravacycline resistance. At the same time, the plasmid can coexist with the blaKPC-2 -carrying plasmid, which may cause multidrug resistance. The emergence of tmexCD-toprJ-positive CRKP in lung transplant patients highlights the potential for rapid nosocomial dissemination and reduced treatment efficacy of last-line antimicrobials. Our findings emphasize the need for enhanced genomic surveillance, infection control measures, and alternative therapeutic strategies to combat the spread of tmexCD-toprJ-mediated resistance in clinical settings.
Collapse
Affiliation(s)
- Haojin Gao
- Department of Clinical Laboratory Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Bingjie Wang
- Department of Clinical Laboratory Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Meilan Li
- Department of Respiratory Intensive Care Unit, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Peiyao Zhou
- Department of Clinical Laboratory Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chunyang Wu
- Department of Endocrinology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Cailing Wan
- Department of Clinical Laboratory Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Li Shen
- Department of Clinical Laboratory Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jiana Fu
- Department of Clinical Laboratory Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Weihua Han
- Department of Clinical Laboratory Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ying Zhou
- Department of Clinical Laboratory Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Fangyou Yu
- Department of Clinical Laboratory Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
10
|
Tang H, Liu Z, Hu B, Zhu L. Hierarchical activation of resistance genes under tetracyclines selective pressure in complex microbial community. JOURNAL OF HAZARDOUS MATERIALS 2025; 493:138399. [PMID: 40300515 DOI: 10.1016/j.jhazmat.2025.138399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 04/21/2025] [Accepted: 04/22/2025] [Indexed: 05/01/2025]
Abstract
The pervasive use of antibiotics exerts selective pressure in both natural and anthropogenic environments, driving the propagation and evolution of antibiotic resistance genes (ARGs) in microbial communities. Understanding the succession of resistome under varying antibiotic stresses is crucial for mitigating the spread of ARGs. This study investigates the succession of resistome under exposure to four structurally different tetracyclines (TC) across concentrations ranging from environmental to clinical levels. A clear hierarchical activation of ARGs was observed, starting with the upregulation of multidrug and TC-specific efflux pump genes, followed by those involved in TC inactivation and ribosomal protection. By identifying the specific thresholds of transcriptional onset times and critical TC concentration ranges that triggered ARG abundance increases, it was found that all ARGs as a whole did not significantly increase when TC concentrations were maintained below 10-5 of the initial minimum inhibitory concentration (MIC0) within 2 h. Similarly, high-risk TC resistance genes do not proliferate when TC concentrations were kept below 10-3 × MIC0 within 24 h. These findings provide quantifiable benchmarks for concentration-time thresholds that can inform the establishment of environmental discharge limits and guide the implementation of targeted treatment technologies to mitigate ARG dissemination.
Collapse
Affiliation(s)
- Huiming Tang
- College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China; Zhejiang Provincial Key Laboratory of Organic Pollution Process and Control, Zhejiang University, Hangzhou 310058, China
| | - Zishu Liu
- College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China
| | - Baolan Hu
- College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China
| | - Lizhong Zhu
- College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China; Zhejiang Provincial Key Laboratory of Organic Pollution Process and Control, Zhejiang University, Hangzhou 310058, China; State Key Laboratory of Soil Pollution Control and Safety, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
11
|
Kinose K, Shinoda K, Kawasaki H. Impact of exporter proteins and their engineering on the productivity of Corynebacterium. Appl Microbiol Biotechnol 2025; 109:98. [PMID: 40261395 PMCID: PMC12014714 DOI: 10.1007/s00253-025-13479-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 04/01/2025] [Accepted: 04/02/2025] [Indexed: 04/24/2025]
Abstract
Enhancing product efflux is crucial in improving fermentative bioproduction. Despite advancements in metabolic engineering guided by the design-build-test-learn cycle, membrane transport engineering of product efflux remains underdeveloped, limiting the efficient production of target chemicals. This review explores the historical findings on product efflux, regardless of passive or active transport, in fermentation engineering, focusing on Corynebacterium species, and highlights the potential of multidrug transporters as valuable screening sources for efflux improvement. Furthermore, the review emphasizes the importance of understanding the machinery of efflux transporters to optimize their functionality. Molecular dynamics simulations are a promising tool for exploring novel strategies to advance fermentation-related processes. These insights provide a framework for overcoming current challenges in membrane transport engineering of product efflux and improving industrial-scale bioproduction. KEY POINTS: • Review of strategies to enhance product efflux in Corynebacterium species. • Multidrug transporters are key tools for optimizing metabolite efflux. • Efflux transporter mechanisms analyzed to improve microbial productivity. • Molecular dynamics simulations employed for understanding transporter mechanisms.
Collapse
Affiliation(s)
- Keita Kinose
- Nagahama Institute for Biochemical Science, Oriental Yeast Co., Ltd., 50 Kano-Cho, Nagahama, Shiga, 526 - 0804, Japan
| | - Keiko Shinoda
- The Institute of Statistical Mathematics, Research Organization of Information and Systems, 10 - 3 Midori-Cho, Tachikawa, Tokyo, 190 - 8562, Japan
| | - Hisashi Kawasaki
- Graduate School of Agriculture and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-Ku, Tokyo, 113 - 8657, Japan.
- Research and Development Division, Kikkoman Corporation, 338 Noda, Noda City, Chiba, 278 - 0037, Japan.
| |
Collapse
|
12
|
Ciobanasu C. Bacterial Extracellular Vesicles and Antimicrobial Peptides: A Synergistic Approach to Overcome Antimicrobial Resistance. Antibiotics (Basel) 2025; 14:414. [PMID: 40298572 PMCID: PMC12024098 DOI: 10.3390/antibiotics14040414] [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: 03/31/2025] [Revised: 04/14/2025] [Accepted: 04/17/2025] [Indexed: 04/30/2025] Open
Abstract
Antimicrobial resistance is already a major global health threat, contributing to nearly 5 million deaths annually. The rise of multidrug-resistant pathogens has made many infections increasingly difficult to treat. This growing threat has driven the search for alternative therapeutic approaches. Among the most promising candidates are bacterial extracellular vesicles (BEVs) and antimicrobial peptides (AMPs), which offer unique mechanisms of action, potential synergistic effects, and the ability to bypass conventional resistance pathways. This review summarizes the current research on synergistic effects of BEVs and AMPs to overcome antimicrobial resistance.
Collapse
Affiliation(s)
- Corina Ciobanasu
- Department of Exact and Natural Sciences, Institute of Interdisciplinary Research, Alexandru I. Cuza University, Bulevardul Carol I, Nr. 11, 700506 Iasi, Romania
| |
Collapse
|
13
|
Shen S, He Z, Zhao S, Zhu Z, Wang X, Tian Y, Han Y, Hu M, Lu C, Li A. Environmental high-risk efflux pumps mediate concurrent enhancement of resistance and virulence in reclaimed water from urban wastewater treatment plants. JOURNAL OF HAZARDOUS MATERIALS 2025; 493:138236. [PMID: 40273845 DOI: 10.1016/j.jhazmat.2025.138236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 03/12/2025] [Accepted: 04/08/2025] [Indexed: 04/26/2025]
Abstract
With the growing demand for urban water, reclaimed water is a key solution for mitigating shortages. However, wastewater treatment plants (WWTPs) can act as reservoirs for antibiotic resistance genes (ARGs) and virulence factors (VFs), posing environmental and public health risks. This study systematically investigated the distribution and co-selection mechanisms of ARGs and VFs across four treatment stages in five WWTPs in Nanjing over two years, with a focus on high-risk efflux pump-related ARGs. A total of 902 persistent ARGs and 1086 persistent VFs were identified, including 12 characteristic ARGs, 12 characteristic VFs, and 20 high-risk priority ARGs. Network analysis and linear regression revealed a significant correlation between efflux pump ARGs and VFs, particularly those linked to outer membrane proteins, pili, and iron ion transport. Their coexistence suggests potential selective advantages during wastewater treatment. Laboratory-simulated aquatic environment experiments, RT-qPCR, and virulence protein assays further confirmed the co-selection between resistance and virulence. Results demonstrated that environmental stress, including antibiotics, heavy metals, and disinfection, induces efflux pump overexpression, enhancing bacterial pathogenicity. These findings highlight the importance of monitoring high-risk efflux pump ARGs in reclaimed water safety. Targeted surveillance and advanced treatment strategies are crucial for controlling antibiotic resistance and ensuring safe water reuse.
Collapse
Affiliation(s)
- Shanqi Shen
- State Key Laboratory of State Key Laboratory of Water Pollution Control and Green Resource Recycling, School of the Environment, Nanjing University, Nanjing 210023, China; Yangze River Innovation Center For Ecological Civilization, Nanjing 210019, China
| | - Zhong He
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Siya Zhao
- State Key Laboratory of State Key Laboratory of Water Pollution Control and Green Resource Recycling, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Zezhang Zhu
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Xinyi Wang
- State Key Laboratory of State Key Laboratory of Water Pollution Control and Green Resource Recycling, School of the Environment, Nanjing University, Nanjing 210023, China; Yangze River Innovation Center For Ecological Civilization, Nanjing 210019, China
| | - Yechao Tian
- State Key Laboratory of State Key Laboratory of Water Pollution Control and Green Resource Recycling, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Yuze Han
- State Key Laboratory of State Key Laboratory of Water Pollution Control and Green Resource Recycling, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Min Hu
- State Key Laboratory of State Key Laboratory of Water Pollution Control and Green Resource Recycling, School of the Environment, Nanjing University, Nanjing 210023, China; Yangze River Innovation Center For Ecological Civilization, Nanjing 210019, China
| | - Chang Lu
- State Key Laboratory of State Key Laboratory of Water Pollution Control and Green Resource Recycling, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Aimin Li
- State Key Laboratory of State Key Laboratory of Water Pollution Control and Green Resource Recycling, School of the Environment, Nanjing University, Nanjing 210023, China; Yangze River Innovation Center For Ecological Civilization, Nanjing 210019, China.
| |
Collapse
|
14
|
Hu M, Chua SL. Antibiotic-Resistant Pseudomonas aeruginosa: Current Challenges and Emerging Alternative Therapies. Microorganisms 2025; 13:913. [PMID: 40284749 PMCID: PMC12029751 DOI: 10.3390/microorganisms13040913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 04/04/2025] [Accepted: 04/10/2025] [Indexed: 04/29/2025] Open
Abstract
Antibiotic-resistant Pseudomonas aeruginosa is a pathogen notorious for its resilience in clinical settings due to biofilm formation, efflux pumps, and the rapid acquisition of resistance genes. With traditional antibiotic therapy rendered ineffective against Pseudomonas aeruginosa infections, we explore alternative therapies that have shown promise, including antimicrobial peptides, nanoparticles and quorum sensing inhibitors. While these approaches offer potential, they each face challenges, such as specificity, stability, and delivery, which require careful consideration and further study. We also delve into emerging alternative strategies, such as bacteriophage therapy and CRISPR-Cas gene editing that could enhance targeted treatment for personalized medicine. As most of them are currently in experimental stages, we highlight the need for clinical trials and additional research to confirm their feasibility. Hence, we offer insights into new therapeutic avenues that could help address the pressing issue of antibiotic-resistant Pseudomonas aeruginosa, with an eye toward practical applications in future healthcare.
Collapse
Affiliation(s)
- Minqi Hu
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Song Lin Chua
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
- State Key Laboratory of Chemical Biology and Drug Discovery, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
- Research Centre for Deep Space Explorations (RCDSE), The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
- Research Institute for Future Food (RiFood), The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| |
Collapse
|
15
|
Wang C, Wei X, Zhong L, Chan CL, Li H, Sun H. Metal-Based Approaches for the Fight against Antimicrobial Resistance: Mechanisms, Opportunities, and Challenges. J Am Chem Soc 2025; 147:12361-12380. [PMID: 40063057 PMCID: PMC12007004 DOI: 10.1021/jacs.4c16035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 04/17/2025]
Abstract
The rapid emergency and spread of antimicrobial-resistant (AMR) bacteria and the lack of new antibiotics being developed pose serious threats to the global healthcare system. Therefore, the development of more effective therapies to overcome AMR is highly desirable. Metal ions have a long history of serving as antimicrobial agents, and metal-based compounds are now attracting more interest from scientific communities in the fight against AMR owing to their unique mechanism. Moreover, they may also serve as antibiotic adjuvants to enhance the efficacy of clinically used antibiotics. In this perspective, we highlight important showcase studies in the last 10 years on the development of metal-based strategies to overcome the AMR crisis. Specifically, we categorize these metallo-antimicrobials into five classes based on their modes of action (i.e., metallo-enzymes and metal-binding enzyme inhibitors, membrane perturbants, uptake/efflux system inhibitors/regulators, persisters inhibitors, and oxidative stress inducers). The significant advantages of metallo-antimicrobials over traditional antibiotics lie in their multitargeted mechanisms, which render less likelihood to generate resistance. However, we notice that such modes of action of metallo-antimicrobials may also raise concern over their potential side effects owing to the low selectivity toward pathogens and host, which appears to be the biggest obstacle for downstream translational research. We anticipate that combination therapy through repurposing (metallo)drugs with antibiotics and the optimization of their absorption route through formulation to achieve a target-oriented delivery will be a powerful way to combat AMR. Despite significant challenges, metallo-antimicrobials hold great opportunities for the therapeutic intervention of infection by resistant bacteria.
Collapse
Affiliation(s)
- Chenyuan Wang
- Department
of Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, PR China
- CAS-HKU
Joint Laboratory of Metallomics for Health and Environment, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, PR China
| | - Xueying Wei
- Department
of Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, PR China
- Department
of Microbiology, The University of Hong
Kong, Pokfulam Road, Hong Kong SAR, PR China
| | - Liang Zhong
- Department
of Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, PR China
| | - Chun-Lung Chan
- Department
of Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, PR China
| | - Hongyan Li
- Department
of Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, PR China
- CAS-HKU
Joint Laboratory of Metallomics for Health and Environment, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, PR China
- State
Key Laboratory of Synthetic Chemistry, The
University of Hong Kong, Pokfulam Road, Hong Kong SAR, PR China
| | - Hongzhe Sun
- Department
of Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, PR China
- CAS-HKU
Joint Laboratory of Metallomics for Health and Environment, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, PR China
- State
Key Laboratory of Synthetic Chemistry, The
University of Hong Kong, Pokfulam Road, Hong Kong SAR, PR China
| |
Collapse
|
16
|
Sun S. Emerging antibiotic resistance by various novel proteins/enzymes. Eur J Clin Microbiol Infect Dis 2025:10.1007/s10096-025-05126-4. [PMID: 40232578 DOI: 10.1007/s10096-025-05126-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 04/02/2025] [Indexed: 04/16/2025]
Abstract
BACKGROUND The emergence and dissemination of antibiotic resistance represents a significant and ever-increasing global threat to human, animal, and environmental health. The explosive proliferation of resistance has ultimately been seen in all clinically used antibiotics. Infections caused by antibiotic-resistant bacteria have been associated with an estimated 4,950,000 deaths annually, with extremely limited therapeutic options and only a few new antibiotics under development. To combat this silent pandemic, a better understanding of the molecular mechanisms of antibiotic resistance is immensely needed, which not only helps to improve the efficacy of current drugs in clinical use but also design new antimicrobial agents that are less susceptible to resistance. RESULTS The past few years have witnessed a number of new advances in revealing the molecular mechanisms of AMR. Following five sophisticated mechanisms (efflux pump, antibiotics inactivation by enzymes, alteration of membrane permeability, target modification, and target protection), the roles of various novel proteins/enzymes in the acquisition of antibiotic resistance are constantly being described. They are widely used by clinical bacterial strains, playing a key role in the emergence of resistance. CONCLUSION While most of these have so far received less attention, expanding our understanding of these emerging resistance mechanisms is of crucial importance to combat the antibiotic resistance crisis in the world. This review summarizes recent advances in our knowledge of emerging resistance mechanisms in bacteria, providing an update on the current antibiotic resistance threats and encouraging researchers to develop critical strategies for overcoming the resistance.
Collapse
Affiliation(s)
- Shengwei Sun
- School of Engineering Sciences in Chemistry, Biotechnology and Health, Department of Fibre and Polymer Technology, KTH Royal Institute of Technology, Stockholm, SE-100 44, Sweden.
- School of Engineering Sciences in Chemistry, Biotechnology and Health, Science for Life Laboratory, Tomtebodavägen 23, Solna, 171 65, Sweden.
| |
Collapse
|
17
|
Santos Barbosa CRD, Menezes Dantas DD, Bezerra SR, Rocha JE, Freitas Alexandre PR, Marinho ES, Queiroz Almeida-Neto FWD, Tintino SR, Ribeiro de Sousa G, Barbosa-Filho JM, Souza-Ferrari JD, Costa MRD, Coutinho HDM, Teixeira AMR, Santos HSD, Cunha FABD. Electronic properties and adjuvant effect of riparins I-IV: Inhibition of β-lactamase and QacC efflux pump in Staphylococcus aureus K4100. Biochem Biophys Res Commun 2025; 758:151636. [PMID: 40112534 DOI: 10.1016/j.bbrc.2025.151636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 03/10/2025] [Accepted: 03/14/2025] [Indexed: 03/22/2025]
Abstract
Staphylococcus aureus is a leading cause of nosocomial infections, posing a significant health threat due to its resistance mechanisms, particularly involving β-lactamase enzymes and efflux pumps. Targeting these mechanisms is crucial to restore the efficacy of antibiotics. This study characterized the electronic properties of riparins I, II, III, and IV and evaluated their effects on the β-lactamase enzyme and the QacC efflux pump in the S. aureus K4100 strain. The electronic properties of the riparins revealed distinct electrophilic characteristics, but similar nucleophilic behavior, as indicated by the HOMO (Highest Occupied Molecular Orbital) and LUMO (Lowest Unoccupied Molecular Orbital) orbital energy values. Microbiological testing showed that riparins I, II, III, and IV did not display direct antibacterial activity against S. aureus K4100. However, riparin III significantly reduced the MIC of oxacillin, suggesting it potentiates the antibiotic's effect, likely by targeting the β-lactamase enzyme. Furthermore, riparins II and III lowered the MIC of ethidium bromide, indicating their potential as inhibitors of the QacC efflux pump. These findings highlight the potential of riparins II and III as adjuvants to enhance the effectiveness of antibiotics against multidrug-resistant strains of S. aureus.
Collapse
Affiliation(s)
| | - Débora de Menezes Dantas
- Department of Biological Chemistry, Regional University of Cariri (URCA), Crato, 63105-000, CE, Brazil
| | - Suieny Rodrigues Bezerra
- Laboratory of Bioprospection of Semiarid and Alternative Methods of the Regional University of Cariri - LABSEMA, Crato, Cear'a, Brazil.
| | - Janaína Esmeraldo Rocha
- Department of Biological Chemistry, Regional University of Cariri (URCA), Crato, 63105-000, CE, Brazil.
| | | | - Emmanuel Silva Marinho
- Center for Exact Sciences and Technology, State University of Ceará (UECE), Fortaleza, 60714-903, CE, Brazil.
| | | | - Saulo Relison Tintino
- Department of Biological Chemistry, Regional University of Cariri (URCA), Crato, 63105-000, CE, Brazil.
| | - Gabriela Ribeiro de Sousa
- Postgraduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraiba, João Pessoa, 58051-900, PB, Brazil
| | - José Maria Barbosa-Filho
- Postgraduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraiba, João Pessoa, 58051-900, PB, Brazil.
| | | | | | - Henrique Douglas Melo Coutinho
- Coordinator of the Laboratory of Microbiology and Molecular Biology (LMBM), Regional University of Cariri (URCA), Crato, CE, Brazil.
| | | | - Hélcio Silva Dos Santos
- Program Rede Nordeste de Biotecnologia (RENORBIO-Nucleadora UECE), Universidade Estadual Vale do Acaraú (UVA), Sobral, CE, Brazil.
| | | |
Collapse
|
18
|
Yu M, Wu Y, Zhao W, Li N, Ding W, Liu J, Miu X, Hou J, Liu Y, Pan B, Xie L, Han L, Wang C. The impact of differential expression levels of smeE gene on antimicrobial susceptibility and other biological functions in Stenotrophomonas maltophilia CYZ. Int J Biol Macromol 2025; 310:143075. [PMID: 40220818 DOI: 10.1016/j.ijbiomac.2025.143075] [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: 10/29/2024] [Revised: 03/28/2025] [Accepted: 04/09/2025] [Indexed: 04/14/2025]
Abstract
Stenotrophomonas maltophilia is an important opportunistic pathogen that utilizes SmeE efflux pump to extrude structurally dissimilar antibiotics. Here, we constructed smeE gene deletion (D531) and overexpression (O531) strains of S. maltophilia CYZ, and investigated changes in biological functions by analyzing the transcriptional and expression levels of differentially expressed genes and proteins, respectively. S. maltophilia D531 showed significant susceptibility to erythromycin, quinolones, and tetracycline. The organism exhibited stronger bacterial motility, which was due to the upregulated expressions of FlhA and MotB proteins. Because the expression levels of SM01853, OhrB, DnaK, GrpE, and HslJ proteins were downregulated, S. maltophilia D531 increased susceptibilities to H2O2 and high temperature. Conversely, S. maltophilia O531 exhibited resistance to H2O2 and high temperature and enhanced biofilm-forming capacity, since the expressions of KatE, SM02481, PilO and PilQ proteins in S. maltophilia O531 were upregulated. However, both S. maltophilia D531 and O531 reduced tolerance towards Zn2+, and the expression of Zn transporter was found to be downregulated. Additionally, it is necessary to demonstrate whether downregulation of SmeN and SM02901 proteins resulted in S. maltophilia O531 reduced resistance to aminoglycosides. The current study may give important leads in elucidating the changes in biological functions via transcriptomic and proteomic analyses.
Collapse
Affiliation(s)
- Menghan Yu
- Department of Pathogenic Biology, Hebei Medical University, Shijiazhuang 050017, PR China; Public Research Platform, School of Basic Medical Sciences, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Yan Wu
- Department of Pathogenic Biology, Hebei Medical University, Shijiazhuang 050017, PR China; Public Research Platform, School of Basic Medical Sciences, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Wenting Zhao
- Department of Pathogenic Biology, Hebei Medical University, Shijiazhuang 050017, PR China; Public Research Platform, School of Basic Medical Sciences, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Nan Li
- First School of Clinical Medicine, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Wenxi Ding
- Department of Pathogenic Biology, Hebei Medical University, Shijiazhuang 050017, PR China; Public Research Platform, School of Basic Medical Sciences, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Jinxu Liu
- First School of Clinical Medicine, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Xiaojin Miu
- First School of Clinical Medicine, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Jiahuan Hou
- First School of Clinical Medicine, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Yawen Liu
- Department of Pathogenic Biology, Hebei Medical University, Shijiazhuang 050017, PR China; Public Research Platform, School of Basic Medical Sciences, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Bingxin Pan
- Department of Pathogenic Biology, Hebei Medical University, Shijiazhuang 050017, PR China; Public Research Platform, School of Basic Medical Sciences, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Lixin Xie
- Department of Experimental Center of Teaching, Hebei Medical University, Shijiazhuang 050017, PR China.
| | - Lujuan Han
- Department of Pathogenic Biology, Hebei Medical University, Shijiazhuang 050017, PR China; Public Research Platform, School of Basic Medical Sciences, Hebei Medical University, Shijiazhuang 050017, PR China.
| | - Changle Wang
- Department of Pathogenic Biology, Hebei Medical University, Shijiazhuang 050017, PR China; Public Research Platform, School of Basic Medical Sciences, Hebei Medical University, Shijiazhuang 050017, PR China.
| |
Collapse
|
19
|
Lawrence R, Athar M, Uddin MR, Adams C, Sousa JS, Durrant O, Lellman S, Sutton L, Keevil CW, Patel N, Prosser C, McMillan D, Zgurskaya HI, Vargiu AV, Ahdash Z, Reading E. Molecular basis for multidrug efflux by an anaerobic RND transporter. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.04.646765. [PMID: 40236129 PMCID: PMC11996581 DOI: 10.1101/2025.04.04.646765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Bacteria can resist antibiotics and toxic substances within demanding ecological settings, such as low oxygen, extreme acid, and during nutrient starvation. MdtEF, a proton motive force-driven efflux pump from the resistance-nodulation-cell division (RND) superfamily, is upregulated in these conditions but its molecular mechanism is unknown. Here, we report cryo-electron microscopy structures of Escherichia coli multidrug transporter MdtF within native-lipid nanodiscs, including a single-point mutant with an altered multidrug phenotype and associated substrate-bound form. We reveal that drug binding domain and channel conformational plasticity likely governs promiscuous substrate specificity, analogous to its closely related, constitutively expressed counterpart, AcrB. Whereas we discover distinct transmembrane state transitions within MdtF, which create a more engaged proton relay network, altered drug transport allostery and an acid-responsive increase in efflux efficiency. Physiologically, this provides means of xenobiotic and metabolite disposal within remodelled cell membranes that presage encounters with acid stresses, as endured in the gastrointestinal tract.
Collapse
|
20
|
Niu N, Dou LN, Yang S, Wang HX, Zhuang S, Fan YP, Liu YQ, Zhang WM, Ma WR. Drug resistance detection of canine origin Escherichia coli in China and inhibition by genipin. Vet J 2025; 310:106307. [PMID: 39889817 DOI: 10.1016/j.tvjl.2025.106307] [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: 09/04/2024] [Revised: 01/24/2025] [Accepted: 01/24/2025] [Indexed: 02/03/2025]
Abstract
Zoonotic transmission from pets to their owners is a major health problem. It is important to determine and control the drug resistance in pets to mitigate risks of human transmission. In this work, the current prevalence of multi-drug resistance (MDR) and resistance gene in Escherichia coli (E. coli) derived from dogs in nine cities across various regions of China initially evaluated using microfluidic dilution methods and polymerase chain reaction (PCR) technology. To control antibiotic resistance, genipin as natural product was used to combat MDR E. coli. Finally, the synergistic effect of genipin and norfloxacin on MDR E. coli was studied using time-kill curves to retard the resistance spread. A total of 126 E. coli strains were isolated from 154 collected fecal samples of dogs. Minimum inhibitory concentrations (MIC) results revealed that the highest detection rate of MDR E. coli appeared in Zhengzhou at 90.9 %, and the lowest in Shenyang at 10.0 %. The results of drug resistance gene testing indicated that the blaTEM gene had the highest detection rate (99.2 %), then tetA and blaCTX-M-1, whose detection rates all exceed 50 %. Furthermore, the MIC of genipin against MDR E. coli was found to be 4096 μg/mL, and genipin at ½ MIC demonstrated significant inhibition on MDR E. coli within 6 h. Finally, the combination of ¼ MIC genipin with ½ MIC norfloxacin showed partial synergistic inhibitory effect on MDR E. coli. Our findings suggest that although antibiotic resistance in canine origin E. coli varies across different regions of China, it remains concerning, and genipin shows potential as a treatment option for MDR E. coli infections.
Collapse
Affiliation(s)
- Nan Niu
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, PR China; Institute of Traditional Chinese Veterinary Medicine, Northwest A&F University, Yangling 712100, PR China
| | - Lei-Na Dou
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, PR China
| | - Shuo Yang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, PR China; Institute of Traditional Chinese Veterinary Medicine, Northwest A&F University, Yangling 712100, PR China
| | - Hai-Xin Wang
- College of Veterinary Medicine, Hebei Agricultural University, Baoding 071000, PR China
| | - Shen Zhuang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, PR China; Institute of Traditional Chinese Veterinary Medicine, Northwest A&F University, Yangling 712100, PR China
| | - Yun-Peng Fan
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, PR China; Institute of Traditional Chinese Veterinary Medicine, Northwest A&F University, Yangling 712100, PR China
| | - Ying-Qiu Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, PR China; Institute of Traditional Chinese Veterinary Medicine, Northwest A&F University, Yangling 712100, PR China
| | - Wei-Min Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, PR China; Institute of Traditional Chinese Veterinary Medicine, Northwest A&F University, Yangling 712100, PR China
| | - Wu-Ren Ma
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, PR China; Institute of Traditional Chinese Veterinary Medicine, Northwest A&F University, Yangling 712100, PR China; Xi'an Veterinary Teaching Hospital, Northwest A&F University, Xi'an 710065, PR China.
| |
Collapse
|
21
|
Freitas PR, Araújo ACJD, Araújo IM, Almeida RS, Oliveira Borges JAD, Lima CMG, Oliveira-Tintino CDM, Paulo CLR, Miranda GM, Araújo-Neto JBD, Almeida-Bezerra JW, Nascimento IJDS, Araújo-Júnior JXD, Silva-Júnior EFD, Aquino TMD, Mendonca Junior FJB, Marinho ES, Santos HSD, Menezes IRAD, Tintino SR, Coutinho HDM. Evaluating efflux pump inhibition in Staphylococcus aureus 1199B strain using thiadiazine-derived compounds: In vitro and in silico approaches. Biochimie 2025; 231:104-109. [PMID: 39708974 DOI: 10.1016/j.biochi.2024.12.009] [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: 07/24/2024] [Revised: 12/03/2024] [Accepted: 12/19/2024] [Indexed: 12/23/2024]
Abstract
Thiadiazines are heterocyclic compounds known for some pharmacological activities. However, the ability of these compounds and their derivatives to act as antibacterial agents and inhibitors of the efflux system in resistant bacteria remains unknown. This study aims to evaluate the antibacterial and NorA efflux pump inhibitory activities of thiadiazine-derived compounds (IJ14, IJ15, IJ16, IJ17, IJ18, IJ19, and IJ20) against the Staphylococcus aureus 1199B strain. Minimum Inhibitory Concentration (MIC) tests and antibacterial activity assessment through NorA efflux system inhibition were performed using microdilution assays in 96-well plates. Additionally, ethidium bromide (EtBr) fluorescence emission assays were conducted to evaluate efflux system inhibition. The methodology revealed that the IJ17 and IJ20 compounds presented MIC values of 256 and 597.3 μg/mL, respectively. The efflux pump inhibition assessment using the microdilution method showed significant results for all compounds, which also increased the fluorescence rates emitted by EtBr. Consequently, thiadiazine-derived compounds exhibit promising results in targeting a key bacterial resistance mechanism, underscoring the need for further studies, such as molecular tests, to evaluate their mechanism of action and clarify the feasibility and efficacy of these compounds as antibacterial agents.
Collapse
Affiliation(s)
| | | | - Isaac Moura Araújo
- Department of Biological Chemistry, Universidade Regional Do Cariri, Crato, CE, Brazil
| | - Ray Silva Almeida
- Department of Biological Chemistry, Universidade Regional Do Cariri, Crato, CE, Brazil
| | | | | | | | | | | | - José Bezerra de Araújo-Neto
- Postgraduate Program in Biological Sciences, Biosciences Center, Federal University of Pernambuco, Recife, PE, 50740-570, Brazil
| | | | - Igor José Dos Santos Nascimento
- Laboratory of Medicinal Chemistry, Institute of Pharmaceutical Sciences, Federal University of Alagoas, 57072-900, Maceió, AL, Brazil
| | - João Xavier de Araújo-Júnior
- Laboratory of Medicinal Chemistry, Institute of Pharmaceutical Sciences, Federal University of Alagoas, 57072-900, Maceió, AL, Brazil
| | - Edeildo Ferreira da Silva-Júnior
- Biological and Molecular Chemistry Research Group, Institute of Chemistry and Biotechnology, Federal University of Alagoas, 57072-900, Maceió, AL, Brazil
| | - Thiago Mendonça de Aquino
- Biological and Molecular Chemistry Research Group, Institute of Chemistry and Biotechnology, Federal University of Alagoas, 57072-900, Maceió, AL, Brazil
| | | | | | | | | | - Saulo Relison Tintino
- Department of Biological Chemistry, Universidade Regional Do Cariri, Crato, CE, Brazil
| | | |
Collapse
|
22
|
Liu X, Song M, Liu Y, Yang S, Chen S, Kang J, Shen J, Zhu K. Rational Design of Natural Xanthones Against Gram-negative Bacteria. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411923. [PMID: 39888292 PMCID: PMC11984908 DOI: 10.1002/advs.202411923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 12/21/2024] [Indexed: 02/01/2025]
Abstract
Most antibiotics are ineffective against Gram-negative bacteria owing to the existence of the outer membrane (OM) barrier. The rational design of compounds to expand their antibacterial spectra of antibiotics solely targeting Gram-positive pathogens remains challenging. Here, the design of skeletons from natural products to penetrate the OM are deciphered. Structure-activity relationship analysis shows the optimization of the model of natural xanthones α-mangostin endows the broad-spectrum antibacterial activity. Mechanistic studies demonstrate the lead compound A20 penetrates the OM in a self-promoted pathway through electronic and hydrophobic interactions with lipopolysaccharides and phospholipids in OM. A20 displays rapid bactericidal activity by targeting the cofactor heme in the respiratory complex. The therapeutic efficacy of A20 is demonstrated in two animal models infected with multidrug-resistant Gram-negative bacterial pathogens. The findings elucidate the structural property and self-promoted transportation of a class of antibacterial compounds, to facilitate the design and discovery of antibacterial agents against increasingly prevalent Gram-negative pathogens associated with infections.
Collapse
Affiliation(s)
- Xiaojia Liu
- National Key Laboratory of Veterinary Public Health and SafetyCollege of Veterinary MedicineChina Agricultural UniversityNo.2 Yuanmingyuan West RoadBeijing100193China
| | - Meirong Song
- National Key Laboratory of Veterinary Public Health and SafetyCollege of Veterinary MedicineChina Agricultural UniversityNo.2 Yuanmingyuan West RoadBeijing100193China
| | - Ying Liu
- National Key Laboratory of Veterinary Public Health and SafetyCollege of Veterinary MedicineChina Agricultural UniversityNo.2 Yuanmingyuan West RoadBeijing100193China
| | - Shuyu Yang
- National Key Laboratory of Veterinary Public Health and SafetyCollege of Veterinary MedicineChina Agricultural UniversityNo.2 Yuanmingyuan West RoadBeijing100193China
| | - Shang Chen
- National Key Laboratory of Veterinary Public Health and SafetyCollege of Veterinary MedicineChina Agricultural UniversityNo.2 Yuanmingyuan West RoadBeijing100193China
| | - Jijun Kang
- National Key Laboratory of Veterinary Public Health and SafetyCollege of Veterinary MedicineChina Agricultural UniversityNo.2 Yuanmingyuan West RoadBeijing100193China
| | - Jianzhong Shen
- National Key Laboratory of Veterinary Public Health and SafetyCollege of Veterinary MedicineChina Agricultural UniversityNo.2 Yuanmingyuan West RoadBeijing100193China
| | - Kui Zhu
- National Key Laboratory of Veterinary Public Health and SafetyCollege of Veterinary MedicineChina Agricultural UniversityNo.2 Yuanmingyuan West RoadBeijing100193China
| |
Collapse
|
23
|
Ojha S, Sinsinwar S, Chatterjee P, Biswal S, Pradhan P, Beuria TK. Efflux pump modulation by Montelukast and its roles in restoring antibiotic susceptibility in multidrug-resistant Staphylococcus aureus. EBioMedicine 2025; 114:105658. [PMID: 40157128 PMCID: PMC11994358 DOI: 10.1016/j.ebiom.2025.105658] [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: 09/11/2024] [Revised: 02/22/2025] [Accepted: 03/05/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND Staphylococcus aureus and its drug-resistant mutants are mentioned among the WHO's high-priority list of pathogens. Antibiotics like fluoroquinolones and cephalosporins are used to treat multidrug-resistant S. aureus infections. However, a higher expression of efflux pumps (NorA, NorB, and AbcA) induces multidrug resistance. The master regulator, MgrA, regulates the expression of most of these efflux pumps in S. aureus. The phosphorylation status of MgrA is determined by the cellular PknB/RsbU ratio, where PknB, a serine-threonine kinase, and RsbU, a serine-threonine phosphatase, are critical for MgrA functioning. METHODS An FDA-approved drug library was screened using an EtBr-accumulation assay to identify efflux pump inhibitors (EPIs). The synergy of EPIs with antibiotics was studied in vitro and in vivo in the murine skin infection model of female BALB/c mice. The effect of EPIs on mgrA, norB, pknB, and rsbU gene expression, interaction with MgrA, and effects on MgrA phosphorylation were studied. FINDINGS We identified Montelukast as an effective EPI, which showed synergy with moxifloxacin, a substrate of the NorB efflux pump, both in vitro and in the murine skin infection model. Further, Montelukast decreased norB expression and increased the pknB/rsbU expression ratio. Our in vitro results demonstrated that Montelukast strongly interacted with MgrA, facilitated MgrA phosphorylation, and enhanced its affinity for the norB promoter. INTERPRETATION Our study showed that Montelukast repressed MgrA expression and promoted MgrA phosphorylation to suppress norB expression and efflux pump activity, leading to the restoration of antibiotic susceptibility in multidrug-resistant S. aureus. FUNDING The study was supported by SERB-DST, India (CRG/2021/005069), and the BRIC-ILS core.
Collapse
Affiliation(s)
- Suvendu Ojha
- Department of Infectious Disease Biology, Institute of Life Sciences, NALCO Square, Chandrasekharpur, Bhubaneswar, Odisha, 751023, India; Regional Centre for Biotechnology, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad Rd, Faridabad, Haryana, 121001, India
| | - Simran Sinsinwar
- Department of Infectious Disease Biology, Institute of Life Sciences, NALCO Square, Chandrasekharpur, Bhubaneswar, Odisha, 751023, India
| | - Puja Chatterjee
- Department of Infectious Disease Biology, Institute of Life Sciences, NALCO Square, Chandrasekharpur, Bhubaneswar, Odisha, 751023, India; Regional Centre for Biotechnology, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad Rd, Faridabad, Haryana, 121001, India
| | - Sarmistha Biswal
- Department of Infectious Disease Biology, Institute of Life Sciences, NALCO Square, Chandrasekharpur, Bhubaneswar, Odisha, 751023, India
| | - Pinkilata Pradhan
- Department of Infectious Disease Biology, Institute of Life Sciences, NALCO Square, Chandrasekharpur, Bhubaneswar, Odisha, 751023, India; Regional Centre for Biotechnology, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad Rd, Faridabad, Haryana, 121001, India
| | - Tushar Kant Beuria
- Department of Infectious Disease Biology, Institute of Life Sciences, NALCO Square, Chandrasekharpur, Bhubaneswar, Odisha, 751023, India.
| |
Collapse
|
24
|
Tanner JD, Richards SN, Corry B. Molecular basis of the functional conflict between chloroquine and peptide transport in the Malaria parasite chloroquine resistance transporter PfCRT. Nat Commun 2025; 16:2987. [PMID: 40140375 PMCID: PMC11947230 DOI: 10.1038/s41467-025-58244-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 03/16/2025] [Indexed: 03/28/2025] Open
Abstract
The Plasmodium falciparum chloroquine resistance transporter (PfCRT) is a key protein contributing to resistance against the antimalarial chloroquine (CQ). Mutations such as K76T enable PfCRT to transport CQ away from its target in the parasite's digestive vacuole, but this comes at a cost to its natural peptide transport function. This creates fitness costs which can drive changes to drug susceptibility in parasite populations, but the molecular basis of this is not well understood. To investigate, here we run 130 μs of molecular dynamics simulations of CQ-sensitive and CQ-resistant PfCRT isoforms with CQ and peptide substrates. We identify the CQ binding site and characterized diverse peptide binding modes. The K76T mutation allows CQ to access the binding site but disrupts peptide binding, highlighting the importance of cavity charge in determining substrate specificity. This study provides insight into PfCRT polyspecific peptide transport and will aid in rational, structure-based inhibitor design.
Collapse
Affiliation(s)
- John D Tanner
- Research School of Biology, Australian National University, Canberra, ACT, Australia
| | - Sashika N Richards
- Research School of Biology, Australian National University, Canberra, ACT, Australia
| | - Ben Corry
- Research School of Biology, Australian National University, Canberra, ACT, Australia.
| |
Collapse
|
25
|
Qiao J, Zhu W, Du D, Morigen M. Characterizing Common Factors Affecting Replication Initiation During H 2O 2 Exposure and Genetic Mutation-Induced Oxidative Stress in Escherichia coli. Int J Mol Sci 2025; 26:2968. [PMID: 40243598 PMCID: PMC11989076 DOI: 10.3390/ijms26072968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/13/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
Oxidative stress is prevalent in organisms, and excessive oxidative damage can trigger cell death. Bacteria have evolved multiple pathways to cope with adverse stress, including the regulation of the cell cycle. Previous studies show that non-lethal exposure to H2O2 and mutations in antioxidant enzymes suppress replication initiation in Escherichia coli. The existence of common regulatory factors governing replication initiation across diverse causes-induced oxidative stress remains unclear. In this study, we utilized flow cytometry to determine the replication pattern of E. coli, and found that oxidative stress also participated in the inhibition of replication initiation by a defective iron regulation (fur-bfr-dps deletion). Adding a certain level of ATP promoted replication initiation in various antioxidant enzyme-deficient mutants and the ΔfurΔbfrΔdps mutant, suggesting that low ATP levels could be a common factor in the inhibition of replication initiation by different causes-induced oxidative stress. More potential common factors were screened using proteomics, followed by genetic validation with H2O2 stress. We found that oxidative stress might mediate the inhibition of replication initiation by interfering with the metabolism of glycine, glutamate, ornithine, and aspartate. Blocking CcmA-dependent cytochrome c biosynthesis, deleting the efflux pump proteins MdtABCD and TolC, or the arabinose transporter AraFHG eliminated the replication initiation inhibition by H2O2. In conclusion, this study uncovers a common multifactorial pathway of different causes-induced oxidative stress inhibiting replication initiation. Dormant and persistent bacteria exhibit an arrested or slow cell cycle, and non-lethal oxidative stress promotes their formation. Our findings contribute to exploring strategies to limit dormant and persistent bacterial formation by maintaining faster DNA replication initiation (cell cycle progression).
Collapse
Affiliation(s)
- Jiaxin Qiao
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences, Inner Mongolia University, Hohhot 010070, China; (J.Q.); (D.D.)
| | - Weiwei Zhu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang-An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Dongdong Du
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences, Inner Mongolia University, Hohhot 010070, China; (J.Q.); (D.D.)
| | - Morigen Morigen
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences, Inner Mongolia University, Hohhot 010070, China; (J.Q.); (D.D.)
| |
Collapse
|
26
|
Vaccaro M, Pilat AM, Gusmano L, Pham MTN, Barich D, Gibson A, Epalle M, Frost DJ, Volin E, Slimak ZC, Menke CC, Fennessy MS, Slonczewski JL. Pond water microbiome antibiotic resistance genes vary seasonally with environmental pH and tannins. Microbiol Spectr 2025; 13:e0303424. [PMID: 40130858 PMCID: PMC12054064 DOI: 10.1128/spectrum.03034-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 02/22/2025] [Indexed: 03/26/2025] Open
Abstract
Microbial communities of small freshwater bodies interact dynamically with environmental factors in unknown ways. Longitudinal sampling of four ponds in Knox County, Ohio, revealed relationships among antibiotic resistance genes (ARGs) and environmental factors such as pH and tannin concentrations. For each site, microbial communities were collected by filtration, and metagenomes were analyzed by short-read sequencing. ARGs were quantified using the ShortBRED pipeline to detect and quantify hits to a marker set derived from the Comprehensive Antibiotic Resistance Database. The top 30 ARGs showed increased abundance at the end of the growing season. The top two ARGs with the largest marker hits encode components of a Stenotrophomonas drug efflux pump powered by proton-motive force (smeABC) and a mycobacterial global regulator that activates a drug pump and acid stress response (mtrA). The smeABC and mtrA prevalence showed a modest correlation with acidifying conditions (low pH and high tannic acids). Acidity amplifies the transmembrane pH difference component of the proton-motive force, thus increasing the cell's energy available for pump function and ARG expression. Association with microbial taxa was tested by the Kraken2/Bracken predictor of taxa profiles. The ARG profiles showed the strongest acid dependence in ponds with a high proportion of Proteobacteria, whereas a pond with high Cyanobacteria showed the lowest ARG counts. Efflux pumps such as SmeABC and transcriptional activation by MtrA incur large energy expenditures whose function may be favored at low external pH, where the cell's proton-motive force is maximal. IMPORTANCE Compared to rivers and lakes, pond microbial ecosystems are understudied despite close contact with agriculture and recreation. Environmental microbes offer health benefits as well as hazards for human contact. Small water bodies may act as reservoirs for drug-resistant organisms and transfer of antibiotic resistance genes (ARGs). Yet, the public is rarely aware of the potential for exposure to ARG-carrying organisms in recreational water bodies. Little is known about the capacity of freshwater microbial communities to remediate drug pollution and which biochemical factors may select against antibiotic resistance genes. This study analyzes how aquatic ARG prevalence may depend on environmental factors such as pH and tannic acid levels.
Collapse
Affiliation(s)
- Maya Vaccaro
- Department of Biology, Kenyon College, Gambier, Ohio, USA
| | | | - Logan Gusmano
- Department of Biology, Kenyon College, Gambier, Ohio, USA
| | | | - Daniel Barich
- Department of Biology, Kenyon College, Gambier, Ohio, USA
| | - Audrey Gibson
- Department of Biology, Kenyon College, Gambier, Ohio, USA
| | - Mwï Epalle
- Department of Biology, Kenyon College, Gambier, Ohio, USA
| | | | | | | | | | | | | |
Collapse
|
27
|
Harmon DE, Adamson K, Wiersma J. Disruption of the EmrAB-TolC efflux pump of Escherichia coli induces global metabolic changes in multiple growth conditions. MICROPUBLICATION BIOLOGY 2025; 2025:10.17912/micropub.biology.001437. [PMID: 40191445 PMCID: PMC11971657 DOI: 10.17912/micropub.biology.001437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 03/05/2025] [Accepted: 03/21/2025] [Indexed: 04/09/2025]
Abstract
Multidrug efflux pumps are transporters that are important for the removal of exogenous toxic molecules in bacteria. Recently, efflux pumps have been implicated in the regulation of metabolic homeostasis in Escherichia coli . Here, we investigated the contribution of EmrAB-TolC to metabolism in various conditions. Deletion of EmrB led to changes in several metabolic pathways, both in standard growth conditions and during nutrient stress. The pathways impacted include the tricarboxylic acid (TCA) cycle, and carbohydrate and amino acid metabolism. Our findings suggest that EmrAB-TolC contributes to maintaining metabolic homeostasis and adapts metabolism based on cellular needs.
Collapse
Affiliation(s)
- Dana E. Harmon
- Biology, California Lutheran University, Thousand Oaks, California, United States
| | - Klay Adamson
- Biology, California Lutheran University, Thousand Oaks, California, United States
| | - Joseph Wiersma
- Biology, California Lutheran University, Thousand Oaks, California, United States
| |
Collapse
|
28
|
Khunweeraphong N, Kuchler K. The human ABCG2 transporter engages three gates to control multidrug extrusion. iScience 2025; 28:112125. [PMID: 40165990 PMCID: PMC11957596 DOI: 10.1016/j.isci.2025.112125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 01/17/2025] [Accepted: 02/25/2025] [Indexed: 04/02/2025] Open
Abstract
The human ABCG2 transporter plays roles in physiological detoxification across barriers and in anticancer multidrug resistance. The translocation pathway for drug extrusion and its gating mechanism remains elusive. Here, we demonstrate that the ABCG2 multidrug transporter holds two cavities that are delineated by three regulatory gates, indicating a substrate translocation channel. Drugs are trapped in the central cavity after entering through the pivotal intracellular entry gate. This flexible cavity is surrounded by a cluster of three highly conserved phenylalanines. Their aromatic side chains enact a "clamp-push-seal" motion to ensure unidirectional substrate movement. The unique residues T435 and N436 act as critical selectors for ligands, determining the broad substrate specificity. The upper cavity is covered by the lid architecture, constituting the final gate before multidrug extrusion. This work unravels deep mechanistic details on how the translocation channel utilizes pivotal gating steps, including the sequence of events that drive ABCG2-mediated multidrug efflux.
Collapse
Affiliation(s)
- Narakorn Khunweeraphong
- Medical University of Vienna, Max Perutz Labs Vienna, Center for Medical Biochemistry Campus Vienna Biocenter, Dr. Bohr-Gasse 9/2, 1030 Vienna, Austria
| | - Karl Kuchler
- Medical University of Vienna, Max Perutz Labs Vienna, Center for Medical Biochemistry Campus Vienna Biocenter, Dr. Bohr-Gasse 9/2, 1030 Vienna, Austria
| |
Collapse
|
29
|
Cueny R, Voter A, McKenzie A, Morgenstern M, Myers K, Place M, Peters J, Coon J, Keck J. Altering translation allows E. coli to overcome G-quadruplex stabilizers. Nucleic Acids Res 2025; 53:gkaf264. [PMID: 40193707 PMCID: PMC11975287 DOI: 10.1093/nar/gkaf264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 03/14/2025] [Accepted: 03/21/2025] [Indexed: 04/09/2025] Open
Abstract
G-quadruplex (G4) structures can form in guanine-rich DNA or RNA and have been found to modulate cellular processes, including replication, transcription, and translation. Many studies on the cellular roles of G4s have focused on eukaryotic systems, with far fewer probing bacterial G4s. Using a chemical-genetic approach, we identified genes in Escherichia coli that are important for growth in G4-stabilizing conditions. Reducing levels of translation elongation factor Tu or slowing translation initiation or elongation with kasugamycin, chloramphenicol, or spectinomycin suppress the effects of G4-stabilizing compounds. In contrast, reducing the expression of specific translation termination or ribosome recycling proteins is detrimental to growth in G4-stabilizing conditions. Proteomic and transcriptomic analyses reveal decreased protein and transcript levels, respectively, for ribosome assembly factors and proteins associated with translation in the presence of G4 stabilizer. Our results support a model in which reducing the rate of translation by altering translation initiation, translation elongation, or ribosome assembly can compensate for G4-related stress in E. coli.
Collapse
Affiliation(s)
- Rachel R Cueny
- Biomolecular Chemistry Department, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53706 United States
| | - Andrew F Voter
- Biomolecular Chemistry Department, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53706 United States
| | - Aidan M McKenzie
- Biomolecular Chemistry Department, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53706 United States
| | - Marcel Morgenstern
- Biomolecular Chemistry Department, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53706 United States
- National Center for Quantitative Biology of Complex Systems, University of Wisconsin-Madison, Madison, WI, 53706 United States
| | - Kevin S Myers
- Great Lakes Bioenergy Research Center and the Wisconsin Energy Institute, University of Wisconsin-Madison, Madison, WI, 53726 United States
| | - Michael M Place
- Great Lakes Bioenergy Research Center and the Wisconsin Energy Institute, University of Wisconsin-Madison, Madison, WI, 53726 United States
| | - Jason M Peters
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53706 United States
| | - Joshua J Coon
- Biomolecular Chemistry Department, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53706 United States
- National Center for Quantitative Biology of Complex Systems, University of Wisconsin-Madison, Madison, WI, 53706 United States
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53706 United States
- Morgridge Institute for Research, Madison, WI, 53715 United States
| | - James L Keck
- Biomolecular Chemistry Department, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53706 United States
| |
Collapse
|
30
|
Xiao-Quan Y, Jian-Ting H, Han-Zhong F, Jun H, Zhi-Ping W, He YX. The flavonoid-sensing regulator AefR is involved in modulating quorum sensing through repressing the MexEF-OprN efflux pump in Pseudomonas fluorescens. mSystems 2025; 10:e0091524. [PMID: 40013790 PMCID: PMC11915855 DOI: 10.1128/msystems.00915-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 02/04/2025] [Indexed: 02/28/2025] Open
Abstract
Flavonoids, a major component of plant root exudates, play a crucial role in mediating plant-microbe interactions. However, the mechanisms by which flavonoids are perceived and trigger downstream signaling events in microbes remain largely unknown. In this study, we characterized AefR, a flavonoid-sensing transcriptional regulator from Pseudomonas fluorescens 2P24, a plant growth-promoting rhizobacterium (PGPR) known for its biocontrol properties. AefR was found to repress the expression of the mexEF-oprN efflux pump, which putatively exports N-acylhomoserine lactones (AHLs). This repression attenuates the PcoR/PcoI quorum-sensing system, leading to decreased production of the antibiotic mupirocin in P. fluorescens 2P24. Furthermore, quantitative proteomic analysis revealed that the PcoR/PcoI quorum-sensing system regulates a diverse range of physiological processes, including mupirocin production and denitrification. Collectively, these findings demonstrate a quorum-quenching role of flavonoids in a PGPR strain, establishing that flavonoids can disrupt quorum-sensing by enhancing the efflux of quorum-sensing signaling molecules. These findings have practical implications for the development of sustainable biocontrol strategies, where leveraging natural plant-microbe interactions could enhance the suppression of plant pathogens without the use of synthetic chemicals.IMPORTANCEFlavonoids are key mediators of plant-microbe interactions; however, their role in regulating microbial signaling remains poorly understood. This study identifies AefR as a flavonoid-sensing regulator in Pseudomonas fluorescens 2P24, revealing a novel quorum-quenching mechanism where flavonoids enhance the efflux of quorum-sensing signals. These findings shed light on the molecular basis of flavonoid-mediated microbial regulation and offer new strategies for sustainable plant health management.
Collapse
Affiliation(s)
- Yu Xiao-Quan
- Institute of Urology, Gansu Province Clinical Research Center for Urinary System Disease, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu, China
| | - Han Jian-Ting
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Feng Han-Zhong
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Hou Jun
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Wang Zhi-Ping
- Institute of Urology, Gansu Province Clinical Research Center for Urinary System Disease, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu, China
| | - Yong-Xing He
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
31
|
Liu J, Zhang L, Ma H, Sun H, Ge SA, Liu J, Fan S, Quan C. Quaternary ammonium chitosan-functionalized mesoporous silica nanoparticles: A promising targeted drug delivery system for the treatment of intracellular MRSA infection. Carbohydr Polym 2025; 352:123184. [PMID: 39843087 DOI: 10.1016/j.carbpol.2024.123184] [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: 08/13/2024] [Revised: 11/28/2024] [Accepted: 12/22/2024] [Indexed: 01/30/2025]
Abstract
The limited membrane permeability and bacterial resistance pose significant challenges in the management of intracellular drug-resistant bacterial infections. To overcome this issue, we developed a bacterial-targeted drug delivery system based on quaternary ammonium chitosan-modified mesoporous silica nanoparticles (MSN-NH2-CFP@HACC) for the treatment of intracellular Methicillin-resistant Staphylococcus aureus (MRSA) infections. This system utilizes amino-functionalized mesoporous silica nanoparticles to efficiently load cefoperazone (CFP), and the nanoparticles' surface is coated with 2-hydroxypropyltrimethyl ammonium chloride chitosan (HACC) to target bacteria and enhance macrophage uptake. The findings indicate that MSN-NH2-CFP@HACC nanoparticles are efficiently internalized by macrophages, demonstrate accelerated drug release in acidic environments, and exhibit enhanced antibacterial properties, effectively suppressing the proliferation and intracellular escape of MRSA. Moreover, HACC enhances the bacterial capture ability of the nanoparticles and reduces resistance by disrupting bacterial membrane structures and inhibiting bacterial β-lactamase activity. In a murine model of MRSA bacteremia, MSN-NH2-CFP@HACC exhibited remarkable antibacterial efficacy and significantly attenuated severe inflammatory responses. In conclusion, MSN-NH2-CFP@HACC represent a promising antibiotic delivery system with exceptional antibacterial efficacy and favorable biocompatibility, thus presenting a novel strategy for addressing intracellular drug-resistant bacterial infections and demonstrating significant potential for clinical application.
Collapse
Affiliation(s)
- Junfeng Liu
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, College of Life Science, Dalian Minzu University, Dalian 116600, PR China; Department of Bioengineering, College of Life Science, Dalian Minzu University, Dalian 116600, PR China
| | - Liying Zhang
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, College of Life Science, Dalian Minzu University, Dalian 116600, PR China; Department of Bioengineering, College of Life Science, Dalian Minzu University, Dalian 116600, PR China
| | - Haodi Ma
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, College of Life Science, Dalian Minzu University, Dalian 116600, PR China; Department of Bioengineering, College of Life Science, Dalian Minzu University, Dalian 116600, PR China
| | - Haoyang Sun
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, College of Life Science, Dalian Minzu University, Dalian 116600, PR China; Department of Bioengineering, College of Life Science, Dalian Minzu University, Dalian 116600, PR China
| | - Shu-Ai Ge
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, College of Life Science, Dalian Minzu University, Dalian 116600, PR China; Department of Bioengineering, College of Life Science, Dalian Minzu University, Dalian 116600, PR China
| | - Jieyi Liu
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, College of Life Science, Dalian Minzu University, Dalian 116600, PR China; Department of Bioengineering, College of Life Science, Dalian Minzu University, Dalian 116600, PR China
| | - Shengdi Fan
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, College of Life Science, Dalian Minzu University, Dalian 116600, PR China; Department of Bioengineering, College of Life Science, Dalian Minzu University, Dalian 116600, PR China
| | - Chunshan Quan
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, College of Life Science, Dalian Minzu University, Dalian 116600, PR China; Department of Bioengineering, College of Life Science, Dalian Minzu University, Dalian 116600, PR China.
| |
Collapse
|
32
|
Zhu C, Diao Z, Yang Y, Liao J, Wang C, Li Y, Liang Z, Xu P, Liu X, Zhang Q, Gong L, Ma Q, Liang L, Lin Z. Recent advances and challenges in metal-based antimicrobial materials: a review of strategies to combat antibiotic resistance. J Nanobiotechnology 2025; 23:193. [PMID: 40059157 PMCID: PMC11892188 DOI: 10.1186/s12951-025-03249-6] [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: 10/06/2024] [Accepted: 02/18/2025] [Indexed: 05/13/2025] Open
Abstract
Despite the availability of a series of classical antibiotic drugs, bacterial infections continue to represent a significant and urgent threat to global human health. The emergence of drug-resistant bacteria and the slow pace of antibiotic development have rendered current treatment methods inadequate in meeting the clinical demands of bacterial infections. Consequently, there is an increasingly urgent and vital need for the development of safe, efficient, and alternative novel antimicrobial agents in the medical and healthcare field. Over the past five years, there has been a notable expansion in the field of nanomedicine with regard to the prevention and control of infectious diseases. The objective of this article is to provide a comprehensive review of the latest research developments in the field of metal nanomaterials for medical antimicrobial therapy. We begin by delineating the gravity of the bacterial infection crisis, subsequently undertaking a comprehensive examination of the potential mechanisms through which nanoparticles may combat bacterial infections and the specific applications of these nanomaterials in the treatment of diverse infectious diseases. In conclusion, we eagerly anticipate the future development directions of metal nanomaterials in the field of antimicrobial therapy. We believe that with continuous technological advancements and innovations, this field will make even more outstanding contributions to safeguarding human health and well-being.
Collapse
Affiliation(s)
- Chuanda Zhu
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Zhenli Diao
- Department of Clinical Laboratory, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100043, China
| | - Yuanyuan Yang
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Jun Liao
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Chao Wang
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Yanglonghao Li
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Zichao Liang
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Pengcheng Xu
- School of Pharmaceutical Sciences, Inner Mongolia Medical University, Hohhot, 010110, China
| | - Xinyu Liu
- Beijing Life Science Academy, Beijing, 102200, China
| | - Qiang Zhang
- School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Lidong Gong
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China.
| | - Qiang Ma
- School of Basic Medical Sciences, North China University of Science and Technology, Tangshan City, 063210, Hebei Province, China.
| | - Ling Liang
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China.
| | - Zhiqiang Lin
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China.
| |
Collapse
|
33
|
Russell Lewis B, Uddin MR, Kuo KM, Shah LMN, Harris NJ, Booth PJ, Hammerschmid D, Gumbart JC, Zgurskaya HI, Reading E. Mg 2+-dependent mechanism of environmental versatility in a multidrug efflux pump. Structure 2025; 33:552-565.e4. [PMID: 39809273 DOI: 10.1016/j.str.2024.12.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 11/15/2024] [Accepted: 12/15/2024] [Indexed: 01/16/2025]
Abstract
Tripartite resistance nodulation and cell division multidrug efflux pumps span the periplasm and are major drivers of multidrug resistance among gram-negative bacteria. Cations, such as Mg2+, become concentrated within the periplasm and, in contrast to the cytoplasm, its pH is sensitive to conditions outside the cell. Here, we reveal an interplay between Mg2+ and pH in modulating the structural dynamics of the periplasmic adapter protein, AcrA, and its function within the prototypical AcrAB-TolC multidrug pump from Escherichia coli. In the absence of Mg2+, AcrA becomes increasingly plastic within acidic conditions, but when Mg2+ is bound this is ameliorated, resulting instead in domain specific organization. We establish a unique histidine residue directs these dynamics and is essential for sustaining pump activity across acidic, neutral, and basic regimes. Overall, we propose Mg2+ conserves AcrA structural mobility to ensure optimal AcrAB-TolC function within rapidly changing environments commonly faced during bacterial infection and colonization.
Collapse
Affiliation(s)
- Benjamin Russell Lewis
- Department of Chemistry, Britannia House, 7 Trinity Street, King's College London, London, SE1 1DB, UK
| | - Muhammad R Uddin
- Department of Chemistry and Biochemistry, University of Oklahoma, 101 Stephenson Parkway, Norman, OK 73019, USA
| | - Katie M Kuo
- School of Physics, Georgia Institute of Technology, 837 State Street NW, Atlanta, GA 30332, USA
| | - Laila M N Shah
- Department of Chemistry, Britannia House, 7 Trinity Street, King's College London, London, SE1 1DB, UK
| | - Nicola J Harris
- Department of Chemistry, Britannia House, 7 Trinity Street, King's College London, London, SE1 1DB, UK
| | - Paula J Booth
- Department of Chemistry, Britannia House, 7 Trinity Street, King's College London, London, SE1 1DB, UK
| | - Dietmar Hammerschmid
- Department of Chemistry, Britannia House, 7 Trinity Street, King's College London, London, SE1 1DB, UK; School of Biological Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| | - James C Gumbart
- School of Physics, Georgia Institute of Technology, 837 State Street NW, Atlanta, GA 30332, USA; School of Chemistry and Biochemistry, Georgia Institute of Technology, 901 Atlantic Drive NW, Atlanta, GA 30332, USA.
| | - Helen I Zgurskaya
- Department of Chemistry and Biochemistry, University of Oklahoma, 101 Stephenson Parkway, Norman, OK 73019, USA.
| | - Eamonn Reading
- Department of Chemistry, Britannia House, 7 Trinity Street, King's College London, London, SE1 1DB, UK; School of Biological Sciences, University of Southampton, Southampton, SO17 1BJ, UK.
| |
Collapse
|
34
|
Deng K, Zhang Y, Lv S, Zhang C, Xiao L. Decoding Pecan's Fungal Foe: A Genomic Insight into Colletotrichum plurivorum Isolate W-6. J Fungi (Basel) 2025; 11:203. [PMID: 40137241 PMCID: PMC11943440 DOI: 10.3390/jof11030203] [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: 01/17/2025] [Revised: 02/19/2025] [Accepted: 02/21/2025] [Indexed: 03/27/2025] Open
Abstract
Pecan (Carya illinoinensis) is a world-renowned nut crop that is highly favored by consumers for its high content of healthy nutrients. For a long time, anthracnose has severely threatened the yield and quality of pecan, causing significant economic losses to the global pecan industry. Here, we report the 54.57-Mb gapless chromosome-level assembly of the pathogenic ascomycetes Colletotrichum plurivorum isolate W-6 from pecan plantations in Southeast China. Six of 12 chromosomes contain, at least, telomeric repeats (CCCTAA)n or (TTAGGG)n at one end. A total of 14,343 protein-coding genes were predicted. Pathogenicity- and virulence-related annotations revealed 137 to 4558 genes associated with the TCDB, PHI, Cyt_P450, DFVF, effector, and secretome databases, respectively. A comparative analysis of isolate W-6, together with 51 other Colletotrichum strains, reveled 13 genes unique to the Orchidearum complex to which isolate W-6 belongs, highlighting the major facilitator superfamily transporters. The detailed analyses of MFS transporters associated with secondary metabolite gene clusters in isolate W-6 led to the identification and protein structure analyses of two key virulence factor candidates in DHA1 subclass, prlG and azaK, which were reported as efflux transporters of antibiotics in other pathogenic fungi. The assembly and further functional investigation of two pathogenic genes identified here potentially provide important resources for better understanding the biology and lifestyle of Colletotrichum and pave the way for designing more efficient strategies to control anthracnose in pecan plantations.
Collapse
Affiliation(s)
- Ke Deng
- College of Forestry and Biotechnology, Zhejiang A&F University, Hangzhou 311300, China; (K.D.); (Y.Z.); (S.L.)
| | - Ying Zhang
- College of Forestry and Biotechnology, Zhejiang A&F University, Hangzhou 311300, China; (K.D.); (Y.Z.); (S.L.)
| | - Saibin Lv
- College of Forestry and Biotechnology, Zhejiang A&F University, Hangzhou 311300, China; (K.D.); (Y.Z.); (S.L.)
| | - Chulong Zhang
- Zhejiang Key Laboratory of Biology and Ecological Regulation of Crop Pathogens and Insects, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, China;
- Ministry of Agriculture and Rural Affairs Key Laboratory of Molecular Biology of Crop Pathogens and Insect Pests, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, China
| | - Lihong Xiao
- College of Forestry and Biotechnology, Zhejiang A&F University, Hangzhou 311300, China; (K.D.); (Y.Z.); (S.L.)
| |
Collapse
|
35
|
Xie P, Zhang Y, Qin Y, Fang Y, Yang N, Bai Y, Zhi S, Niu W, Wang F, Yuan X. Macrolide resistance in Mycoplasma pneumoniae in adult patients. Front Cell Infect Microbiol 2025; 15:1496521. [PMID: 40166372 PMCID: PMC11955812 DOI: 10.3389/fcimb.2025.1496521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 02/12/2025] [Indexed: 04/02/2025] Open
Abstract
Mycoplasma pneumoniae is one of the most significant pathogens responsible for respiratory infections in humans. Macrolides are recommended as the first-line treatment for M. pneumoniae infection. The prevalence of macrolide-resistant M. pneumoniae has increased significantly in recent decades, particularly in China. The mechanisms of resistance in M. pneumoniae to macrolides have been extensively studied in pediatric patients. However, a paucity reports regarding the resistance characteristics and mechanisms exhibited in adults. The aim of this study was to elucidate the resistance of M. pneumoniae to macrolides and the underlying mechanisms in adult patients. Pharyngeal swab specimens were collected from adult patients presenting with subacute cough or community-acquired pneumonia at our hospital from January 2011 to June 2017 to identify and isolate M. pneumoniae strains. The antimicrobial susceptibility of these isolates to 3 macrolide antibiotics was assessed using broth microdilution method. The 23S rRNA genes of macrolide-resistant M. pneumoniae strains were sequenced, and the presence of target methylation genes (ermA, ermB, and ermC), efflux pump genes (mefA, mefA/E, msrA, and msrA/B), and the macrolide resistance gene mphC was identified through polymerase chain reaction (PCR) testing. Additionally, MICs were determined with and without the efflux pump inhibitor reserpine. A total of 72 M. pneumoniae strains were isolated from adult patients, with 41.7% (30/72) exhibiting macrolide resistance. Among the 3 macrolides tested, the 16-membered-ring midecamycin exhibited the greatest activity (MIC90: 16 µg/ml) against M. pneumoniae. All macrolide-resistant M. pneumoniae strains harbored mutations at the 2063 site in domain V of the 23S rRNA gene. Two macrolide-resistant M. pneumoniae clinical isolates were found to harbor the efflux pump genes msrA/B and mefA. The efflux pump inhibitor reserpine reduced the MIC for azithromycin in these two strains to a quarter of their original values. In summary, macrolide-resistant M. pneumoniae is commonly observed among adults in Beijing. Point mutations are the primary mechanism responsible for macrolide resistance in adults with M. pneumoniae. Additionally, the efflux pump mechanism may contribute partially to this resistance. Midecamycin presents a promising alternative drug for treating M. pneumoniae infections, particularly in cases of azithromycin-resistant M. pneumoniae infection in young children.
Collapse
Affiliation(s)
- Panpan Xie
- Department of Respiratory and Critical Care Medicine, Senior Department of Infectious Diseases, the Fifth Medical Center of PLA General Hospital, Beijing, China
- The Fifth Clinical Medical College, Anhui Medical University, Hefei, Anhui, China
| | - Yue Zhang
- Department of Respiratory and Critical Care Medicine, Senior Department of Infectious Diseases, the Fifth Medical Center of PLA General Hospital, Beijing, China
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Yanhong Qin
- Department of Respiratory and Critical Care Medicine, Senior Department of Infectious Diseases, the Fifth Medical Center of PLA General Hospital, Beijing, China
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Yun Fang
- Department of Respiratory and Critical Care Medicine, Senior Department of Infectious Diseases, the Fifth Medical Center of PLA General Hospital, Beijing, China
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Ning Yang
- Department of Respiratory and Critical Care Medicine, Senior Department of Infectious Diseases, the Fifth Medical Center of PLA General Hospital, Beijing, China
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Yunbiao Bai
- Department of Respiratory and Critical Care Medicine, Senior Department of Infectious Diseases, the Fifth Medical Center of PLA General Hospital, Beijing, China
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Shimeng Zhi
- Department of Respiratory and Critical Care Medicine, Senior Department of Infectious Diseases, the Fifth Medical Center of PLA General Hospital, Beijing, China
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Wenkai Niu
- Department of Respiratory and Critical Care Medicine, Senior Department of Infectious Diseases, the Fifth Medical Center of PLA General Hospital, Beijing, China
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Fusheng Wang
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Xin Yuan
- Department of Respiratory and Critical Care Medicine, Senior Department of Infectious Diseases, the Fifth Medical Center of PLA General Hospital, Beijing, China
- The Fifth Clinical Medical College, Anhui Medical University, Hefei, Anhui, China
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| |
Collapse
|
36
|
Tao E, Corry B. Drugs exhibit diverse binding modes and access routes in the Nav1.5 cardiac sodium channel pore. J Gen Physiol 2025; 157:e202413658. [PMID: 39774837 PMCID: PMC11706274 DOI: 10.1085/jgp.202413658] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 11/05/2024] [Accepted: 12/07/2024] [Indexed: 01/11/2025] Open
Abstract
Small molecule inhibitors of the sodium channel are common pharmacological agents used to treat a variety of cardiac and nervous system pathologies. They act on the channel via binding within the pore to directly block the sodium conduction pathway and/or modulate the channel to favor a non-conductive state. Despite their abundant clinical use, we lack specific knowledge of their protein-drug interactions and the subtle variations between different compound structures. This study investigates the binding and accessibility of nine different compounds in the pore cavity of the Nav1.5 sodium channel using enhanced sampling simulations. We find that most compounds share a common location of pore binding-near the mouth of the DII-III fenestration-associated with the high number of aromatic residues in this region. In contrast, some other compounds prefer binding within the lateral fenestrations where they compete with lipids, rather than binding in the central cavity. Overall, our simulation results suggest that the drug binding within the pore is highly promiscuous, with most drugs having multiple low-affinity binding sites. Access to the pore interior via two out of four of the hydrophobic fenestrations is favorable for the majority of compounds. Our results indicate that the polyspecific and diffuse binding of inhibitors in the pore contributes to the varied nature of their inhibitory effects and can be exploited for future drug discovery and optimization.
Collapse
Affiliation(s)
- Elaine Tao
- Division of Biomedical Science and Biochemistry, Research School of Biology, Australian National University, Canberra, Australia
| | - Ben Corry
- Division of Biomedical Science and Biochemistry, Research School of Biology, Australian National University, Canberra, Australia
| |
Collapse
|
37
|
Yan J, Zhang X, Shi X, Wu J, Zhou Z, Tang Y, Bao Z, Luo N, Zhang D, Chen J, Zhang H. Metagenomic insights into the rapid recovery mechanisms of prokaryotic community and spread of antibiotic resistance genes after seawater disinfection. WATER RESEARCH 2025; 271:122887. [PMID: 39637691 DOI: 10.1016/j.watres.2024.122887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 10/26/2024] [Accepted: 11/28/2024] [Indexed: 12/07/2024]
Abstract
Disinfectants, such as bleaching powder, are widely employed in marine aquaculture worldwide to control the bacterial pathogens and eliminate antibiotic resistance genes (ARGs). Nevertheless, the rapid recovery of prokaryotic community compositions (PCCs) after disinfection may significantly influence the overall efficacy of disinfection. Presently, little is known about the rapid recovery mechanisms of PCCs and its impact on the removal of ARGs in seawater. In this study, 16S rRNA gene sequencing and metagenomic analysis were used to address the above concerns through simulating the disinfection process in aquaculture. The results showed that recovery of PCCs began within 16 h. The underlying mechanisms of the rapid recovery of PCCs were the synergistic interactions between microbes and the residues of disinfection-resistant bacteria (DRB). Disinfection resistance genes (DRGs) related to efflux pump serve as the primary molecular foundation providing DRB to resist disinfection. Among the 78 annotated ARGs, only 10 ARGs exhibited a significant decrease (P < 0.05) after 72 h, implying the ineffective removal of ARGs by bleaching powder. Furthermore, bacterial co-resistance to disinfectants and antibiotics was observed. Genome analysis of two highly resistant DRB from Pseudomonadaceae revealed that both DRB carried 16 DRGs, aiding the recovery of PCCs and the spread of ARGs. These findings provide novel insights in the mechanisms of the rapid recovery of PCCs and bacterial co-resistance to disinfectants and antibiotics, which can be crucial for the management of pathogens and antibiotic resistance in seawater.
Collapse
Affiliation(s)
- Jiaojiao Yan
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China; Shanghai Treatgut Biotechnology Co., Ltd., Shanghai, 200441, China; Key Laboratory of Aquacultural Biotechnology, Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, 315211, China
| | - Xinxu Zhang
- Archaeal Biology Center, Synthetic Biology Research Center, Shenzhen Key Laboratory of Marine Microbiome Engineering, Key Laboratory of Marine Microbiome Engineering of Guangdong Higher Education Institutes, Institute for Advanced Study, Shenzhen University, Shenzhen, 518060, China; Key Laboratory of Aquacultural Biotechnology, Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, 315211, China
| | - Xinyong Shi
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China; Key Laboratory of Aquacultural Biotechnology, Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, 315211, China
| | - Jialin Wu
- Ningbo Haiwei Ecological Technology Co., Ltd., Ningbo, 315141, China
| | - Ziang Zhou
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China; Key Laboratory of Aquacultural Biotechnology, Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, 315211, China
| | - Yawen Tang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China; Key Laboratory of Aquacultural Biotechnology, Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, 315211, China
| | - Zhen Bao
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China; Key Laboratory of Aquacultural Biotechnology, Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, 315211, China
| | - Nan Luo
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China; Key Laboratory of Aquacultural Biotechnology, Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, 315211, China
| | - Demin Zhang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China; Key Laboratory of Aquacultural Biotechnology, Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, 315211, China.
| | - Jiong Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China; Key Laboratory of Aquacultural Biotechnology, Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, 315211, China
| | - Huajun Zhang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China; Marine Economic Research Center, Donghai Academy, Ningbo University, Ningbo, 315211, China; Key Laboratory of Aquacultural Biotechnology, Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, 315211, China.
| |
Collapse
|
38
|
Dhanda G, Singh H, Gupta A, Abdul Mohid S, Biswas K, Mukherjee R, Mukherjee S, Bhunia A, Nair NN, Haldar J. Dual-Functional Antibiotic Adjuvant Displays Potency against Complicated Gram-Negative Bacterial Infections and Exhibits Immunomodulatory Properties. ACS CENTRAL SCIENCE 2025; 11:279-293. [PMID: 40028349 PMCID: PMC11868958 DOI: 10.1021/acscentsci.4c02060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 12/31/2024] [Accepted: 01/03/2025] [Indexed: 03/05/2025]
Abstract
The treatment of Gram-negative bacterial infections is challenged by antibiotic resistance and complicated forms of infection like persistence, multispecies biofilms, intracellular infection, as well as infection-associated hyperinflammation and sepsis. To overcome these challenges, a dual-functional antibiotic adjuvant has been developed as a novel strategy to target complicated forms of bacterial infection and exhibit immunomodulatory properties. The lead adjuvant, D-LBDiphe showed multimodal mechanisms of action like weak outer membrane permeabilization, weak membrane depolarization, and inhibition of efflux machinery, guided primarily by hydrogen bonding and electrostatic interactions, along with weak van der Waals forces. D-LBDiphe potentiated antibiotics up to ∼4100-fold, targeted phenotypic forms of antibiotic tolerance, and revitalized antibiotics against topical and systemic infections of P. aeruginosa in mice. The aromatic moiety in D-LBDiphe was instrumental for interaction with lipopolysaccharide (LPS) micelles, and this interaction was the driving factor in reducing pro-inflammatory cytokines by 61.8-79% in mice challenged with LPS. Such multifarious properties of a weak-membrane perturbing, nonactive and nontoxic adjuvant have been discussed for the first time, supported by detailed mechanistic understanding and elucidation of structure-guided properties. This work expands the scope of antibiotic adjuvants and validates them as a promising approach for treatment of complicated bacterial infections and inflammation.
Collapse
Affiliation(s)
- Geetika Dhanda
- Antimicrobial
Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru 560064, Karnataka, India
| | - Himani Singh
- Antimicrobial
Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru 560064, Karnataka, India
| | - Abhinav Gupta
- Department
of Chemistry, Indian Institute of Technology
Kanpur, Kanpur 208016, India
| | - Sk Abdul Mohid
- Department
of Chemical Sciences, Bose Institute, Kolkata 700091, India
| | - Karishma Biswas
- Department
of Chemical Sciences, Bose Institute, Kolkata 700091, India
| | - Riya Mukherjee
- Antimicrobial
Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru 560064, Karnataka, India
| | - Smriti Mukherjee
- Antimicrobial
Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru 560064, Karnataka, India
| | - Anirban Bhunia
- Department
of Chemical Sciences, Bose Institute, Kolkata 700091, India
| | - Nisanth N. Nair
- Department
of Chemistry, Indian Institute of Technology
Kanpur, Kanpur 208016, India
| | - Jayanta Haldar
- Antimicrobial
Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru 560064, Karnataka, India
- School
of Advanced Materials, Jawaharlal Nehru
Centre for Advanced Scientific Research, Jakkur, Bengaluru 560064, Karnataka, India
| |
Collapse
|
39
|
Ongwae GM, Liu Z, Feng S, Chordia MD, Sharifian Gh M, Dash R, Dalesandro BE, Guo T, Sharpless KB, Dong J, Siegrist MS, Im W, Pires MM. Click-Based Determination of Accumulation of Molecules in Escherichia coli. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.06.20.545103. [PMID: 40027664 PMCID: PMC11870406 DOI: 10.1101/2023.06.20.545103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Gram-negative bacterial pathogens pose a significant challenge in drug development due to their outer membranes, which impede the permeation of small molecules. The lack of widely adoptable methods to measure the cytosolic accumulation of compounds in bacterial cells has hindered drug discovery efforts. To address this challenge, we developed the CHloroalkane Azide Membrane Permeability (CHAMP) assay, specifically designed to assess molecule accumulation in the cytosol of Gram-negative bacteria. The CHAMP analysis utilizes biorthogonal epitopes anchored within HaloTag-expressing bacteria and measures the cytosolic arrival of azide-bearing test molecules through strain-promoted azide-alkyne cycloaddition. This workflow allows for robust and rapid accumulation measurements of thousands of azide-tagged small molecules. Our approach consistently yields a large number of accumulation profiles, significantly exceeding the scale of previous measurements in Escherichia coli ( E. coli ). We have validated the CHAMP assay across various chemical and biological contexts, including hyperporinated cells, membrane-permeabilized cells, and E. coli strains with impaired TolC function, a key component of the efflux pump. The CHAMP platform provides a simple, high-throughput, and accessible method that enables the analysis of over 1,000 molecules within hours. This technique addresses a critical gap in antimicrobial research, potentially accelerating the development of effective agents against Gram-negative pathogens.
Collapse
|
40
|
Xu J, Zou D, Dong E, Jiang X, Xu S, Xiao Y, Gong W, Xu Y, Zeng Z, Deng K, Li Y, Xu X. Biomedical Engineering on Smart Polymeric Nanoparticle-Hydrogel Platforms for Efficient Antibiotic Delivery against Bacterial-Infected Wounds. ACS Biomater Sci Eng 2025. [PMID: 39977696 DOI: 10.1021/acsbiomaterials.4c02045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
The rising incidence of bacterial infections poses a significant challenge to global public health. The development of safe and effective antibacterial treatment strategies is an urgent need in the field of biomedicine. In this work, we developed a smart nanoparticle-hydrogel platform to address bacterial infections in wounds. Rifampicin-loaded chitosan-functionalized nanoparticles (R-CNP) could break bacterial barriers and enhance antibiotic internalization. R-CNP reduced the minimum inhibitory concentration of rifampicin against Staphylococcus aureus and greatly enhanced the bactericidal effect of rifampicin. Furthermore, R-CNP was incorporated into thermosensitive hydrogels (HG) to construct HG(R-CNP) for enhanced antibiotic accumulation and wound protection. In the mouse model with a bacterial-infected wound, treatment with R-CNP reduced the bacterial content by 98.5% as compared to treatment with free rifampicin. Therefore, this smart nanoparticle-hydrogel platform constructed by FDA-approved or natural polymers, offers significant therapeutic efficacy on bacterial-infected wounds, showing great promise for clinical translation.
Collapse
Affiliation(s)
- Jiahao Xu
- Department of Pharmacy, College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Dongzhe Zou
- Department of Pharmacy, College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Enyu Dong
- Department of Pharmacy, College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Xingyu Jiang
- Department of Pharmacy, College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Shuo Xu
- Department of Pharmacy, College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Yusheng Xiao
- Department of Pharmacy, College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Wensheng Gong
- Department of Pharmacy, College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Yini Xu
- Department of Pharmacy, College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Zenan Zeng
- Department of Pharmacy, College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Kefurong Deng
- Department of Pharmacy, College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Yachao Li
- Department of Pharmacy, College of Biology, Hunan University, Changsha, Hunan 410082, China
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, Hunan 410082, China
| | - Xianghui Xu
- Department of Pharmacy, College of Biology, Hunan University, Changsha, Hunan 410082, China
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, Hunan 410082, China
| |
Collapse
|
41
|
Roson-Calero N, Gomis Font MA, Ruiz-Soriano A, Just-Baringo X, Pachón-Ibáñez ME, Salvador JP, Marco MP, Giralt E, Oliver A, Ballesté-Delpierre C, Vila J. In vitro potentiation of tetracyclines in Pseudomonas aeruginosa by RW01, a new cyclic peptide. Antimicrob Agents Chemother 2025; 69:e0145924. [PMID: 39714156 PMCID: PMC11823630 DOI: 10.1128/aac.01459-24] [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: 09/28/2024] [Accepted: 12/03/2024] [Indexed: 12/24/2024] Open
Abstract
The pipeline for new drugs against multidrug-resistant Pseudomonas aeruginosa remains limited, highlighting the urgent need for innovative treatments. New strategies, such as membrane-targeting molecules acting as adjuvants, aim to enhance antibiotic effectiveness and combat resistance. RW01, a cyclic peptide with low antimicrobial activity, was selected as an adjuvant to enhance drug efficacy through membrane permeabilization. RW01's activity was evaluated via antimicrobial susceptibility testing in combination with existing antibiotics on 10 P. aeruginosa strains and analog synthesis. Synergy was assessed using checkerboard assays, and one-step mutants were generated to identify altered pathways through whole-genome sequencing and variant analysis. Permeabilizing activity was studied using flow cytometry and real-time fluorescence measurement. In vivo toxicity was assessed in female C57BL/6J mice, and possible interaction with mouse serum was also evaluated. Susceptibility testing revealed specific synergy with tetracyclines, with up to a 16-fold reduction in minimum inhibitory concentrations. Sequencing revealed that resistance to the RW01-minocycline combination involved mutations in the pmrB gene, affecting outer membrane lipopolysaccharide composition. This was further confirmed by the identification of cross-resistance to colistin in these mutants. RW01 reduced the mutant prevention concentration of minocycline from 64 to 8 mg/L. RW01 was demonstrated to enhance membrane permeabilization and therefore minocycline uptake with statistical significance. Synthetic derivatives of RW01 showed a complete loss of activity, highlighting the importance of RW01's D-proline(NH2) residue. No acute or cumulative in vivo toxicity was observed in mice. These findings suggest that RW01 could revitalize obsolete antimicrobials and potentially expand therapeutic options against multidrug-resistant P. aeruginosa.
Collapse
Affiliation(s)
- Natalia Roson-Calero
- Barcelona Institute for Global Health (ISGlobal), Barcelona, Spain
- Department of Basic Clinical Practice, School of Medicine, University of Barcelona, Barcelona, Spain
| | - María A. Gomis Font
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Department of Microbiology, Hospital Universitario Son Espases, Health Research Institute of the Balearic Islands (IdISBa), Palma de Mallorca, Spain
| | - Albert Ruiz-Soriano
- Laboratori de Química Orgànica, Facultat de Farmàcia, Universitat de Barcelona, Barcelona, Spain
| | - Xavier Just-Baringo
- Laboratori de Química Orgànica, Facultat de Farmàcia, Universitat de Barcelona, Barcelona, Spain
| | - María Eugenia Pachón-Ibáñez
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Clinical Unit of Infectious Diseases, Microbiology and Parasitology, Institute of Biomedicine of Seville (IBiS), Virgen del Rocio University Hospital/CSIC/University of Seville, Seville, Spain
| | - J. Pablo Salvador
- Nanobiotechnology for Diagnostics (Nb4D), Department of Chemical and Biomolecular Nanotechnology, Institute for Advanced Architecture of Catalonia (IQAC) of the Spanish Council for Scientific Research (CSIC), Barcelona, Spain
- Centro de Investigación Biomédica en Red Bioingeniería, Biomateriales y Nanomedicina (CIBER_BBN), Madrid, Spain
| | - M. Pilar Marco
- Nanobiotechnology for Diagnostics (Nb4D), Department of Chemical and Biomolecular Nanotechnology, Institute for Advanced Architecture of Catalonia (IQAC) of the Spanish Council for Scientific Research (CSIC), Barcelona, Spain
- Centro de Investigación Biomédica en Red Bioingeniería, Biomateriales y Nanomedicina (CIBER_BBN), Madrid, Spain
| | - Ernest Giralt
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology, Barcelona, Spain
- Department of Inorganic and Organic Chemistry, University of Barcelona, Barcelona, Spain
| | - Antonio Oliver
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Department of Microbiology, Hospital Universitario Son Espases, Health Research Institute of the Balearic Islands (IdISBa), Palma de Mallorca, Spain
| | - Clara Ballesté-Delpierre
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Department of Clinical Microbiology, Biomedical Diagnostic Center, Hospital Clinic, Barcelona, Spain
| | - Jordi Vila
- Barcelona Institute for Global Health (ISGlobal), Barcelona, Spain
- Department of Basic Clinical Practice, School of Medicine, University of Barcelona, Barcelona, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Department of Clinical Microbiology, Biomedical Diagnostic Center, Hospital Clinic, Barcelona, Spain
| |
Collapse
|
42
|
Zhao W, Zhang B, Zheng S, Yan W, Yu X, Ye C. High temperatures promote antibiotic resistance genes conjugative transfer under residual chlorine: Mechanisms and risks. JOURNAL OF HAZARDOUS MATERIALS 2025; 483:136675. [PMID: 39603126 DOI: 10.1016/j.jhazmat.2024.136675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 11/16/2024] [Accepted: 11/25/2024] [Indexed: 11/29/2024]
Abstract
The impact of residual chlorine on the dissemination of antibiotic resistance during the distribution and storage of water has become a critical concern. However, the influence of rising temperatures attributed to global warming on this process remains ambiguous, warranting further investigation. This study investigated the effects of different temperatures (17, 27, 37, and 42°C) on the conjugative transfer of antibiotic resistance genes (ARGs) under residual chlorine (0, 0.1, 0.3, and 0.5 mg/L). The results indicated that high temperatures significantly increased the conjugative transfer frequency of ARGs in intra-species under residual chlorine. Compared to 17°C, the transfer frequencies at 27°C, 37°C, and 42°C increased by 1.07-2.43, 1.20-4.80, and 1.24-2.82 times, respectively. The promoting effect of high temperatures was mainly due to the generation of reactive oxygen species, the triggered SOS response, and the formation of pilus channels. Transcriptomic analysis demonstrated that higher temperature stimulates the electron transport chain, thereby enhancing ATP production and facilitating the processes of conjugative, as confirmed by inhibitor validation. Additionally, rising temperatures similarly promoted the frequency of conjugative transfer in inter-species and communities under residual chlorine. These further highlighted the risk of antibiotic resistance spread in extreme and prolonged high-temperature events. The increased risk of antibiotic resistance in the process of drinking water transmission under the background of climate warming is emphasized.
Collapse
Affiliation(s)
- Wenya Zhao
- College of the Environment & Ecology, Xiamen University, Xiamen 361102, China; Fujian Key Laboratory of Coastal Pollution Prevention and Control, Xiamen 361102, China
| | - Binghuang Zhang
- College of the Energy, Xiamen University, Xiamen 361102, China
| | - Shikan Zheng
- College of the Environment & Ecology, Xiamen University, Xiamen 361102, China; Fujian Key Laboratory of Coastal Pollution Prevention and Control, Xiamen 361102, China
| | - Wanli Yan
- College of the Environment & Ecology, Xiamen University, Xiamen 361102, China; Fujian Key Laboratory of Coastal Pollution Prevention and Control, Xiamen 361102, China
| | - Xin Yu
- College of the Environment & Ecology, Xiamen University, Xiamen 361102, China; Fujian Key Laboratory of Coastal Pollution Prevention and Control, Xiamen 361102, China
| | - Chengsong Ye
- College of the Environment & Ecology, Xiamen University, Xiamen 361102, China; Fujian Key Laboratory of Coastal Pollution Prevention and Control, Xiamen 361102, China.
| |
Collapse
|
43
|
Wu W, Guo Z, Zhang J, Li W, Liu C, Jiang B, Su Y. Integration of transcriptomics and metabolomics reveals the mechanism of enrofloxacin resistance in Aeromonas schubertii. Microb Pathog 2025; 199:107262. [PMID: 39730098 DOI: 10.1016/j.micpath.2024.107262] [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: 07/25/2024] [Revised: 11/18/2024] [Accepted: 12/24/2024] [Indexed: 12/29/2024]
Abstract
Aeromonas schubertii infections has caused severe economic losses in aquaculture in China. In this study, we first induced enrofloxacin (ENR) resistance in A. schubertii strains and then analyzed the mechanisms of drug resistance using transcriptomics and metabolomics. We found that the minimal inhibitory concentration (MIC) was 0.03125 μg/mL for the sensitive strain (WL23S) and 32 μg/mL for the resistant strain (WL23R), which is a 1024-fold increase. After 40 serial passages, the WL23R strain maintained a MIC of 32 μg/mL, even in the absence of ENR-induced stress. Notably, it had also developed resistance to several other antibiotics, such as neomycin sulfate and flumequine. There was no significant difference in the growth rates of the two strains, highlighting the strong adaptability and growth characteristics of the WL23R strain. Comparison of the transcriptome data between the WL23R and WL23S strains identified 579 differentially expressed genes. Expression of the efflux pump-related genes (e.g., acrA, acrB, pstB, pstC, pstS) was significantly upregulated in the WL23R strain (P < 0.05). The highest enrichment of differential genes in the Gene Ontology analysis was in the catabolism of various amino acids, and that in the Kyoto Encyclopedia of Genes and Genomes pathway was in ATP-binding cassette (ABC) transport. Comparison of the metabolomics data between the WL23R and WL23S strains revealed 1, 059 differentially expressed metabolites. Metabolomics analysis revealed the impact of drug resistance on the levels of amino acids, the activity of amino acid biosynthesis/metabolism pathways, and the ABC transport protein pathway, which confirmed the transcriptomics results. The joint analysis results showed that ABC transporters were most prominent in the shared pathways between enriched differentially expressed genes and metabolites. To further validate the resistance mechanism of A. schubertii, we exposed the WL23R strain to the efflux pump inhibitor carbonyl cyanide 3-chlorophenylhydrazone. The minimal inhibitory concentration of the induced resistant strain decreased by 4-fold after the addition of the inhibitor, indicating the overexpression of active efflux pumps in WL23R. Our results indicate that the efflux system and ABC transporters play crucial roles during the development of multidrug resistance in A. schubertii. This study will serve as an important reference for understanding bacterial resistance to quinolones and multidrug resistance in aquatic environments.
Collapse
Affiliation(s)
- Wenhao Wu
- Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, 510225, Guangdong, China
| | - Zihong Guo
- Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, 510225, Guangdong, China
| | - Jiahao Zhang
- Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, 510225, Guangdong, China
| | - Wei Li
- Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, 510225, Guangdong, China
| | - Chun Liu
- Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, 510225, Guangdong, China
| | - Biao Jiang
- Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, 510225, Guangdong, China.
| | - Youlu Su
- Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, 510225, Guangdong, China.
| |
Collapse
|
44
|
Wang M, Li W, Li S, Xiang J, Shao Y, Yin C, Sedjoah RCAA, Xin Z. Functional characterization and mechanism of the multidrug resistance transport potein YoeA in Bacillus subtilis. Int J Biol Macromol 2025; 291:139115. [PMID: 39719240 DOI: 10.1016/j.ijbiomac.2024.139115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 12/19/2024] [Accepted: 12/21/2024] [Indexed: 12/26/2024]
Abstract
Transport proteins are essential for bacterial resistance to antibiotics and toxins, but their mechanisms remain poorly understood in Bacillus subtilis. In the present study, overexpression of yoeA enhanced resistance to various antibiotics, with its expression induced by these antibiotics, especially penicillin and plipastatin. The ΔyoeA strain exhibited significant growth inhibition at 100 μg/mL of plipastatin, while as high as 10,000 μg/mL of iturin/surfactin are required to achieve comparable inhibition, suggesting a higher sensitivity of ΔyoeA to plipastatin. The transcript level of yoeA gene was increased 2.71-fold in response to plipastatin, significantly higher than the levels induced by surfactin and iturin. The ethidium bromide (EtBr) efflux activity of YoeA was inhibited by carbonyl cyanide chlorophenylhydrazone (CCCP) and enhanced by Na+. Molecular modeling studies revealed that cation-π interactions of Na+ with Y287 and Y434 residues in the C-terminal domain of YoeA contribute to its ion channel function, and Cu2+ can form coordination bonds with the N atoms of H278 and H421 residues on the C-terminal surface of YoeA, promoting plipastatin efflux in a dose-dependent manner. The present study characterized the main factors influencing YoeA's efflux activity, revealed its transport mechanism, and provided new insights into enhancing antimicrobial peptide production and controlling bacterial resistance.
Collapse
Affiliation(s)
- Mengxi Wang
- Key Laboratory of Food Processing and Quality Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210000, PR China
| | - Wenqing Li
- Key Laboratory of Food Processing and Quality Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210000, PR China
| | - Siwei Li
- Key Laboratory of Food Processing and Quality Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210000, PR China
| | - Jiahui Xiang
- Key Laboratory of Food Processing and Quality Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210000, PR China
| | - Yuting Shao
- Key Laboratory of Food Processing and Quality Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210000, PR China
| | - ChenYue Yin
- Key Laboratory of Food Processing and Quality Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210000, PR China
| | - Rita-Cindy Aye-Ayire Sedjoah
- Key Laboratory of Food Processing and Quality Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210000, PR China
| | - Zhihong Xin
- Key Laboratory of Food Processing and Quality Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210000, PR China.
| |
Collapse
|
45
|
Domen A, Porter J, Johnson J, Molyneux J, McIntyre L, Kovacevic J, Waite-Cusic J. Variability in cadmium tolerance of closely related Listeria monocytogenes isolates originating from dairy processing environments. Appl Environ Microbiol 2025; 91:e0128124. [PMID: 39570037 PMCID: PMC11784300 DOI: 10.1128/aem.01281-24] [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: 06/29/2024] [Accepted: 10/18/2024] [Indexed: 11/22/2024] Open
Abstract
Increased tolerance to cadmium in Listeria monocytogenes has been suggested to contribute to their persistence in natural and food production environments. This study investigated the phenotypic cadmium response of L. monocytogenes strains with efflux pump cadAC (variants 1-4) and related strains with cadA1C1. Growth of cadAC variant strains (n = 5) in 0 µM-120 µM cadmium salts (CdCl2, CdSO4) in Mueller-Hinton broth (MHB) was evaluated. Additionally, 88 L. monocytogenes strains from dairy processing facilities were exposed to 43.8 µM CdCl2 in MHB, and their lag phase duration (LPD) was measured. Strains with cadA1 through cadA3 showed similar growth trends in the presence of cadmium, while the cadA4 variant (Scott A) had the highest CdCl2 minimum inhibitory concentration (175 µM). Growth varied between the two salts, with CdSO4 significantly increasing LPD (P < 0.05) compared to CdCl2. In 43.8 µM CdCl2, cadA1 strains displayed LPDs ranging from 0.99 ± 0.14 h to 6.44 ± 0.08 h, with no clear genomic differences explaining this variability. Strains without cadA did not grow at 43.8 µM CdCl2 but exhibited low tolerance (10.9 µM CdCl2), potentially due to non-specific soft metal ATPases (626 aa; 737 aa) and soft metal resistance proteins encoded by czc genes (289 aa; 291 aa; 303 aa) within their chromosomes. These findings enhance our understanding of L. monocytogenes cadmium tolerance and underscore the need for further research to explore the genetic and physiological factors underlying these trends. IMPORTANCE Mobile genetic elements in Listeria monocytogenes contribute to its survival in natural and food processing environments. This study focused on how different genetic variants of the efflux pump gene cadAC and group of closely related cadA1C1 strains respond to cadmium exposure. When exposed to two cadmium salts, cadmium chloride and cadmium sulfate, we observed varying growth patterns, with a significantly longer lag phase in cadmium sulfate compared to cadmium chloride. Strains with cadA1 to cadA3 had similar growth trends, whereas a strain with the cadA4 variant had the highest minimum inhibitory concentration value. Among 88 strains from dairy processing facilities, significant phenotypic differences were observed despite core genome similarities, indicating other underlying genetic and physiological factors contribute to cadmium tolerance. Since cadmium tolerance studies in L. monocytogenes are limited, with rare phenotypic comparisons between closely related strains, our study makes an important observation and contribution to understanding of L. monocytogenes tolerance to cadmium by providing phenotypic comparisons between numerous strains within the same clonal group (<16 single nucleotide polymorphisms).
Collapse
Affiliation(s)
- Andrea Domen
- Food Innovation Center, Oregon State University, Portland, Oregon, USA
- Department of Food Science and Technology, Oregon State University, Corvallis, Oregon, USA
| | - Jenna Porter
- Department of Food Science and Technology, Oregon State University, Corvallis, Oregon, USA
| | - Jared Johnson
- Department of Food Science and Technology, Oregon State University, Corvallis, Oregon, USA
| | - James Molyneux
- Department of Statistics, Oregon State University, Corvallis, Oregon, USA
| | | | - Jovana Kovacevic
- Food Innovation Center, Oregon State University, Portland, Oregon, USA
- Department of Food Science and Technology, Oregon State University, Corvallis, Oregon, USA
| | - Joy Waite-Cusic
- Department of Food Science and Technology, Oregon State University, Corvallis, Oregon, USA
| |
Collapse
|
46
|
Haripriyan J, Binu CR, Menon ND, Vanuopadath M, Hari MB, Namitha N, Binoy K, Kumar A, Nair BG, Nizet V, Kumar GB. Essential oils modulate virulence phenotypes in a multidrug-resistant pyomelanogenic Pseudomonas aeruginosa clinical isolate. Sci Rep 2025; 15:3738. [PMID: 39885214 PMCID: PMC11782693 DOI: 10.1038/s41598-025-86515-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 01/13/2025] [Indexed: 02/01/2025] Open
Abstract
Pyomelanogenic P. aeruginosa, frequently isolated from patients with urinary tract infections and cystic fibrosis, possesses the ability to withstand oxidative stress, contributing to virulence and resulting in persistent infections. Whole genome sequence analysis of U804, a pyomelanogenic, multidrug-resistant, clinical isolate, demonstrates the mechanism underlying pyomelanin overproduction. Seven essential oils (EOs) were screened for pyomelanin inhibition. Garlic, cinnamon and thyme EOs were selected for further studies based on their significant anti-virulent properties, like inhibition of pyomelanin production and biofilm formation. Additionally, downregulation of the expression of virulence genes regulated by quorum sensing (QS) and a decrease in levels of the QS signaling molecule, C12-HSL, were also observed. The EO treatment inhibited the survival of U804 in human blood and increased survival of C. elegans, a whole animal model of pathogenesis. EO treatment also resulted in a significant reduction of efflux pump activity, indicative of their effect on antibiotic sensitization. Garlic oil enhanced the permeability of the bacterial membrane, resulting in decreased survival, when combined with sub-MIC concentrations of colistin. This study demonstrates that thyme, cinnamon and garlic EOs can attenuate pyomelanogenic P. aeruginosa virulence traits. Additionally, garlic potentiates drug sensitivity, suggesting its promising therapeutic use in combating pyomelanogenic MDR infections.
Collapse
Affiliation(s)
| | - Christy Rose Binu
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Amritapuri, Kerala, India
| | - Nitasha D Menon
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Amritapuri, Kerala, India
| | | | - Malavika B Hari
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Amritapuri, Kerala, India
| | - N Namitha
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Amritapuri, Kerala, India
| | - Kesiya Binoy
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Amritapuri, Kerala, India
| | - Anil Kumar
- Department of Microbiology, Amrita Institute of Medical Sciences, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Bipin G Nair
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Amritapuri, Kerala, India
| | - Victor Nizet
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
| | - Geetha B Kumar
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Amritapuri, Kerala, India.
- Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, Clappana, Kollam, Kerala, 690525, India.
| |
Collapse
|
47
|
Wimalasekara RL, White D, Kumar A. Targeting Acinetobacter baumannii resistance-nodulation-division efflux pump transcriptional regulators to combat antimicrobial resistance. NPJ ANTIMICROBIALS AND RESISTANCE 2025; 3:4. [PMID: 39863717 PMCID: PMC11762787 DOI: 10.1038/s44259-024-00074-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 12/17/2024] [Indexed: 01/27/2025]
Abstract
Regulatory elements controlling gene expression fine-tune bacterial responses to environmental cues, including antimicrobials, to optimize survival. Acinetobacter baumannii, a pathogen notorious for antimicrobial resistance, relies on efficient efflux systems. Though the role of efflux systems in antibiotic expulsion are well recognized, the regulatory mechanisms controlling their expression remain understudied. This review explores the current understanding of these regulators, aiming to inspire strategies to combat bacterial resistance and improve therapeutic outcomes.
Collapse
Affiliation(s)
| | - Dawn White
- Department of Microbiology, University of Manitoba, Winnipeg, MB, Canada
| | - Ayush Kumar
- Department of Microbiology, University of Manitoba, Winnipeg, MB, Canada.
| |
Collapse
|
48
|
Wright GD. The Janus Effect: The Biochemical Logic of Antibiotic Resistance. Biochemistry 2025; 64:301-311. [PMID: 39772429 DOI: 10.1021/acs.biochem.4c00585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Antibiotics are essential medicines threatened by the emergence of resistance in all relevant bacterial pathogens. The engagement of the molecular targets of antibiotics offers multiple opportunities for resistance to emerge. Successful target engagement often requires passage of the antibiotic from outside into the cell interior through one or two distinct membrane barriers. Resistance can occur by preventing the accumulation of antibiotics in sufficient quantities outside the cell, decreasing the rates of entry into the cell, and modifying the antibiotic or the target once inside the cell. These competing equilibria and rates are the lens through which the balance of antibiotic efficacy or failure can be viewed. The two faces of antibiotics, cell growth inhibition or resistance, are reminiscent of Janus, the Roman god of doorways and beginnings and endings, and offer a framework through which antibiotic discovery, use, and the emergence of resistance can be rationally viewed.
Collapse
Affiliation(s)
- Gerard D Wright
- David Braley Centre for Antibiotic Discovery, M.G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4K1, Canada
| |
Collapse
|
49
|
Pan M, Colpo RA, Roussou S, Ding C, Lindblad P, Krömer JO. Engineering a Photoautotrophic Microbial Coculture toward Enhanced Biohydrogen Production. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2025; 59:337-348. [PMID: 39668362 PMCID: PMC11741097 DOI: 10.1021/acs.est.4c08629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 12/05/2024] [Accepted: 12/05/2024] [Indexed: 12/14/2024]
Abstract
The application of synthetic phototrophic microbial consortia holds promise for sustainable bioenergy production. Nevertheless, strategies for the efficient construction and regulation of such consortia remain challenging. Applying tools of genetic engineering, this study successfully constructed a synthetic community of phototrophs using Rhodopseudomonas palustris (R. palustris) and an engineered strain of Synechocystis sp PCC6803 for acetate production (Synechocystis_acs), enabling the production of biohydrogen and fatty acids during nitrogen and carbon dioxide fixation. Elemental balance confirmed carbon capture and nitrogen fixation into the consortium. The strategy of circadian illumination effectively limited oxygen levels in the system, ensuring the activity of the nitrogenase in R. palustris, despite oxygenic photosynthesis happening in Synechocystis. When infrared light was introduced into the circadian illumination, the production of H2 (9.70 μmol mg-1) and fatty acids (especially C16 and C18) was significantly enhanced. Proteomic analysis indicated acetate exchange and light-dependent regulation of metabolic activities. Infrared illumination significantly stimulated the expression of proteins coding for nitrogen fixation, carbohydrate metabolism, and transporters in R. palustris, while constant white light led to the most upregulation of photosynthesis-related proteins in Synechocystis_acs. This study demonstrated the successful construction and light regulation of a phototrophic community, enabling H2 and fatty acid production through carbon and nitrogen fixation.
Collapse
Affiliation(s)
- Minmin Pan
- Department
of Microbial Biotechnology, Helmholtz Centre
for Environmental Research - UFZ, Leipzig 04318, Germany
| | - Rodrigo Amarante Colpo
- Department
of Microbial Biotechnology, Helmholtz Centre
for Environmental Research - UFZ, Leipzig 04318, Germany
| | - Stamatina Roussou
- Microbial
Chemistry, Department of Chemistry-Ångström, Uppsala University, Box 523, Uppsala 75120, Sweden
| | - Chang Ding
- Department
of Molecular Environmental Biotechnology, Helmholtz Centre for Environmental Research - UFZ, Leipzig 04318, Germany
| | - Peter Lindblad
- Microbial
Chemistry, Department of Chemistry-Ångström, Uppsala University, Box 523, Uppsala 75120, Sweden
| | - Jens O. Krömer
- Department
of Microbial Biotechnology, Helmholtz Centre
for Environmental Research - UFZ, Leipzig 04318, Germany
| |
Collapse
|
50
|
Liu Z, Yao X, Chen C, Zhao Y, Dong C, Sun L, Zhao J, Zhang B, Yu Z, Cheng D, Zhu L, Hu B. Growth of microbes in competitive lifestyles promotes increased ARGs in soil microbiota: insights based on genetic traits. MICROBIOME 2025; 13:8. [PMID: 39806455 PMCID: PMC11730135 DOI: 10.1186/s40168-024-02005-6] [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: 06/02/2024] [Accepted: 12/12/2024] [Indexed: 01/16/2025]
Abstract
BACKGROUND The widespread selective pressure of antibiotics in the environment has led to the propagation of antibiotic resistance genes (ARGs). However, the mechanisms by which microbes balance population growth with the enrichment of ARGs remain poorly understood. To address this, we employed microcosm cultivation at different antibiotic (i.e., Oxytetracycline, OTC) stresses across the concentrations from the environmental to the clinical. Paired with shot-gun metagenomics analysis and quantification of bacterial growth, trait-based assessment of soil microbiota was applied to reveal the association between key ARG subtypes, representative bacterial taxa, and functional-gene features that drive the growth of ARGs. RESULTS Our results illuminate that resistome variation is closely associated with bacterial growth. A non-monotonic change in ARG abundance and richness was observed over a concentration gradient from none to 10 mg/l. Soil microbiota exposed to intermediate OTC concentrations (i.e., 0.1 and 0.5 mg/l) showed greater increases in the total abundance of ARGs. Community compositionally, the growth of representative taxa, i.e., Pseudomonadaceae was considered to boost the increase of ARGs. It has chromosomally carried kinds of multidrug resistance genes such as mexAB-oprM and mexCD-oprJ could mediate the intrinsic resistance to OTC. Streptomycetaceae has shown a better adaptive ability than other microbes at the clinical OTC concentrations. However, it contributed less to the ARGs growth as it represents a stress-tolerant lifestyle that grows slowly and carries fewer ARGs. In terms of community genetic features, the community aggregated traits analysis further indicates the enhancement in traits of resource acquisition and growth yield is driving the increase of ARGs abundance. Moreover, optimizations in energy production and conversion, alongside a streamlining of bypass metabolic pathways, further boost the growth of ARGs in sub-inhibitory antibiotic conditions. CONCLUSION The results of this study suggest that microbes with competitive lifestyles are selected under the stress of environmental sub-inhibitory concentrations of antibiotics and nutrient scarcity. They possess greater substrate utilization capacity and carry more ARGs, due to this they were faster growing and leading to a greater increase in the abundance of ARGs. This study has expanded the application of trait-based assessments in understanding the ecology of ARGs propagation. And the finding illustrated changes in soil resistome are accompanied by the lifestyle switching of the microbiome, which theoretically supports the ARGs control approach based on the principle of species competitive exclusion. Video Abstract.
Collapse
Affiliation(s)
- Zishu Liu
- Key Laboratory of Environment Remediation and Ecological Health, Ministry of Education, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, 310058, China
- College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, 310058, China
- Key Laboratory of Water Pollution Control and Environmental Safety of Zhejiang Province, Hangzhou, 310058, China
| | - Xiangwu Yao
- College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Chengyi Chen
- College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yuxiang Zhao
- College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Chifei Dong
- College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Lingtao Sun
- College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Junxian Zhao
- College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Baofeng Zhang
- Hangzhou Ecological and Environmental Monitoring Center, Hangzhou, 310007, China
| | - Zhendi Yu
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Dongqing Cheng
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Lizhong Zhu
- Key Laboratory of Environment Remediation and Ecological Health, Ministry of Education, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, 310058, China
- College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Baolan Hu
- Key Laboratory of Environment Remediation and Ecological Health, Ministry of Education, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, 310058, China.
- College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, 310058, China.
- Key Laboratory of Water Pollution Control and Environmental Safety of Zhejiang Province, Hangzhou, 310058, China.
| |
Collapse
|