1
|
Yao KS, Zhou PL, Tan LJ, Yang HQ, Qiao LK, Gao FZ, Liu YS, Peng FJ, Ying GG, Van den Brink PJ. Antibiotic-induced multi-trophic effects and their cascades in a sub-tropical freshwater ecosystem. JOURNAL OF HAZARDOUS MATERIALS 2025; 492:138281. [PMID: 40245710 DOI: 10.1016/j.jhazmat.2025.138281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 04/02/2025] [Accepted: 04/12/2025] [Indexed: 04/19/2025]
Abstract
Antibiotics are commonly detected in aquatic ecosystems worldwide due to their extensive use and excretion by humans and animals, posing potential risks to the health of these ecosystems. This study aimed to assess the ecological effects of the antibiotic ciprofloxacin on both structural (microbes, phytoplankton, zooplankton, and macroinvertebrates) and functional endpoints of a sub-tropical freshwater ecosystem. Ciprofloxacin was applied at concentrations of 0, 0.5, 5, 50, and 500 µg/L for 21 consecutive days in outdoor mesocosms, followed by a five-week recovery period. Ciprofloxacin significantly affected the structure of microbial, phytoplankton, and zooplankton communities, with calculated NOECs of 5, 0.5, and 5 µg/L, respectively. Notably, the microbial community composition, particularly taxa within the phyla Cyanobacteria and Bacteroidetes, exhibited marked shifts. Among phytoplankton, the filamentous cyanobacterium Cylindrospermopsis sp. exhibited the largest negative response to ciprofloxacin, while Microcystis sp. displayed the largest increase in abundance. Ciprofloxacin exposure also indirectly led to significant increases in zooplankton populations belonging to Cladocera, Copepoda, and Rotifera. Significant effects of ciprofloxacin on physicochemical parameters related to carbon and nitrogen cycling were observed. Structural equation models revealed that ciprofloxacin induced both direct and indirect effects across multiple trophic levels through cascading effects, further impacting ecosystem-level endpoints. Overall, this study provides an understanding of the potential ecological risks posed by antibiotic pollution on freshwater ecosystems.
Collapse
Affiliation(s)
- Kai-Sheng Yao
- Aquatic Ecology and Water Quality Management Group, Wageningen University, P.O. Box 47, Wageningen 6700 AA, the Netherlands; SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, South China Normal University, Guangzhou 510006, China
| | - Pei-Liang Zhou
- SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, South China Normal University, Guangzhou 510006, China; School of Environment, South China Normal University, Guangzhou 510006, China
| | - Li-Juan Tan
- SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, South China Normal University, Guangzhou 510006, China; School of Environment, South China Normal University, Guangzhou 510006, China
| | - Hua-Qiao Yang
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Lu-Kai Qiao
- SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, South China Normal University, Guangzhou 510006, China; School of Environment, South China Normal University, Guangzhou 510006, China
| | - Fang-Zhou Gao
- SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, South China Normal University, Guangzhou 510006, China; School of Environment, South China Normal University, Guangzhou 510006, China
| | - You-Sheng Liu
- SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, South China Normal University, Guangzhou 510006, China; School of Environment, South China Normal University, Guangzhou 510006, China
| | - Feng-Jiao Peng
- SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, South China Normal University, Guangzhou 510006, China; School of Environment, South China Normal University, Guangzhou 510006, China
| | - Guang-Guo Ying
- SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, South China Normal University, Guangzhou 510006, China; School of Environment, South China Normal University, Guangzhou 510006, China
| | - Paul J Van den Brink
- Aquatic Ecology and Water Quality Management Group, Wageningen University, P.O. Box 47, Wageningen 6700 AA, the Netherlands.
| |
Collapse
|
2
|
Wang H, Wu X, Xu J, Lu Z, Hu B, Zhu L, Lu H. Proline mitigates antibiotic resistance evolution induced by ciprofloxacin at environmental concentrations. JOURNAL OF HAZARDOUS MATERIALS 2025; 489:137561. [PMID: 39938368 DOI: 10.1016/j.jhazmat.2025.137561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/21/2025] [Accepted: 02/09/2025] [Indexed: 02/14/2025]
Abstract
Antibiotics-induced resistance development in the environment has emerged as a critical issue under the 'one health' framework. Although there have been approaches to control antibiotic resistance evolution in clinical settings, they are rarely applicable in environmental contexts. Amino acids can affect the metabolic states of bacteria and potentially influence their resistance evolution. In this study, we screened 18 amino acids and identified proline as an efficient agent capable of mitigating ciprofloxacin-induced resistance of a soil-isolated Escherichia coli by over 50 % during a 24-day evolutionary experiment. Using transcriptomics and 13C metabolic flux analysis, we revealed the evolution mitigation mechanism of proline, which mainly involves down-regulated gene expressions and reduced metabolic flux of the TCA cycle, thereby decreasing NADH production, proton motive force, and uptake of ciprofloxacin. Based on single-cell RNA-seq, proline also reduced the size of resistant subgroups in the evolved E. coli population. Based on soil microcosm experiments, proline not only reduced the overall antibiotic resistance but also increased community diversity and robustness (optimal dosage: 5 mg/kg). Moreover, proline's evolution mitigation potentials likely extend to other antibiotics (e.g., streptomycin) and populations (e.g., Pseudomonas and Serratia spp.). Overall, proline addition holds promising potentials for mitigating antibiotic resistance in diverse antibiotics-polluted environments.
Collapse
Affiliation(s)
- Hanqing Wang
- Key Laboratory of Environment Remediation and Ecological Health, Ministry of Education, College of Environmental Resource Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Xiujing Wu
- Key Laboratory of Environment Remediation and Ecological Health, Ministry of Education, College of Environmental Resource Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Jing Xu
- Key Laboratory of Environment Remediation and Ecological Health, Ministry of Education, College of Environmental Resource Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Zhenmei Lu
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Baolan Hu
- Key Laboratory of Environment Remediation and Ecological Health, Ministry of Education, College of Environmental Resource Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China; Zhejiang Province Key Laboratory for Water Pollution Control and Environmental Safety, Zhejiang University, Hangzhou 310058, China
| | - Lizhong Zhu
- Zhejiang Provincial Key Laboratory of Organic Pollution Process and Control, Zhejiang University, Hangzhou 310058, China
| | - Huijie Lu
- Key Laboratory of Environment Remediation and Ecological Health, Ministry of Education, College of Environmental Resource Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China; Zhejiang Province Key Laboratory for Water Pollution Control and Environmental Safety, Zhejiang University, Hangzhou 310058, China; Academy of Ecological Civilization, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
3
|
Manik MRK, Mishu ID, Mahmud Z, Muskan MN, Emon SZ. Association of fluoroquinolone resistance with rare quinolone resistance-determining region (QRDR) mutations and protein-quinolone binding affinity (PQBA) in multidrug-resistant Escherichia coli isolated from patients with urinary tract infection. J Infect Public Health 2025; 18:102766. [PMID: 40153979 DOI: 10.1016/j.jiph.2025.102766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND Urinary tract infections (UTIs) caused by Escherichia coli pose significant public health risks, particularly in developing countries like Bangladesh. This study aimed to elucidate resistance patterns among UTI isolates and comprehensively investigate the mutational spectrum and its impact on drug-microbe interactions. METHODS We collected and identified E. coli isolates from hospitalized UTI patients at Dhaka Medical College Hospital and determined their resistance patterns using the disc diffusion method and broth microdilution. Quinolone resistance-determining regions (QRDRs) of the target genes (gyrA, gyrB, parC, and parE) associated with fluoroquinolone resistance were amplified by polymerase chain reaction (PCR) and analyzed through BTSeq™ sequencing for mutations, followed by molecular docking analysis using PyMOL and AutoDock for the protein-quinolone binding affinity (PQBA) study. RESULTS All isolates (100 %) displayed multidrug resistance, with chloramphenicol (16 % resistant) and colistin (28 % resistant) demonstrating superior efficacy compared to other antibiotics. The isolates resistant to colistin, as determined by disc diffusion testing, exhibited remarkably high minimum inhibitory concentrations (MICs), with one isolate registering an MIC exceeding 512 µg/mL. Alarming resistance rates were observed for five antibiotic classes, except for polymyxins (28 % resistant) and protein synthesis inhibitors (48 % resistant). Fifty-two percent (52 %) of the isolates exhibited resistance to all five tested quinolones. Sequence analysis revealed a novel L88Q mutation in ParC, affecting PQBA and binding conformation. Additionally, three ParC mutations (S80I, E84V, and E84G) and two ParE mutations (S458A and I529L) were identified, which had not been previously reported in Bangladesh. Among these, S80I appeared in all isolates. Double-mutations (S83L+D87N) in GyrA, L88Q and S80I in ParC, and I529L in ParE were identified as key drivers of fluoroquinolone resistance. CONCLUSION Our findings underscore the accumulation of significant mutations within QRDRs of UTI isolates, potentially compromising fluoroquinolone efficacy. The emergence of these novel mutations warrants further investigation to impede their dissemination and combat quinolone resistance.
Collapse
Affiliation(s)
- Md Rasel Khan Manik
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka 1000, Bangladesh
| | | | - Zimam Mahmud
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka 1000, Bangladesh.
| | | | - Sharmin Zaman Emon
- Centre for Advanced Research in Sciences, University of Dhaka, Dhaka 1000, Bangladesh
| |
Collapse
|
4
|
Yehya A, Ezzeddine Z, Chakkour M, Dhaini Z, Bou Saba MS, Bou Saba AS, Nohra L, Nassar NB, Yassine M, Bahmad HF, Ghssein G. The intricacies of Acinetobacter baumannii: a multifaceted comprehensive review of a multidrug-resistant pathogen and its clinical significance and implications. Front Microbiol 2025; 16:1565965. [PMID: 40444001 PMCID: PMC12121509 DOI: 10.3389/fmicb.2025.1565965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 04/07/2025] [Indexed: 06/02/2025] Open
Abstract
Acinetobacter baumannii, a highly adaptive and formidable nosocomial pathogen, has emerged as a symbol of modern medicine's struggle against multidrug resistance (MDR). As a Gram-negative dweller in moist hospital environments, A. baumannii has proven its ability to colonize the most vulnerable-critically ill patients-leaving behind a trail of infections highlighted by high morbidity and mortality and rendering nearly all antibiotics ineffective. This literature review aims to provide an in-depth, comprehensive overview of microbiological features, virulence factors, clinical manifestations, epidemiology, and antibiotic resistance mechanisms of A. baumannii. It also highlights the different diagnostic approaches, possible treatment strategies, and infection control, as well as the profound public health burden this pathogen imposes. The genus Acinetobacter has undergone a pivotal taxonomic journey and categorization. In addition, the intricate virulence mechanisms and factors of A. baumannii, including but not limited to outer membrane components and nutrient acquisition systems, have contributed to its pathogenicity and severe clinical manifestations ranging from respiratory tract infections and meningitis to urinary tract infections, skin infections, and bloodstream infections. This review also describes the epidemiological trend of A. baumannii established by its global prevalence and distribution, risk factors, hospital-acquired vs. community-acquired infections, and its geographical variations. In terms of antibiotic resistance, this pathogen has demonstrated resilience to a wide range of first-line and last-resort antibiotics due to its different evasion mechanisms. The current diagnostic approaches, treatment strategies, and infection control measures are further analyzed in detail, underscoring the need for prompt and precise identification of A. baumannii to guide appropriate therapy and reinforce the optimal approaches to limit its transmission and control outbreaks. Finally, the review addresses the substantial public health implications, reflecting on the hindrance that A. baumannii brings to healthcare systems, and the urgent need for global surveillance, effective infection control protocols, innovative research, and therapeutic approaches to mitigate its global threat.
Collapse
Affiliation(s)
- Amani Yehya
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Zeinab Ezzeddine
- High Council for Scientific Research and Publication (HCSRP), Islamic University of Lebanon (IUL), Khalde, Lebanon
| | - Mohamed Chakkour
- Department of Biological Sciences, Wayne State University, Detroit, MI, United States
| | - Zahraa Dhaini
- Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | | | | | - Lea Nohra
- Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Nagham B. Nassar
- Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Mahdi Yassine
- Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Hisham F. Bahmad
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Ghassan Ghssein
- High Council for Scientific Research and Publication (HCSRP), Islamic University of Lebanon (IUL), Khalde, Lebanon
| |
Collapse
|
5
|
Chen J, Kuang R, Liu H, Yang L, Li J, Li J, Xia J, Huang Y, Cui M, Zou L, Han X. Emergence of multidrug resistant Escherichia coli coharboring fosA3 and ESBL genes from retail ducks along slaughter line. Int J Food Microbiol 2025; 435:111185. [PMID: 40179607 DOI: 10.1016/j.ijfoodmicro.2025.111185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 03/10/2025] [Accepted: 03/28/2025] [Indexed: 04/05/2025]
Abstract
The excessive and indiscriminate use of antimicrobial agents in poultry production has increased antimicrobial resistance in E. coli, posing a significant threat to public health. This study investigated the prevalence of multidrug-resistant (MDR) E. coli co-harboring fosA3 and extended-spectrum β-lactamases (ESBLs) genes in large-scale retail duck slaughterhouses in Sichuan Province, China. The antimicrobial susceptibility to 9 categories and 16 types of antimicrobial agents was assessed by using the broth microdilution method. Phylogenetic grouping and pulsed-field gel electrophoresis (PFGE) were used to investigate the phylogenetic groups and genetic characteristics of the isolates. Moreover, whole-genome sequencing (WGS) was performed on 35 representative E. coli isolates. Among 1, 059 samples collected, 895 E. coli were isolated by MacConkey (MAC) agar and eosin methylene blue (EMB) agar and PCR identification, and 150 strains of MDR E. coli co-carrying fosA3 and ESBL genes were finally screened for further analysis. The results indicated that 141 isolates were resistant to fosfomycin and exhibited high resistance to β-lactam antibiotics. All isolates demonstrated MDR patterns, with 148 isolates resistant to six or more classes of antimicrobial agents. The majority (70/150, 46.67 %) belonged to phylogenetic group B1. Based on an 85 % similarity threshold, all isolates were categorized into 40 distinct PFGE types. Genomic analysis revealed 27 different serotypes and 19 sequence types (STs), with O8 (14.29 %, 5/35) and ST155 (22.86 %, 8/35) being the most common. The IncFIB (AP001918) was the most prevalent plasmid replicon type, identified in 74.29 % (26/35) of the isolates. In addition, 49 acquired antimicrobial resistance genes (ARGs) associated with resistance to 11 types of antimicrobial agents were identified and chromosomal mutation of p.S83L in gyrA gene (88.57 %, 31/35) was the most common. Furthermore, 110 virulence factors (VFs) were identified, with those related to iron uptake and storage, adhesion, secretion and endotoxin production being the most prevalent. This study underscored the importance of rational use of antimicrobial agents in poultry farming and provided critical insights into the distribution of ARGs and VFs among retail duck-derived MDR E. coli.
Collapse
Affiliation(s)
- Jialan Chen
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Ruyi Kuang
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Haotian Liu
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Lei Yang
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Jiarui Li
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Junlin Li
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Jing Xia
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China; Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of China, Chengdu, China; Key Laboratory of Animal Diseases and Human Health of Sichuan Province, Chengdu, China
| | - Yong Huang
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China; Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of China, Chengdu, China; Key Laboratory of Animal Diseases and Human Health of Sichuan Province, Chengdu, China
| | - Min Cui
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China; Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of China, Chengdu, China; Key Laboratory of Animal Diseases and Human Health of Sichuan Province, Chengdu, China
| | - Likou Zou
- College of Resources, Sichuan Agricultural University, Chengdu, China
| | - Xinfeng Han
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China; Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of China, Chengdu, China; Key Laboratory of Animal Diseases and Human Health of Sichuan Province, Chengdu, China.
| |
Collapse
|
6
|
Qv M, Wu Q, Wang W, Wang H, Zhu L. Metagenomic insights into the response of microbial metabolic function and extracellular polymeric substances from microalgae-bacteria consortia to fluoroquinolone antibiotics. JOURNAL OF ENVIRONMENTAL MANAGEMENT 2025; 381:125283. [PMID: 40203710 DOI: 10.1016/j.jenvman.2025.125283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Revised: 03/19/2025] [Accepted: 04/04/2025] [Indexed: 04/11/2025]
Abstract
Microalgae-bacteria consortia (MBC) are considered a promising bioremediation technology for removing pollutants from swine wastewater. However, the overuse of antibiotics poses challenges to the effective functioning of MBC. In this study, the removal efficiency of nutrients in wastewater by MBC under different antibiotic concentrations (0, 1, 5, 10 and 50 mg/L) was evaluated. The changes of functional microbial abundance were elucidated and the response mechanism of MBC against antibiotics was investigated. Antibiotics inhibited the accumulation of MBC biomass and reduced the removal efficiency of ammonia nitrogen and total phosphorus in wastewater by 8.39 % and 8.74 % respectively. In addition, antibiotics affected the relative abundance of microorganisms (Raineyella, from 30.72 % to 15.96 %) and functional genes (glnA, gudB, NirK, NirBD, NarB, NapAB, NorBC and NosZEPS) involved in N metabolism. MBC could defend against the adverse effects of antibiotics by regulating the content of proteins in the extracellular polymeric substances.
Collapse
Affiliation(s)
- Mingxiang Qv
- School of Resource and Environmental Sciences, Hubei Key Laboratory of Biomass-Resources Chemistry and Environmental Biotechnology, and Hubei International Scientific and Technological Cooperation Base of Sustainable Resource and Energy, Wuhan University, Wuhan, 430079, China
| | - Qirui Wu
- School of Resource and Environmental Sciences, Hubei Key Laboratory of Biomass-Resources Chemistry and Environmental Biotechnology, and Hubei International Scientific and Technological Cooperation Base of Sustainable Resource and Energy, Wuhan University, Wuhan, 430079, China
| | - Wei Wang
- School of Resource and Environmental Sciences, Hubei Key Laboratory of Biomass-Resources Chemistry and Environmental Biotechnology, and Hubei International Scientific and Technological Cooperation Base of Sustainable Resource and Energy, Wuhan University, Wuhan, 430079, China
| | - Hanzhi Wang
- School of Resource and Environmental Sciences, Hubei Key Laboratory of Biomass-Resources Chemistry and Environmental Biotechnology, and Hubei International Scientific and Technological Cooperation Base of Sustainable Resource and Energy, Wuhan University, Wuhan, 430079, China
| | - Liandong Zhu
- School of Resource and Environmental Sciences, Hubei Key Laboratory of Biomass-Resources Chemistry and Environmental Biotechnology, and Hubei International Scientific and Technological Cooperation Base of Sustainable Resource and Energy, Wuhan University, Wuhan, 430079, China; State Key Laboratory of Water Resources Engineering and Management, Wuhan University, Wuhan, 430072, China.
| |
Collapse
|
7
|
Turbant F, Lewandowska N, Bloch S, Wien F, Chauvet H, Węgrzyn G, Arluison V. Hfq influences ciprofloxacin accumulation in Escherichia coli independently of ompC and ompF post-transcriptional regulation. J Appl Genet 2025; 66:449-457. [PMID: 39920540 DOI: 10.1007/s13353-025-00945-9] [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: 10/04/2024] [Revised: 01/21/2025] [Accepted: 01/22/2025] [Indexed: 02/09/2025]
Abstract
The antibiotic resistance of pathogenic bacteria is currently one of the major problems in medicine, and finding novel antibacterial agents is one of the most difficult tasks in the field of biomedical sciences. Studies on such tasks can be successful only if genetic and molecular mechanisms leading to antibiotic resistance/sensitivity are understood. Previous reports indicated that the bacterial protein Hfq, discovered as an RNA chaperone but subsequently demonstrated to play also other functions in cells, is involved in the mechanisms of the response of bacterial cells to antibiotics. Recently, it was found that Hfq dysfunction resulted in more effective accumulation of an antibiotic ciprofloxacin in Escherichia coli cells irrespective of the presence or absence of the AcrB efflux pump. However, small RNA-mediated impairment of expression of the ompF gene, which encodes a porin involved in antibiotics influx, reversed the effects of the absence of Hfq on the antibiotic accumulation. This led to the hypothesis that Hfq might influence ciprofloxacin accumulation in the manner independent on its RNA chaperone function, as this protein might also influence cellular membrane structure and functions. Here, we demonstrate that in ompC and ompF mutants of E. coli, accumulation of ciprofloxacin is significantly impaired in the absence of Hfq or its C-terminal domain. These results corroborate the above-mentioned hypothesis on a sRNA-independent mechanism of Hfq-mediated modulation of the antibiotic transmembrane transport. Since fluoroquinolones use both protein- and lipid-mediated pathways to cross the outer membrane, Hfq may influence both processes. This possibility will be discussed herein.
Collapse
Affiliation(s)
- Florian Turbant
- Synchrotron SOLEIL, L Orme Des Merisiers, Saint Aubin BP48, 91192, Gif-Sur-Yvette, France
- Laboratoire Léon Brillouin LLB, UMR12, CEA CNRS, CEA Saclay, 91191, Gif-Sur-Yvette, France
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308, Gdansk, Poland
| | - Natalia Lewandowska
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308, Gdansk, Poland
| | - Sylwia Bloch
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308, Gdansk, Poland
| | - Frank Wien
- Synchrotron SOLEIL, L Orme Des Merisiers, Saint Aubin BP48, 91192, Gif-Sur-Yvette, France
| | - Hugo Chauvet
- Synchrotron SOLEIL, L Orme Des Merisiers, Saint Aubin BP48, 91192, Gif-Sur-Yvette, France
| | - Grzegorz Węgrzyn
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308, Gdansk, Poland.
| | - Véronique Arluison
- Laboratoire Léon Brillouin LLB, UMR12, CEA CNRS, CEA Saclay, 91191, Gif-Sur-Yvette, France.
- UFR SDV, Université Paris Cité, 75013, Paris, France.
| |
Collapse
|
8
|
Yaffe E, Dethlefsen L, Patankar AV, Gui C, Holmes S, Relman DA. Brief antibiotic use drives human gut bacteria towards low-cost resistance. Nature 2025; 641:182-191. [PMID: 40269166 DOI: 10.1038/s41586-025-08781-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 02/12/2025] [Indexed: 04/25/2025]
Abstract
Understanding the relationship between antibiotic use and the evolution of antimicrobial resistance is vital for effective antibiotic stewardship. Yet, animal models and in vitro experiments poorly replicate real-world conditions1. To explain how resistance evolves in vivo, we exposed 60 human participants to ciprofloxacin and used longitudinal stool samples and a new computational method to assemble the genomes of 5,665 populations of commensal bacterial species within participants. Analysis of 2.3 million polymorphic sequence variants revealed 513 populations that underwent selective sweeps. We found convergent evolution focused on DNA gyrase and evidence of dispersed selective pressure at other genomic loci. Roughly 10% of susceptible bacterial populations evolved towards resistance through sweeps that involved substitutions at a specific amino acid in gyrase. The evolution of gyrase was associated with large populations that decreased in relative abundance during exposure. Sweeps persisted for more than 10 weeks in most cases and were not projected to revert within a year. Targeted amplification showed that gyrase mutations arose de novo within the participants and exhibited no measurable fitness cost. These findings revealed that brief ciprofloxacin exposure drives the evolution of resistance in gut commensals, with mutations persisting long after exposure. This study underscores the capacity of the human gut to promote the evolution of resistance and identifies key genomic and ecological factors that shape bacterial adaptation in vivo.
Collapse
Affiliation(s)
- Eitan Yaffe
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.
- Infectious Diseases Section, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA.
| | - Les Dethlefsen
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Arati V Patankar
- Infectious Diseases Section, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Chen Gui
- Infectious Diseases Section, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Susan Holmes
- Department of Statistics, Stanford University, Stanford, CA, USA
| | - David A Relman
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Infectious Diseases Section, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
9
|
Ball G, Stevenson J, Boroujeni FA, Jacobson B, Kuehne SA, Lucas M, Walmsley AD, Prentice P, Pikramenou Z. Non-porous silica nanoparticles as a cavitation sensitive vehicle for antibiotic delivery. ULTRASONICS SONOCHEMISTRY 2025; 116:107316. [PMID: 40120341 PMCID: PMC11981769 DOI: 10.1016/j.ultsonch.2025.107316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/28/2025] [Accepted: 03/16/2025] [Indexed: 03/25/2025]
Abstract
Ultrasound stimulated drug delivery is attractive for controlled dose and localised delivery to reduce excess loss of drug and side effects, which for antibiotics is pertinent to the fight against antimicrobial resistance. Low frequency ultrasound is commonly used in dental clinical practice for bacterial biofilm removal and is an attractive versatile stimulus for drug release. Here we introduce nonporous (amorphous) silica nanoparticles as a biocompatible, encapsulant for triggered drug release by low frequency ultrasound. A 20 kHz ultrasonic sonotrode is used in to evaluate the release of the antibiotic ciprofloxacin, CPX, from non-porous particles, CPX ⊂ SiO2. Laser doppler vibrometry (LDV) was employed to characterise the ultrasonic vibration displacement of the sonotrode. Drug release from CPX ⊂ SiO2 was monitored for increasing the tip displacement. Clinically relevant quantities of CPX release (5.7 mg/L) occurred at 40 μm tip displacement in our studies. A strong correlation was observed between cavitation features in the acoustic spectra and drug release from CPX ⊂ SiO2. Silica nanoparticles with and without encapsulated CPX, CPX ⊂ SiO2 and SiO2, respectively, were found to promote cavitation at lower amplitudes confirmed by high-speed imaging, in contrast to mesoporous particles with and without adsorbed CPX, CPX@m-SiO2 and m-SiO2. Spectra of the emissions collected via an acoustic cavitation detector supported these results. Our studies demonstrate a novel platform for drug delivery employing low frequency ultrasound for synergistic enhancement of cavitation effects and triggered drug release.
Collapse
Affiliation(s)
- Grace Ball
- School of Chemistry, University of Birmingham, Edgbaston B15 2TT, UK
| | - Jack Stevenson
- Centre for Medical & Industrial Ultrasonics, James Watt School of Engineering, University of Glasgow, Glasgow G12 8QQ, UK
| | - Faraz Amini Boroujeni
- Centre for Medical & Industrial Ultrasonics, James Watt School of Engineering, University of Glasgow, Glasgow G12 8QQ, UK
| | - Ben Jacobson
- Centre for Medical & Industrial Ultrasonics, James Watt School of Engineering, University of Glasgow, Glasgow G12 8QQ, UK
| | - Sarah A Kuehne
- School of Science & Technology, Nottingham Trent University, Nottingham NG11 8NS, UK
| | - Margaret Lucas
- Centre for Medical & Industrial Ultrasonics, James Watt School of Engineering, University of Glasgow, Glasgow G12 8QQ, UK
| | - Anthony Damien Walmsley
- School of Dentistry, College of Medical and Dental Sciences, University of Birmingham, Birmingham B5 7EG, UK
| | - Paul Prentice
- Centre for Medical & Industrial Ultrasonics, James Watt School of Engineering, University of Glasgow, Glasgow G12 8QQ, UK
| | - Zoe Pikramenou
- School of Chemistry, University of Birmingham, Edgbaston B15 2TT, UK
| |
Collapse
|
10
|
Selvam PK, Mudipalli Elavarasu S, C GPD, Vasudevan K. Genetic insights into Staphylococcus aureus resistance: exploring AMR genes and molecular interactions. Pathog Glob Health 2025; 119:87-98. [PMID: 40079538 DOI: 10.1080/20477724.2025.2477337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/15/2025] Open
Abstract
Antimicrobial resistance (AMR) among microorganisms remains a significant global concern in this century, posing an ongoing challenge for humanity. To solve this issue effectively, it is crucial to understand the genes responsible for AMR and how they create resistance. Staphylococcus aureus, which has AMR genes imparting resistance against numerous antibiotics, was the main subject of our investigation. We conducted a phylogenetic investigation to explore the evolutionary history of the gene network comprising rpl, rpoC, parE, and gyrB, providing insights into their genetic relationships and evolutionary connections. A gene interaction network with 46 functional partners was built and examined from the STRING Database and Cytoscape to increase our understanding. According to Cluego's enrichment analysis, 20 genes are significantly involved in biological processes, as are 14 genes in cellular components and 16 genes in molecular functions. RpoB, RpoC, FusA, RplI, and RpsL had the most interactions by Cytohubba when the degree and closeness of the network were studied, according to the gene interaction network analysis. Understanding the molecular basis of AMR requires analysis of the enriched pathways and Gene Ontologies (GO). The proposed study may also help researchers find new ways to battle the multidrug resistance of Staphylococcus aureus.
Collapse
Affiliation(s)
- Prasanna Kumar Selvam
- Manipal Academy of Higher Education (MAHE), Manipal, India
- Institute of Bioinformatics, International Technology Park, Bangalore, India
| | | | | | - Karthick Vasudevan
- Manipal Academy of Higher Education (MAHE), Manipal, India
- Institute of Bioinformatics, International Technology Park, Bangalore, India
| |
Collapse
|
11
|
Soukup CRM, Duffin RN, Burke KJ, Meagher L, Andrews PC. The antibacterial activity and selectivity of bismuth(III) tris(8-hydroxyquinolinates). J Inorg Biochem 2025; 266:112836. [PMID: 39919434 DOI: 10.1016/j.jinorgbio.2025.112836] [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: 01/27/2025] [Accepted: 01/30/2025] [Indexed: 02/09/2025]
Abstract
The series of bismuth(III) tris(8-hydroxyquinolinates); [Bi(Q")3] (1), [Bi(Q'Cl)3] (2), [Bi(QCl2)3] (3), [Bi(QBr2)3] (4), and [Bi(QI2)3] (5) (where Q"-H = C9H7NO; Q'Cl-H = C9H6NOCl, QCl2-H = C9H5NOCl2; QBr2-H = C9H5NOBr2; and QI2-H = C9H5NOI2) were synthesised, fully characterised, and evaluated for their antibacterial activity towards three Gram-positive bacteria (vancomycin-resistant E. faecalis, S. aureus, methicillin-resistant S. aureus), and four Gram-negative bacteria (A. baumannii, P. aeruginosa, K. pneumoniae, and E. coli) and also their cytotoxicity towards mammalian cells. New crystallographic data on 4 indicates it is dimeric in the solid state through 'Bi2O2' bridging which is consistent with data previously reported for 5. The five complexes (1-5) all exhibited good but variable antibacterial activity and selectivity. Complexes 2 and 5 showed significant activity towards Gram-positive bacteria with MIC (minimum inhibitory concentration) values ranging from 0.78 μM - 3.13 μM and selectivity indices of 6.2 - ≥16.0. For Gram-negative species, complexes 3 and 4 exhibited highly selective activity towards multi-drug resistant strains of A. baumannii with a range of MIC values 0.39-1.56 μM and selectivity indices of 3.14-7.23 respectively. While some of the 8-hydroxyquinolines themselves show reasonable antibacterial activity this is generally enhanced through complexation to bismuth(III).
Collapse
Affiliation(s)
- Charles R M Soukup
- School of Chemistry, Monash University, Clayton, Melbourne, VIC 3800, Australia
| | - Rebekah N Duffin
- School of Chemistry, Monash University, Clayton, Melbourne, VIC 3800, Australia
| | - Kirralee J Burke
- School of Chemistry, Monash University, Clayton, Melbourne, VIC 3800, Australia
| | - Laurence Meagher
- Department of Materials Science and Engineering, Monash University, Clayton, Melbourne, VIC 3800, Australia
| | - Philip C Andrews
- School of Chemistry, Monash University, Clayton, Melbourne, VIC 3800, Australia.
| |
Collapse
|
12
|
Sharma V, Saini M, Das R, Chauhan S, Sharma D, Mujwar S, Gupta S, Mehta DK. Recent Updates on Antibacterial Quinolones: Green Synthesis, Mode of Interaction and Structure-Activity Relationship. Chem Biodivers 2025; 22:e202401936. [PMID: 39756027 DOI: 10.1002/cbdv.202401936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 12/21/2024] [Accepted: 12/23/2024] [Indexed: 01/07/2025]
Abstract
Quinolone antibiotics are a crucial class of synthetic antibacterial agents, widely utilized due to their broad spectrum of antibacterial activity. Due to the development of antimicrobial resistance, the potency of quinolone drugs decreased. Many conventional methods have been developed to elevate amination rate and to improve yield. These methods are generally characterized by prolonged reaction durations, high boiling solvents, harsh conditions, costly reagents and excessive heat generation, which have adversely affected the therapeutic efficacy of these compounds. Recently, green chemistry has focused on sustainable chemistry-dependent quinolone analogue synthesis methods that significantly reduce bacterial infections. These methods include one-pot synthesis, photoredox catalysis, phase transfer catalysis, ultrasonic irradiation, microwave-assisted, green solvent and catalyst-free synthesis, which often utilize energy-efficient, non-toxic and less time-consuming techniques, aligning with green chemistry principles to improve safety and environmental impact. Researchers continuously explore innovative approaches to applying these methods in synthetic reactions. This review includes a comprehensive analysis of synthetic literature from the past 15 years from Scopus, PubMed, Embase and WOS using keywords, such as green chemistry, quinolone and antibacterial, highlighting significant advancements and emerging trends. This work's importance lies in its extensive literature overview on green synthesis methods for quinolones and related heterocyclic compounds. Furthermore, to provide useful information for the generation of future antibacterial drugs, some structural-activity relationship studies and in silico studies have also been included to investigate the stable binding interactions between quinolone leads and various target proteins.
Collapse
Affiliation(s)
- Vishal Sharma
- Department of Pharmaceutical Chemistry, MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Ambala, India
| | - Monika Saini
- Department of Pharmaceutical Chemistry, MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Ambala, India
| | - Rina Das
- Department of Pharmaceutical Chemistry, MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Ambala, India
| | - Samrat Chauhan
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Diksha Sharma
- Department of Pharmaceutical Chemistry, Swami Devidyal College of Pharmacy, Barwala, India
| | - Somdutt Mujwar
- Department of Pharmaceutical Chemistry, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Sumeet Gupta
- Department of Pharmacology, MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Ambala, India
| | - Dinesh Kumar Mehta
- Department of Pharmaceutical Chemistry, MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Ambala, India
| |
Collapse
|
13
|
Alraey Y, Assiry MM, Ahmad I, Rab SO, Alqahtani A, Bawazeer AAS, Hussien T, Syed Khaja AS, Saleem M. Hospital epidemiology and antimicrobial susceptibility of isolated methicillin-resistant Staphylococcus aureus: a 5-year retrospective study at a tertiary care centre in Aseer Region, Saudi Arabia. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04218-4. [PMID: 40299023 DOI: 10.1007/s00210-025-04218-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Accepted: 04/22/2025] [Indexed: 04/30/2025]
Abstract
Between 2019 and 2023, an analysis at the tertiary care centre revealed that 849 out of 1,951 Staphylococcus aureus isolates were methicillin-resistant (Staphylococcus aureus), commonly known as MRSA. According to statistical results, the rate of MRSA infection was markedly higher in patients who received inpatient department care (IPD) at 46.8% than in outpatient department (OPD) at 37.0% (p < 0.0001). Results showed males infected at a rate of 45.3% compared to females who had a rate of 39.4% (p = 0.0198) and age groups demonstrated no significant association (p > 0.05). The choice of specimen type affected MRSA detection rates as endotracheal tubes (32.5%, p = 0.004) together with 'Other' samples (e.g.: CSF, bone, bone marrow, bronchial lavage, abdominal aspirate, semen, ETT tip, femoral tip, jugular tip) (54.2%, p = 0.0068) presented higher proportions of infections. Individuals with benign prostatic hyperplasia showed an increased risk of MRSA infection (OR, 1.8; p < 0.0001) along with patients who had chronic lung disease (OR, 1.2; p = 0.048) or recent antibacterial substance use (OR, 2.5; p < 0.0001) while steroid use reduced the risk of MRSA infection (OR, 0.8; p = 0.002). MRSA showed complete resistance against β-lactam antibacterial substances, while all samples remained susceptible to Daptomycin, Linezolid, Nitrofurantoin, and Tigecycline. The sensitivity rate of vancomycin reached 95%, but MRSA displayed significantly reduced susceptibility to fluoroquinolones at 39.3% to 46.3% compared to MSSA, with rates at 81.5% to 85.5%. The percentage of macrolide-resistant bacteria was higher in MRSA, since they showed 28.3%-37.1% susceptibility rates, whereas MSSA had 61.3%-61.8% susceptibility rates. The anti-staphylococcal activity between MRSA and MSSA exceeded 94% for Rifampicin, Teicoplanin, and Fosfomycin. The antibacterial substances, Gentamicin and Tobramycin, showed high sensitivity against MSSA, since their sensitivity reached 92.3% and 91.3%, respectively. Both agents had good sensitivity against MRSA, with rates of 82.4% for gentamicin and 88.9% for tobramycin. Strict antimicrobial stewardship should be implemented as a priority to control the spread of MRSA. Last-line therapies such as vancomycin, daptomycin, and linezolid remain essential treatment options. Regular antimicrobial susceptibility testing is crucial for healthcare professionals to optimize therapy and prevent the development of drug resistance.
Collapse
Affiliation(s)
- Yasser Alraey
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | | | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Safia Obaidur Rab
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Abdulaziz Alqahtani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | | | - Tarig Hussien
- Department of Obstetrics and Gynaecology, College of Medicine, University of Ha'il, Hail, Saudi Arabia
| | | | - Mohd Saleem
- Department of Pathology, College of Medicine, University of Ha'il, Hail, Saudi Arabia.
| |
Collapse
|
14
|
Islam MT, Tasnim J, Basri R, Sakib MN, Ullah W, Nahar KS, Sadique A, Sultana M, Arakawa E, Morita M, Watanabe H, Boucher YF, Huq A, Colwell RR, Alam M. Vibrio cholerae O47 associated with a cholera-like diarrheal outbreak concurrent with seasonal cholera in Bangladesh. mSphere 2025; 10:e0083124. [PMID: 40172221 PMCID: PMC12039230 DOI: 10.1128/msphere.00831-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 03/04/2025] [Indexed: 04/04/2025] Open
Abstract
The Ganges delta of the Bay of Bengal is a recognized hotspot for the emergence and spread of novel variants of Vibrio cholerae. Despite being a diverse species, very little information is available concerning environmental and human-associated aspects of V. cholerae serogroups, other than the two major epidemic-related serogroups O1 and O139. This represents a crucial gap in understanding the spectrum of diversity, ecology, and epidemiology of the species influencing the dynamics of global cholera. In this study, we describe an emerging variant of V. cholerae displaying the antigenic property of serogroup O47, associated with a cholera-like outbreak in coastal Bangladesh where cholera has been endemic for centuries. This outbreak coincides with a rise in cases of cholera caused by V. cholerae O1, as well as frequency of isolation of serogroups O47 and O1 from the environment. The V. cholerae O47 isolates proved clonal in nature, and their genome biology revealed distinct features, with respect to multidrug resistance (MDR), serogroup-specific genes, genomic island combinations, and overall phylogenetic properties. Genome comparison confirmed the absence of canonical virulence factors of V. cholerae O1 and O139, namely, cholera toxin (CTX) and toxin-co-regulated pili (TCP), and the presence of putative virulence factors including type 3 secretion system (T3SS) and an MDR pseudo-compound transposon, carrying genes for macrolide resistance and extended spectrum beta-lactamase. Results of the study suggest that V. cholerae O47 could represent an emerging Vibrio pathogen with the potential to spread virulence and antimicrobial resistance traits impacting the management of cholera-like diseases.IMPORTANCEDespite the global insurgence of human diseases caused by Vibrios in recent years, most research focuses only on the O1 serogroup of V. cholerae, leaving a significant gap concerning the environmental and human-associated aspects of other serogroups found in nature. Although other serogroups are often found associated with sporadic diarrhea cases, in 1992-1993, a massive cholera-like diarrhea epidemic was initiated by a "non-O1" serogroup, namely, O139 that temporally displaced O1 from endemic cholera in the Bay of Bengal villages of Bangladesh and India, highlighting the potential threat they might pose. This study describes yet another emerging variant of V. cholerae, displaying the antigenic property of serogroup O47, associated with a cholera-like outbreak in a coastal locality in Bangladesh. Findings of the study offer critical insights into the genome biology of V. cholerae O47 and its potential implications for understanding their ecology and epidemiology of cholera-like diseases.
Collapse
Affiliation(s)
- Mohammad Tarequl Islam
- International Centre for Diarrhoeal Disease Research, Bangladesh (iccdr,b), Dhaka, Bangladesh
| | - Jarin Tasnim
- International Centre for Diarrhoeal Disease Research, Bangladesh (iccdr,b), Dhaka, Bangladesh
| | - Rabeya Basri
- International Centre for Diarrhoeal Disease Research, Bangladesh (iccdr,b), Dhaka, Bangladesh
| | - Mohammad Nazmus Sakib
- International Centre for Diarrhoeal Disease Research, Bangladesh (iccdr,b), Dhaka, Bangladesh
| | - Wali Ullah
- International Centre for Diarrhoeal Disease Research, Bangladesh (iccdr,b), Dhaka, Bangladesh
| | - Kazi Sumaita Nahar
- International Centre for Diarrhoeal Disease Research, Bangladesh (iccdr,b), Dhaka, Bangladesh
| | - Abdus Sadique
- NSU Genome Research Institute, North South University, Dhaka, Bangladesh
| | - Marzia Sultana
- International Centre for Diarrhoeal Disease Research, Bangladesh (iccdr,b), Dhaka, Bangladesh
| | - Eiji Arakawa
- Department of Bacteriology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Masatomo Morita
- Department of Bacteriology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Haruo Watanabe
- Department of Bacteriology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Yann F. Boucher
- Saw Swee Hock School of Public Health, National University of Singapore, , Singapore
| | - Anwar Huq
- Maryland Pathogen Research Institute, University of Maryland, College Park, Maryland, USA
| | - Rita R. Colwell
- Maryland Pathogen Research Institute, University of Maryland, College Park, Maryland, USA
- Institute for Advanced Computer Studies and Department of Cell Biology and Molecular Biology, University of Maryland, College Park, Maryland, USA
| | - Munirul Alam
- International Centre for Diarrhoeal Disease Research, Bangladesh (iccdr,b), Dhaka, Bangladesh
| |
Collapse
|
15
|
Liang S, Cai W, Mao R, Chen M, Dai X, Jin X, Kong W. Three simple and cost-effective assays for AAC(6')-Ib-cr enzyme activity. Front Microbiol 2025; 16:1513425. [PMID: 40351320 PMCID: PMC12061968 DOI: 10.3389/fmicb.2025.1513425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 04/03/2025] [Indexed: 05/14/2025] Open
Abstract
The enzyme AAC(6')-Ib-cr belongs to plasmid-mediated quinolone resistance (PMQR), first reported in 2006 and now widely disseminating. Here, we developed three phenotypic methods to detect AAC(6')-Ib-cr enzyme-producing Enterobacteriaceae (APE), two of which are proposed innovatively in this research. These tests are based on the following principles: (i) Matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF MS) can measure the mass shift of 42 Da resulting from ciprofloxacin acetylation by the AAC(6')-Ib-cr enzyme. (ii) Co-incubation of ciprofloxacin disks with APE results in inactivation of the drug activity, making it unable to inhibit the growth of the indicator organism. We named this test the quinolone inactivation method (QIM). (iii) Based on the principles of the modified Hodge test, we designed the quinolone Hodge test (QHT). Through exploration of optimal conditions for three methods, we found that MALDI-TOF MS provides the most intuitive results after 1 h of incubation. The interpretability of the QIM and QHT results was significantly improved when the indicator organism E. coli ATCC25922 was replaced with a quinolone-slightly-resistant isolate. However, Proteus mirabilis was excluded from both QIM and QHT due to its swarming motility. Next, a validation study was conducted using a prospectively collected set of 187 clinical strains, demonstrating 100% specificity (MSM: 141/141; QIM, QHT: 135/135) and 100% sensitivity (MSM: 46/46; QIM, QHT: 33/33) compared to the genotype. In a word, this study presented three simple, efficient, and cost-effective methods for detecting APE, suitable for clinical microbiology laboratories under various conditions for the prevention and control of hospital infections.
Collapse
Affiliation(s)
- Shizhou Liang
- Department of Clinical Laboratory, Wenzhou TCM Hospital of Zhejiang Chinese Medical University, Wenzhou, Zhejiang, China
| | - Wenpin Cai
- Department of Clinical Laboratory, Wenzhou TCM Hospital of Zhejiang Chinese Medical University, Wenzhou, Zhejiang, China
- Department of TCM Science and Research Center, Wenzhou, Zhejiang, China
| | - Ruiben Mao
- Department of Clinical Laboratory, Wenzhou TCM Hospital of Zhejiang Chinese Medical University, Wenzhou, Zhejiang, China
| | - Mengquan Chen
- Department of Clinical Laboratory, Wenzhou TCM Hospital of Zhejiang Chinese Medical University, Wenzhou, Zhejiang, China
| | - Xianning Dai
- Department of Clinical Laboratory, Wenzhou People’s Hospital, Wenzhou Women and Children’s Hospital, Wenzhou, Zhejiang, China
| | - Xiaoli Jin
- Department of Clinical Laboratory, Wenzhou TCM Hospital of Zhejiang Chinese Medical University, Wenzhou, Zhejiang, China
| | - Wanzhong Kong
- Department of Clinical Laboratory, Wenzhou TCM Hospital of Zhejiang Chinese Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
16
|
Bui Thi TM, Chen T, Luo T, Leroux Y, Hanna K, Boily JF. Ligand-limited oxidation of ciprofloxacin by Mn(III). JOURNAL OF HAZARDOUS MATERIALS 2025; 493:138373. [PMID: 40306247 DOI: 10.1016/j.jhazmat.2025.138373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 04/04/2025] [Accepted: 04/21/2025] [Indexed: 05/02/2025]
Abstract
Mn(III) species play critical roles in determining the environmental fate of antibiotics released into natural systems. However, their reactivity is strongly influenced by complexation reactions with (in)organic ligands. This study investigates the impact of Mn(III) complexation with pyrophosphate (PP), a model environmental ligand, on the redox-driven degradation of ciprofloxacin (CIP), a widely used antibiotic and environmental contaminant. Spectroscopic analysis and thermodynamic modeling revealed that Mn(III)-PP complexes initially dissociate into MnOH2+ species, which can then disproportionate and form MnO2 colloids. Both dissociation and disproportionation reactions had comparable trends at pH 4 and 7, with reactivities that were strongly dependent on Mn(III):PP ratios. The progress of CIP oxidation following direct coordination with Mn compounds over time was sigmoidal, with an initial lag phase attributed to Mn(III)-PP complex dissociation and disproportionation. CIP degradation was predominantly governed by pH, with maximal rate constants decreasing from k = 0.390 h-1 at pH 3 to k = 0.065 h-1 at pH 5, and no CIP removal under circumneutral to alkaline conditions. Cyclic voltammetry also confirmed that the strongly pH-dependent redox potential of the Mn(III)/Mn(II) couple aligned with facile CIP oxidation under acidic conditions. These collective findings indicated that ligand complexation, such as with PP, enhanced Mn(III) stability and mitigated dissociation and disproportionation reactions. The new insight provided by this work on the speciation and redox activity of Mn(III) should thereby be considered for understanding ciprofloxacin degradation in contaminated water systems.
Collapse
Affiliation(s)
- Tra My Bui Thi
- Univ. Rennes, Ecole Nationale Supérieure de Chimie de Rennes, CNRS, ISCR-UMR 6226, Rennes F-35000, France
| | - Tao Chen
- Univ. Rennes, Ecole Nationale Supérieure de Chimie de Rennes, CNRS, ISCR-UMR 6226, Rennes F-35000, France
| | - Tao Luo
- Department of Chemistry, Umeå University, Umeå SE-901 87, Sweden
| | - Yann Leroux
- Univ. Rennes, CNRS, ISCR-UMR 6226, Rennes F-35000, France
| | - Khalil Hanna
- Univ. Rennes, Ecole Nationale Supérieure de Chimie de Rennes, CNRS, ISCR-UMR 6226, Rennes F-35000, France
| | | |
Collapse
|
17
|
Islam MZ, Hossain F, Yamazaki M. Single-cell analysis of antimicrobial compound-induced cell death of bacterial cells. J Antimicrob Chemother 2025:dkaf116. [PMID: 40238567 DOI: 10.1093/jac/dkaf116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2025] Open
Abstract
Due to the stochasticity of metabolic reactions and cell cycles of bacterial cells, it is necessary to examine the antibacterial activities of antimicrobial compounds (AMCs) such as antibiotics and antimicrobial peptides (AMPs) at the single-cell level. Here, we review recent studies of the bactericidal activities of AMCs at the single-cell level. First, we discuss recent investigations of the interaction of various AMPs with single bacterial cells, as monitored in real time using optical microscopy. This strategy provides information on AMP-induced membrane damage in single cells [e.g. the onset time of damage to the cell membrane (CM) and outer membrane of single cells]. The rate of AMP-induced CM damage is estimated as the fraction of cells with CM damage [Pdamage (t)] at a specific interaction time t. Second, we discuss the use of single-cell analysis of the bactericidal activity of AMCs. The fraction of dead cells after the exposure to AMCs for time t is determined as the fraction of the microcolonies containing only one cell [Psingle (t)]. For some AMPs, the Pdamage (t) and Psingle (t) values are similar, indicating that AMP-induced CM damage is the direct cause of cell death. Third, we discuss single-cell analysis of the processes and mechanisms of antibiotic-induced cell death. For example, fluoroquinolones and aminoglycosides are observed to induce cytoplasmic condensation and cell lysis, leading to cell death. Based on these studies, we provide our perspective on future investigations using single-cell analysis to assess the processes and the mechanisms of the bactericidal activities of AMCs.
Collapse
Affiliation(s)
- Md Zahidul Islam
- Nanomaterials Research Division, Research Institute of Electronics, Shizuoka University, Shizuoka 422-8529, Japan
- Department of Biotechnology and Genetic Engineering, Jahangirnagar University, Savar, Dhaka 1342, Bangladesh
| | - Farzana Hossain
- Nanomaterials Research Division, Research Institute of Electronics, Shizuoka University, Shizuoka 422-8529, Japan
- Department of Biochemistry and Microbiology, School of Health and Life Sciences, North South University, Bashundhara, Dhaka 1229, Bangladesh
| | - Masahito Yamazaki
- Nanomaterials Research Division, Research Institute of Electronics, Shizuoka University, Shizuoka 422-8529, Japan
- Integrated Bioscience Section, Graduate School of Science and Technology, Shizuoka University, Shizuoka 422-8529, Japan
- Department of Science, Graduate School of Integrated Science and Technology, Shizuoka University, 836 Oya, Suruga-ku, Shizuoka 422-8529, Japan
| |
Collapse
|
18
|
El-Saghier AM, Abosella L, Hassan A, Elakesh EO, Bräse S, Abuo-Rahma GEDA, Aziz HA. Design, Synthesis, and In Silico Studies of New Norfloxacin Analogues with Broad Spectrum Antibacterial Activity via Topoisomerase II Inhibition. Pharmaceuticals (Basel) 2025; 18:545. [PMID: 40283980 PMCID: PMC12030355 DOI: 10.3390/ph18040545] [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: 03/07/2025] [Revised: 04/02/2025] [Accepted: 04/05/2025] [Indexed: 04/29/2025] Open
Abstract
Background: Novel norfloxacin derivatives were synthesized, characterized, and screened for their antibacterial activity against Gram-positive strain S. aureus ATCC 6538 and Gram-negative strains; E. coli ATCC 25923, K. pneumoniae ATCC 10031, and P. aeruginosa ATCC 27853 using the agar cup diffusion method. Results: The results revealed that compounds 6-17 exhibited more potent activity towards S. aureus ATCC 6538 with MIC values of 0.21-3.61 µM than norfloxacin with a MIC of 7.83 µM. The most potent compound, 6, showed 37-fold more potency than norfloxacin. More importantly, compound 7 exhibited more potent activity against MRSA than norfloxacin, with MIC values of 0.80 and 1.96 µM, respectively. Meanwhile, compounds 15 and 16 have potent activity towards the Gram-negative strains with MIC values of 0.20-0.79 µM compared with norfloxacin with a MIC of 0.24 µM. Moreover, the potent compounds showed higher activity towards topoisomerase II enzymes, especially against topoisomerase IV, which confirms the docking study with the S. aureus gyrase enzyme active binding site (PDB ID: 2XCT). In addition, cytotoxicity assays of the most potent compounds showed that compounds 6, 7, 15, and 16 have negligible risks of toxic effects when evaluated against the normal cell line WI 38. Conclusions: The docking study of the most potent compounds 6, 7, 15, and 16 on the gyrase enzyme active site (PDB: 2XCT) aligns their antibacterial activity and topoisomerase inhibition. The physicochemical and pharmacokinetic characteristics of the target derivatives were forecasted via SwissADME. Hence, these compounds are considered promising antibacterial candidates that require further optimization.
Collapse
Affiliation(s)
- Ahmed M. El-Saghier
- Chemistry Department, Faculty of Science, Sohag University, Sohag 82524, Egypt;
| | - Laila Abosella
- Chemistry Department, Faculty of Science, Sohag University, Sohag 82524, Egypt;
- Medicinal Chemistry Department, Faculty of Pharmacy—Al-Jmail, Sabratha University, Sabratha P.O. Box 250, Libya
| | - Abdelfattah Hassan
- Medicinal Chemistry Department, Faculty of Pharmacy, South Valley University, Qena 83523, Egypt;
- Medicinal Chemistry Department, Faculty of Pharmacy, National South Valley University, Qena 83523, Egypt
| | - Esmail O. Elakesh
- Chemistry Department, Faculty of Science, University of Zawia, Al Zawiya 16418, Libya;
| | - Stefan Bräse
- Institute for Biological and Chemical System, Karlsruhe Institute of Technology, 76131 Karlsruhe, Germany
| | - Gamal El-Din A. Abuo-Rahma
- Medicinal Chemistry Department, Faculty of Pharmacy, Minia University, Minia 61519, Egypt;
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Deraya University, New Minia-61768, Egypt
| | - Hossameldin A. Aziz
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, New Vallely University, New Valley 72511, Egypt;
| |
Collapse
|
19
|
Yang Q, Kaw HY, Yu J, Ma X, Yang K, Zhu L, Wang W. Basic Nitrogenous Heterocyclic Rings at the 7-Position of Fluoroquinolones Foster Their Induction of Antibiotic Resistance in Escherichia coli. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2025; 59:6787-6798. [PMID: 40116633 DOI: 10.1021/acs.est.4c11346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/23/2025]
Abstract
The extensive prescription of fluoroquinolone antibiotics has resulted in their ubiquitous presence in the environment, fueling the ongoing development of antibiotic resistance. Besides antibiotics, fluoroquinolone production intermediates, an overlooked category of pollutants that oftentimes possess the intact fluoroquinolone core structure, may also contribute to this public health crisis. To assess their relative potency and collectively examine the structural effects of fluoroquinolones on resistance development, wild-type Escherichia coli K12 was exposed to ten fluoroquinolone antibiotics and five intermediates at their environmentally relevant concentrations for 30 days. Phenotypic resistance alterations revealed that the absence of the C7 ring system in fluoroquinolones significantly impaired their capacity to induce resistance in E. coli, potentially due to diminished oxidative DNA damage and gyrase-mediated dsDNA breaks. Genetic and transcriptional analyses indicated that a uniform resistance mechanism emerged under both antibiotic and intermediate stress. Quantitative structure-activity relationship (QSAR) analysis further emphasized the positive impact of both basic nitrogenous heterocyclic rings at C7 (particularly the hydrogen-bond-donor pharmacophores) and aromatic rings at N1 in promoting resistance development, while highlighting the adverse effects of hydrophobic and hydrogen-bond-donor groups at N1. A robust QSAR model was developed and applied to assess the relative risks of other 105 fluoroquinolones. This study underscored the direct role of fluoroquinolone production intermediates in promoting environmental antibiotic resistance and illustrated how different structural features of fluoroquinolone pollutants will influence this process, offering theoretical insights for future antibiotic design and environmental regulation efforts.
Collapse
Affiliation(s)
- Qi Yang
- State Key Laboratory of Soil Pollution Control and Safety, Zhejiang University, Hangzhou, Zhejiang Province 310058, China
- Department of Environmental Science, Zhejiang University, Hangzhou, Zhejiang Province 310058, China
- Zhejiang Provincial Key Laboratory of Organic Pollution Process and Control, Hangzhou, Zhejiang Province 310058, China
| | - Han Yeong Kaw
- State Key Laboratory of Soil Pollution Control and Safety, Zhejiang University, Hangzhou, Zhejiang Province 310058, China
- Department of Environmental Science, Zhejiang University, Hangzhou, Zhejiang Province 310058, China
- Zhejiang Provincial Key Laboratory of Organic Pollution Process and Control, Hangzhou, Zhejiang Province 310058, China
| | - Jing Yu
- State Key Laboratory of Soil Pollution Control and Safety, Zhejiang University, Hangzhou, Zhejiang Province 310058, China
- Department of Environmental Science, Zhejiang University, Hangzhou, Zhejiang Province 310058, China
- Zhejiang Provincial Key Laboratory of Organic Pollution Process and Control, Hangzhou, Zhejiang Province 310058, China
| | - Xuejing Ma
- State Key Laboratory of Soil Pollution Control and Safety, Zhejiang University, Hangzhou, Zhejiang Province 310058, China
- Department of Environmental Science, Zhejiang University, Hangzhou, Zhejiang Province 310058, China
- Zhejiang Provincial Key Laboratory of Organic Pollution Process and Control, Hangzhou, Zhejiang Province 310058, China
| | - Kun Yang
- State Key Laboratory of Soil Pollution Control and Safety, Zhejiang University, Hangzhou, Zhejiang Province 310058, China
- Department of Environmental Science, Zhejiang University, Hangzhou, Zhejiang Province 310058, China
- Zhejiang Provincial Key Laboratory of Organic Pollution Process and Control, Hangzhou, Zhejiang Province 310058, China
| | - Lizhong Zhu
- State Key Laboratory of Soil Pollution Control and Safety, Zhejiang University, Hangzhou, Zhejiang Province 310058, China
- Department of Environmental Science, Zhejiang University, Hangzhou, Zhejiang Province 310058, China
- Zhejiang Provincial Key Laboratory of Organic Pollution Process and Control, Hangzhou, Zhejiang Province 310058, China
| | - Wei Wang
- State Key Laboratory of Soil Pollution Control and Safety, Zhejiang University, Hangzhou, Zhejiang Province 310058, China
- Department of Environmental Science, Zhejiang University, Hangzhou, Zhejiang Province 310058, China
- Zhejiang Provincial Key Laboratory of Organic Pollution Process and Control, Hangzhou, Zhejiang Province 310058, China
| |
Collapse
|
20
|
Romero N, Kergaravat SV, Regaldo L, Hernández SR, Seabra AB, Ferreira FF, Lourenço IM, Castro GR, Gagneten AM. Multiple physiological response analyses of Chlorella vulgaris exposed to silver nanoparticles, ciprofloxacin, and their combination. ENVIRONMENTAL TOXICOLOGY AND CHEMISTRY 2025; 44:1051-1065. [PMID: 39820261 DOI: 10.1093/etojnl/vgaf013] [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: 08/14/2024] [Revised: 12/11/2024] [Accepted: 12/29/2024] [Indexed: 01/19/2025]
Abstract
The combination of silver nanoparticles (AgNPs) and ciprofloxacin (CIP) can be considered an alternative to combat multidrug-resistant microbial infections. However, knowledge about their combined toxicity after being released in an aquatic environment is scarce. This study evaluated the individual toxicity of AgNPs and CIP and their combined toxicity on the unicellular green microalga Chlorella vulgaris, evaluating cellular responses and conducting metabolomic analysis. The median effect concentrations at 96 h (EC50-96h) for AgNPs, CIP, and the mixture were 132 µg L-1, 7,000 µg L-1, and 452 µg L-1, respectively. Ciprofloxacin exhibited a synergistic effect with AgNPs. The toxic ranking for C. vulgaris was AgNPs > AgNPs + CIP > CIP. The growth rate was the most evident parameter of toxicity. Cell diameter significantly increased (p < 0.001) at 96 h for the highest concentrations tested of AgNPs, CIP, and the mixture, with increases of 24%, 41%, and 19%, respectively, compared with the control. Photosynthetic pigment analyses revealed that C. vulgaris upregulated chlorophyll, carotenoids, and pheophytin. Cell exposure to CIP caused an emergency response involving increased protein and carbohydrate concentrations to tolerate antibiotic stress. Exposure to AgNPs and CIP increased catalase and glutathione S-transferase activity, but the mixture decreased the activity. Silver nanoparticles increased malondialdehyde content in exposed cells due to fatty acid peroxidation. These pollutants revealed their potential risks in interfering with survival and metabolism. Our findings highlight the possible hazards of copollutants at environmentally relevant quantities, providing insights into the individual and combined ecotoxicity of AgNPs and CIP.
Collapse
Affiliation(s)
- Natalí Romero
- Laboratorio de Ecotoxicología, Facultad de Humanidades y Ciencias, Universidad Nacional del Litoral (UNL), Santa Fe, Argentina
- CONICET, CCT Santa Fe, Santa Fe, Argentina
| | - Silvina V Kergaravat
- Laboratorio de Ecotoxicología, Facultad de Humanidades y Ciencias, Universidad Nacional del Litoral (UNL), Santa Fe, Argentina
- CONICET, CCT Santa Fe, Santa Fe, Argentina
- Laboratorio de Sensores y Biosensores, Facultad de Bioquímica y Ciencias Biológicas, UNL-CONICET, CCT Santa Fe, Santa Fe, Argentina
| | - Luciana Regaldo
- Laboratorio de Ecotoxicología, Facultad de Humanidades y Ciencias, Universidad Nacional del Litoral (UNL), Santa Fe, Argentina
- CONICET, CCT Santa Fe, Santa Fe, Argentina
| | - Silvia R Hernández
- Laboratorio de Sensores y Biosensores, Facultad de Bioquímica y Ciencias Biológicas, UNL-CONICET, CCT Santa Fe, Santa Fe, Argentina
| | - Amedea B Seabra
- Center for Natural and Human Sciences (CCNH), Universidade Federal do ABC (UFABC), Santo André, SP, Brazil
| | - Fábio F Ferreira
- Center for Natural and Human Sciences (CCNH), Universidade Federal do ABC (UFABC), Santo André, SP, Brazil
| | - Isabella M Lourenço
- Center for Natural and Human Sciences (CCNH), Universidade Federal do ABC (UFABC), Santo André, SP, Brazil
| | - Guillermo R Castro
- Center for Natural and Human Sciences (CCNH), Universidade Federal do ABC (UFABC), Santo André, SP, Brazil
- Nanomedicine Research Unit (Nanomed), Universidade Federal do ABC (UFABC), Santo André, SP, Brazil
| | - Ana M Gagneten
- Laboratorio de Ecotoxicología, Facultad de Humanidades y Ciencias, Universidad Nacional del Litoral (UNL), Santa Fe, Argentina
- Center for Natural and Human Sciences (CCNH), Universidade Federal do ABC (UFABC), Santo André, SP, Brazil
| |
Collapse
|
21
|
Le Tran TN, Bui NN, Nguyen VT, Tran NA. Novel DNA gyrase mutations in levofloxacin-resistant Helicobacter pylori isolates from southern Vietnam. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2025:S1684-1182(25)00074-X. [PMID: 40164521 DOI: 10.1016/j.jmii.2025.03.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 03/17/2025] [Accepted: 03/25/2025] [Indexed: 04/02/2025]
Abstract
BACKGROUND The escalating antibiotic resistance in Helicobacter pylori (H. pylori) has become a significant concern in the management of infections. This study aimed to investigate the mutations associated with levofloxacin resistance in H. pylori isolated from southern Vietnam. METHODS Levofloxacin-resistant H. pylori strains were isolated from peptic ulcer patients and DNA sequencing of DNA gyrase (gyrA and gyrB) genes was performed. RESULTS We found that 97 % of resistant isolates bearing at least one mutation in GyrA and 32.3 % had a simultaneous mutation in GyrB. Additionally, we identified three previously known mutations, G85C (1.5 %), N87 (34.3 %), and D91 (14.9 %), within the quinolone-resistance-determining region (QRDR) region of GyrA, which confer resistance through natural transformation and were the most prevalent in our study. Notably, we demonstrated a mutation (A88V MIC 4 μg/mL) in GyrA and four novel mutations (E422R MIC 16 μg/mL, A426G MIC 32 μg/mL, S429E MIC 4 μg/mL, and P443A MIC 4 μg/mL) in GyrB, belonging to the QRDR. Additionally, several mutations outside the QRDR were observed, including R190K (16.4 %), P219A/G (13.4 %), L45F (9 %), P220E (6 %), Y28I (4.5 %), A27L (3 %) in GyrA, and S457A (7.5 %), A382F (5.9 %), L389F (4.5 %), D412R (1.5 %), L460P (1.5 %), I482V (1.5 %), and L518E (1.5 %) that have not been documented in any previous studies and were not functionally validated. CONCLUSION These results highlight the high genetic diversity and prevalence of levofloxacin-resistant H. pylori strains in southern Vietnam and provide valuable insights into levofloxacin resistance mechanisms for future diagnostic and treatment advancements.
Collapse
Affiliation(s)
- Thi Nhu Le Tran
- Department of Microbiology, Can Tho University of Medicine and Pharmacy, Viet Nam; Department of Microbiology, Hanoi Medical University, Hanoi, Viet Nam
| | - Ngoc-Niem Bui
- Department of Microbiology, Can Tho University of Medicine and Pharmacy, Viet Nam; Graduate Institute of Biomedical Sciences, Department of Microbiology and Immunology, Chang Gung University, Taoyuan, Taiwan; Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung, Taiwan.
| | - Vu Trung Nguyen
- Department of Microbiology, Hanoi Medical University, Hanoi, Viet Nam; Pasteur Institute in Ho Chi Minh, Ho Chi Minh, Viet Nam; Administration of Science, Technology and Training, Ministry of Health, Hanoi, Viet Nam.
| | - Ngoc Anh Tran
- Department of General Internal Medicine, Hanoi Medical University, Hanoi, Viet Nam; Department of Gastroenterology, Hanoi Medical University Hospital, Hanoi, Viet Nam
| |
Collapse
|
22
|
Baraka K, Abozahra R, Okda F, Abdelhamid SM. Chlorpromazine inhibits the plasmid-mediated oqxAB multidrug efflux pump in Escherichia coli isolates of Egyptian patients with utis. BMC Microbiol 2025; 25:171. [PMID: 40140979 PMCID: PMC11938646 DOI: 10.1186/s12866-025-03850-7] [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: 08/08/2024] [Accepted: 02/26/2025] [Indexed: 03/28/2025] Open
Abstract
Over the past ten years, the prevalence of the OqxAB efflux pump, a plasmid-mediated quinolone resistance determinant, has increased among Escherichia coli (E. coli) isolates. The aim of this study was to genotypically and phenotypically investigate quinolone resistance of isolates and transferability of oqxAB genes by conjugation. One hundred E. coli isolates were collected from UTIs samples and identified using biochemical techniques and confirmed by VITEK-2 System. Antibiotic resistance of isolates was determined by disc diffusion method. MIC of levofloxacin was determined using the broth microdilution method. Efflux pump-mediated resistance was assessed using the chlorpromazine-based efflux-pump inhibitor microplate assay. PMQR genes (oqxA, oqxB) were detected by conventional PCR. A conjugation experiment was run to investigate the transferability of the quinolone resistance in having plasmids carrying oqxAB. DNA sequencing was performed for the identification of genes in both donors and tranconjugants. 80% of E. coli isolates were resistant for levofloxacin. Chlorpromazine significantly decreased the levofloxacin MIC values. oqxA and oqxB genes were detected in 44% and 39% of levofloxacin resistant isolates, respectively. The conjugation experiment revealed the transfer of resistance. MICs of levofloxacin in transconjugants carrying oqxAB significantly increased as compared to the parental recipients MICs. In conclusion, plasmid-mediated quinolone resistance linked to oqxAB may be a factor in rapid rise in and spread of bacterial quinolone resistance among Egyptian E. coli isolates. Chlorpromazine could inhibit efflux pump activity leading to decreased quinolones resistance improving their effectiveness in treatment infectious diseases.
Collapse
Affiliation(s)
- Kholoud Baraka
- Microbiology and Immunology Department, Faculty of Pharmacy, Damanhour University, El Gomhoreya Street, El Behira, Egypt.
| | - Rania Abozahra
- Microbiology and Immunology Department, Faculty of Pharmacy, Damanhour University, El Gomhoreya Street, El Behira, Egypt
| | - Fatma Okda
- Microbiology and Immunology Department, Faculty of Pharmacy, Damanhour University, El Gomhoreya Street, El Behira, Egypt
| | - Sarah M Abdelhamid
- Microbiology and Immunology Department, Faculty of Pharmacy, Damanhour University, El Gomhoreya Street, El Behira, Egypt
| |
Collapse
|
23
|
Shen J, Cai Z, Zhang C, Feng X, Zhang C, Zhao H, Yin C, Wang B, Yu X, Zhang B. Dual-Mode Quantitative Immunochromatographic Assay for Highly Sensitive On-Site Detection of Ciprofloxacin in Fish Products. Foods 2025; 14:1132. [PMID: 40238245 PMCID: PMC11989092 DOI: 10.3390/foods14071132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2025] [Revised: 03/21/2025] [Accepted: 03/21/2025] [Indexed: 04/18/2025] Open
Abstract
Ciprofloxacin has been extensively utilized in aquaculture due to its remarkable efficacy in preventing and treating bacterial infections in fish animals. However, the widespread application of ciprofloxacin has led to significant residue accumulation, necessitating the development of rapid, sensitive and specific detection methods. In this study, we developed a novel dual-mode quantitative immunochromatographic assay based on a portable reader and a photothermal instrument, enabling on-site ciprofloxacin detection. Under optimized conditions, the portable reader mode (Mode 1) achieved a detection range of 0.1-100.0 ng/L with a limit of detection (LOD) of 0.1 ng/mL. The photothermal instrument mode (Mode 2) achieved a detection range of 0.1-500.0 ng/mL with an LOD of 0.1 ng/mL. The sensitivity and accuracy of the method were validated using an Enzyme-Linked Immunosorbent Assay. This developed method successfully detected ciprofloxacin residues in samples of Parabramis pekinensis, Larimichthys crocea, Channa argus, Carassius auratus and Micropterus salmoides, with satisfactory recovery rates. The results demonstrated excellent specificity and applicability across various fish product matrices, offering a reliable and efficient solution for the on-site monitoring of ciprofloxacin residues in fish products.
Collapse
Affiliation(s)
- Junqi Shen
- Key Laboratory of Microbiological Metrology, Measurement & Bio-Product Quality Security, State Administration for Market Regulation, College of Life Sciences, China Jiliang University, Hangzhou 310018, China; (J.S.); (Z.C.); (C.Z.); (X.F.); (C.Z.); (H.Z.); (X.Y.)
| | - Zhengyi Cai
- Key Laboratory of Microbiological Metrology, Measurement & Bio-Product Quality Security, State Administration for Market Regulation, College of Life Sciences, China Jiliang University, Hangzhou 310018, China; (J.S.); (Z.C.); (C.Z.); (X.F.); (C.Z.); (H.Z.); (X.Y.)
| | - Cheng Zhang
- Key Laboratory of Microbiological Metrology, Measurement & Bio-Product Quality Security, State Administration for Market Regulation, College of Life Sciences, China Jiliang University, Hangzhou 310018, China; (J.S.); (Z.C.); (C.Z.); (X.F.); (C.Z.); (H.Z.); (X.Y.)
| | - Xinyue Feng
- Key Laboratory of Microbiological Metrology, Measurement & Bio-Product Quality Security, State Administration for Market Regulation, College of Life Sciences, China Jiliang University, Hangzhou 310018, China; (J.S.); (Z.C.); (C.Z.); (X.F.); (C.Z.); (H.Z.); (X.Y.)
| | - Chenzhi Zhang
- Key Laboratory of Microbiological Metrology, Measurement & Bio-Product Quality Security, State Administration for Market Regulation, College of Life Sciences, China Jiliang University, Hangzhou 310018, China; (J.S.); (Z.C.); (C.Z.); (X.F.); (C.Z.); (H.Z.); (X.Y.)
| | - Huan Zhao
- Key Laboratory of Microbiological Metrology, Measurement & Bio-Product Quality Security, State Administration for Market Regulation, College of Life Sciences, China Jiliang University, Hangzhou 310018, China; (J.S.); (Z.C.); (C.Z.); (X.F.); (C.Z.); (H.Z.); (X.Y.)
| | - Chuanlin Yin
- Key Laboratory of Microbiological Metrology, Measurement & Bio-Product Quality Security, State Administration for Market Regulation, College of Life Sciences, China Jiliang University, Hangzhou 310018, China; (J.S.); (Z.C.); (C.Z.); (X.F.); (C.Z.); (H.Z.); (X.Y.)
- College of Life Sciences, China Jiliang University, Hangzhou 310018, China
| | - Bo Wang
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225009, China;
| | - Xiaoping Yu
- Key Laboratory of Microbiological Metrology, Measurement & Bio-Product Quality Security, State Administration for Market Regulation, College of Life Sciences, China Jiliang University, Hangzhou 310018, China; (J.S.); (Z.C.); (C.Z.); (X.F.); (C.Z.); (H.Z.); (X.Y.)
| | - Biao Zhang
- Key Laboratory of Microbiological Metrology, Measurement & Bio-Product Quality Security, State Administration for Market Regulation, College of Life Sciences, China Jiliang University, Hangzhou 310018, China; (J.S.); (Z.C.); (C.Z.); (X.F.); (C.Z.); (H.Z.); (X.Y.)
| |
Collapse
|
24
|
Kos D, Jelinski M, Ruzzini A. Retrospective analysis of antimicrobial resistance associated with bovine respiratory disease. Appl Environ Microbiol 2025; 91:e0190924. [PMID: 39918326 PMCID: PMC11921372 DOI: 10.1128/aem.01909-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 01/19/2025] [Indexed: 03/20/2025] Open
Abstract
The administration and utility of antibiotics to control and treat bovine respiratory disease (BRD) in beef cattle feedlots is a growing concern. Antimicrobial resistance (AMR) among BRD-associated bacterial pathogens has been the subject of cultivation-dependent and cultivation-independent surveillance. Bacterial genome sequencing and metagenomic approaches facilitate the characterization of AMR in the beef industry; however, the current collection of cattle-associated AMR research programs lack connections to each other. A more integrated view of how antimicrobial use (AMU) is related to resistance at a gene level is needed. We sought to establish a catalog of commonly observed AMR genes (ARGs) in opportunistic bacterial pathogens that contribute to BRD using publicly available data sets that were generated by the scientific community with and without AMU in mind. The presence of these clinically relevant ARGs appeared to differ by geography. Greater sampling in North America facilitated the generation of a list of ARGs often encoded by Mannheimia haemolytica and Pasteurella multocida. Detection of clinically relevant ARGs in shotgun metagenomes of cattle-associated and accessible feedlot samples such as water, soil, and feces was possible but limited by relative sequence read abundance. An exception was the tylosin esterase-encoding gene estT, which is among the most frequently observed ARGs in M. haemolytica and feedlot-related metagenomic data sets. Finally, by re-evaluating studies on the impact of AMU on AMR in beef production systems, we show that conventional practices, including in-feed antibiotic use, increase the relative abundance of ARGs in animal-derived samples.IMPORTANCEThis retrospective analysis delivers a list of ARGs found in opportunistic pathogens that contribute to BRD. The high incidence of BRD in North America is linked to the origin and implementation of metaphylaxis to mitigate detrimental animal losses at feedlots. Notably, ARGs commonly observed in these pathogens isolated in North America were not conserved across the globe, underscoring the relationship between regional AMU and AMR. A positive relationship was also observed between the relative abundance of ARGs in cattle-associated metagenomes with greater exposure to antibiotics. Overall, this analysis should help to guide future surveillance efforts and experimental designs to more directly evaluate the impacts of feedlot practices on AMR.
Collapse
Affiliation(s)
- Daniel Kos
- Department of Large Animal Clinical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Murray Jelinski
- Department of Large Animal Clinical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Antonio Ruzzini
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Department of Biochemistry, Microbiology and Immunology, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
25
|
Shi Q, Mao D, Zhang Z, Qudsi AI, Wei M, Cheng Z, Zhang Y, Wang Z, Chen K, Xu X, Lu X, Liang Q. Epidemiological and Antimicrobial Resistance Trends in Bacterial Keratitis: A Hospital-Based 10-Year Study (2014-2024). Microorganisms 2025; 13:670. [PMID: 40142562 PMCID: PMC11945485 DOI: 10.3390/microorganisms13030670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 03/05/2025] [Accepted: 03/13/2025] [Indexed: 03/28/2025] Open
Abstract
Bacterial keratitis (BK) is a severe ocular infection that can lead to vision loss, with antimicrobial resistance (AMR) posing a growing challenge. This study retrospectively analyzed 1071 bacterial isolates from corneal infections over a 10-year period (2014-2024) at a tertiary ophthalmic center in Beijing, categorizing them into three distinct phases: pre-COVID-19, during COVID-19, and post-COVID-19. The results indicated significant changes in pathogen distribution, including a marked decrease in Gram-positive cocci (from 69.8% pre-COVID-19 to 49.3% in post-COVID-19, p < 0.001), particularly in Staphylococcus epidermidis. In contrast, Gram-positive bacilli, particularly Corynebacterium spp., increased from 4.2% to 16.1% (p < 0.001). The susceptibility to gatifloxacin, moxifloxacin, and ciprofloxacin significantly declined in both Gram-positive cocci and bacilli during the COVID-19 period (all p < 0.01). Gatifloxacin resistance in Staphylococcus rose from pre-COVID-19 (15.2%) to COVID-19 (32.7%), remaining high post-COVID-19 (29.7%). A similar trend was observed in Streptococcus and Corynebacterium, where resistance rose sharply from 12.0% and 22.2% pre-COVID-19 to 42.9% during COVID-19, and remained elevated at 40.0% and 46.4% post-COVID-19, respectively (p < 0.01). These findings emphasize the rapid rise of fluoroquinolone resistance in several bacterial groups, underscoring the urgent need for continuous surveillance and improved antimicrobial stewardship to enhance treatment outcomes.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Qingfeng Liang
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing 100005, China; (Q.S.); (D.M.); (Z.Z.); (A.I.Q.); (M.W.); (Z.C.); (Y.Z.); (Z.W.); (K.C.); (X.X.); (X.L.)
| |
Collapse
|
26
|
Rocha GR, Lemos FFB, Silva LGDO, Luz MS, Correa Santos GL, Rocha Pinheiro SL, Calmon MS, de Melo FF. Overcoming antibiotic-resistant Helicobacter pylori infection: Current challenges and emerging approaches. World J Gastroenterol 2025; 31:102289. [PMID: 40093672 PMCID: PMC11886534 DOI: 10.3748/wjg.v31.i10.102289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 11/28/2024] [Accepted: 01/17/2025] [Indexed: 02/26/2025] Open
Abstract
Recent studies have shown a noticeable increase in global Helicobacter pylori (H. pylori) resistance, with clarithromycin resistance surpassing 15% in various areas. However, inadequate epidemiological monitoring, especially in developing countries, and the absence of uniform testing methods lead to discrepancies between regions and a possible underestimation of resistance levels. The complexity of treating H. pylori is driven by its highly dynamic genome, which is prone to frequent mutations contributing to phenotypical resistance. The usual course of action in empirical treatment involves using a combination of various drugs simultaneously, leading to significant resistance selection pressure and potential side effects. The emergence of H. pylori strains resistant to multiple drugs is closely tied to failures in first-line treatment, highlighting the need to prevent further resistance by using optimal initial empirical therapy or regimens guided by antibiotic susceptibility testing, requiring a collection of mixed samples and multiple isolates for accurate assessment. The emergence of new treatments like potassium-competitive acid blockers offers a hopeful approach to decrease antimicrobial usage while still ensuring effectiveness in comparison to traditional therapies with proton pump inhibitors. Additionally, the use of probiotics is under investigation to identify specific strains and formulations that may mitigate therapy-associated adverse effects.
Collapse
Affiliation(s)
- Gabriel Reis Rocha
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Fabian Fellipe Bueno Lemos
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | | | - Marcel Silva Luz
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Gabriel Lima Correa Santos
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Samuel Luca Rocha Pinheiro
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Mariana Santos Calmon
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Fabrício Freire de Melo
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| |
Collapse
|
27
|
Oh H, Choi Y, Lee J. Antibiotic-Resistant Salmonella in Animal Products Jeopardize Human Health. Food Sci Anim Resour 2025; 45:409-428. [PMID: 40093628 PMCID: PMC11907419 DOI: 10.5851/kosfa.2025.e4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 01/21/2025] [Accepted: 01/22/2025] [Indexed: 03/19/2025] Open
Abstract
Despite the significance of antibiotics in treating bacterial infections, antibiotic resistance is continuously increasing, thus posing a significant threat. In addition to strains resistant to individual drugs, multidrug-resistant (MDR) and pandrug-resistant strains, are emerging. Salmonella, a primary cause of global foodborne illness, is often transmitted through animal products. Antibiotic treatment is crucial for immunocompromised individuals, such as older adults and patients with weakened immune systems, due to their increased susceptibility to severe effects. MDR Salmonella, which can arise following antibiotic use in food animals, may transfer to humans, leading to significant health challenges. The emergence of Salmonella strains resistant to carbapenems, often considered a last-resort antibiotic class, is particularly concerning. Salmonella neutralizes antibiotics through mechanisms, such as horizontal gene transfer via plasmids, efflux/influx system regulation, and enzyme production that deactivate or alter antibiotics. The rise of megaplasmids in Salmonella is particularly alarming, as it may enable resistance to a broader range of antibiotics. This review summarizes the current state of the growing threat of MDR Salmonella and underscores the urgent need for a coordinated response.
Collapse
Affiliation(s)
- Hyemin Oh
- Risk Analysis Research Center, Sookmyung Women’s University, Seoul 04310, Korea
- Department of Food and Nutrition, Sookmyung Women’s University, Seoul 04310, Korea
| | - Yukyung Choi
- Chong Kun Dang Bio Research Institute, Ansan 15604, Korea
| | - Jeeyeon Lee
- Department of Food & Nutrition, Dong-eui University, Busan 47340, Korea
| |
Collapse
|
28
|
Robert M, Ruffier d'Epenoux L, Paquin A, Boutoille D, Guillouzouic A, Corvec S. Ciprofloxacin-susceptible but levofloxacin-resistant Pseudomonas aeruginosa clinical strains with Vitek ®2: which mechanism involved and consequences in case of fluoroquinolone treatment? Eur J Clin Microbiol Infect Dis 2025; 44:549-558. [PMID: 39704919 DOI: 10.1007/s10096-024-05006-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 11/27/2024] [Indexed: 12/21/2024]
Abstract
PURPOSE Pseudomonas aeruginosa clinical strains isolated harbored sometimes an atypical phenotype using the automated Vitek2®: ciprofloxacin-susceptibility but levofloxacin-resistance according to 2019 CA-SFM criteria. The aims of this study are to investigate the resistance mechanism(s) involved and to identify the consequences on fluoroquinolone treatment. METHODS Strain resistance profile, patient's data were recovered and reviewed from the database. Minimum inhibitory concentrations of levofloxacin, ciprofloxacin, moxifloxacin and delafloxacin were determined by using a concentration gradient strip. gyrA, gyrB, parC, parE and mexR genes were PCR amplified and sequenced. A PFGE analysis was performed for strains, recovered in a short period of time from the same patient. RESULTS 46 strains were studied. A couple of seldom mutations were detected in gyrA, gyrB, parC and parE genes. Phenotypically, most of the strains (91%) were resistant to ticarcillin/ clavulanic acid combination and aztreonam suggesting a MexAB-OprM efflux-pump overexpression. mexR sequencing demonstrated either a deletion, a mutation or a premature stop codon appearance leading to amino acid substitution for 75% of the strains. Interestingly, four patients presented successively a fully fluoroquinolone susceptible isolate, thereafter a ciprofloxacin-susceptible but levofloxacin-resistant isolate (discordant phenotype) and finally a fluoroquinolone-resistant isolate. Molecular typing of these strains highlighted a strong relatedness between those isolates. CONCLUSION The phenotype detected by the automate Vitek2® is linked to a likely efflux-pump overexpression mechanism and not fluoroquinolone-target mutation. Regarding this discordant phenotype, an alert should be provided to clinicians concerning the high risk of selecting a fluoroquinolone-resistant mutant.
Collapse
Affiliation(s)
- Manon Robert
- Service de Bactériologie et des Contrôles Microbiologiques, CHU Nantes, Nantes, France
| | - Louise Ruffier d'Epenoux
- Service de Bactériologie et des Contrôles Microbiologiques, CHU Nantes, Nantes, France
- INSERM, Immunology and New Concepts in ImmunoTherapy, INCIT, UMR 1302, Nantes Université, Nantes, France
| | - Axelle Paquin
- Service de Bactériologie et des Contrôles Microbiologiques, CHU Nantes, Nantes, France
| | - David Boutoille
- Service des Maladies Infectieuses, Hôtel-Dieu, Centre Hospitalier Universitaire, Nantes, France
- Centre d'Investigation Clinique Unité d'Investigation Clinique, Centre Hospitalier Universitaire, Nantes, France
| | - Aurélie Guillouzouic
- Service de Bactériologie et des Contrôles Microbiologiques, CHU Nantes, Nantes, France
| | - Stéphane Corvec
- Service de Bactériologie et des Contrôles Microbiologiques, CHU Nantes, Nantes, France.
- INSERM, Immunology and New Concepts in ImmunoTherapy, INCIT, UMR 1302, Nantes Université, Nantes, France.
| |
Collapse
|
29
|
Hu X, Tao J, Yan L, Hong W, Wang W, Wang L, Li G, Jia W. Molecular epidemiology of Escherichia coli in bloodstream infections from a general hospital in Ningxia, China, 2022-2023. BMC Infect Dis 2025; 25:293. [PMID: 40021996 PMCID: PMC11871600 DOI: 10.1186/s12879-025-10658-3] [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: 09/10/2024] [Accepted: 02/17/2025] [Indexed: 03/03/2025] Open
Abstract
OBJECTIVE To analyse the antibiotic resistance, resistance genes and clonal relationship of Escherichia coli in bloodstream infections in Ningxia from 2022 to 2023. METHODS We retrospectively analyzed the antibiotic susceptibilities of 257 isolates. PCR was used to detect blaTEM, blaSHV, blaCTX-M, qnrS, qnrA, qnrB, oqxA, qepA, gyrA, gyrB, parC, and parE, and the clonal relationship through multilocus sequence typing (MLST). RESULTS One hundred and twenty-nine of 257 patients were male (50.2%). The 257 E. coli isolates were mainly obtained from the Emergency, Hepatobiliary Surgery, and Haematology Departments, accounting for 56.6%, 7.3%, and 6.2%, respectively. There is no significant difference in sex and genes between the two groups over and under 60 years old (P > 0.05), but there is a significant difference in ST between them(P<0.05). The antimicrobial susceptibility testing showed that the 257 isolates had the highest rates of resistance to ampicillin (82.8%), followed by cefazolin (71.6%), and all isolates were susceptible to tigecycline. Based on the antibiotic susceptibility results for ceftriaxone, we tested 126 isolates of E. coli for extended-spectrum beta-lactamase (ESBL) resistance genes. As a result, blaCTX-M was detected in 76 isolates (60.32%), blaSHV in 26 isolates (20.63%), and blaTEM in 38 isolates (30.16%). Based on the ciprofloxacin and levofloxacin antibiotic susceptibility results, we tested for quinolone resistance genes in 148 isolates, revealing 66 isolates of aac(6')-Ib-cr (44.60%), 3 isolates of oqxA (2.02%), 32 isolates of qnrS (21.62%), and 2 isolates of qepA (1.35%). We did not detect qnrA or qnrB. The detection rates of gyrA, gyrB, parC, and parE were 98%, 42.6%, 91.2%, and 87.8%, respectively and the main amino acid mutations were Ser83 to Leu, Asp87 to Asn(75.2%), Leu417 to Ser, Ser418 to Leu (6.3%), Ser80 to Ile (65.2%), and Ser458 to Ala (21.5%), respectively. MLST revealed that the most common sequence types (STs) were ST69 (12.5%), ST131 (8.2%), and ST1193 (7.8%). CONCLUSION In our hospital, E. coli was resistant to most commonly used antibiotics, and cefoperazone/sulbactam, cefotetan, amikacin, and tigecycline were empirically selected for the treatment of bloodstream infections. The predominant ESBL genotype in our hospital was blaCTX-M and the major quinolone resistance gene was aac(6')-Ib-cr. Clonal relationship analysis revealed genetic diversity among the isolates.
Collapse
Affiliation(s)
- Xinxin Hu
- College of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia, 750004, China
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, General Hospital of Ningxia Medical University, Yinchua, China
| | - Jia Tao
- College of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia, 750004, China
- Center of Medical Laboratory, General Hospital of Ningxia Medical University, Yinchuan, China
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, General Hospital of Ningxia Medical University, Yinchua, China
| | - Lixin Yan
- College of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia, 750004, China
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, General Hospital of Ningxia Medical University, Yinchua, China
| | - Wei Hong
- College of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia, 750004, China
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, General Hospital of Ningxia Medical University, Yinchua, China
| | - Wen Wang
- College of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia, 750004, China
- Center of Medical Laboratory, General Hospital of Ningxia Medical University, Yinchuan, China
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, General Hospital of Ningxia Medical University, Yinchua, China
| | - Liru Wang
- College of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia, 750004, China
- Center of Medical Laboratory, General Hospital of Ningxia Medical University, Yinchuan, China
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, General Hospital of Ningxia Medical University, Yinchua, China
| | - Gang Li
- College of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia, 750004, China.
- Center of Medical Laboratory, General Hospital of Ningxia Medical University, Yinchuan, China.
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, General Hospital of Ningxia Medical University, Yinchua, China.
| | - Wei Jia
- College of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia, 750004, China.
- Center of Medical Laboratory, General Hospital of Ningxia Medical University, Yinchuan, China.
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, General Hospital of Ningxia Medical University, Yinchua, China.
| |
Collapse
|
30
|
Yang M, Xu Q, Gu K, Wen R, Zhou C, Zhao Y, Guo B, Xu W, Zhang Y, Li C, Lei C, Wang H. Development of a nanobody-horseradish peroxidase fusion-based competitive ELISA to rapidly and sensitively detect Enrofloxacin residues in animal-derived foods. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2025; 327:125309. [PMID: 39490174 DOI: 10.1016/j.saa.2024.125309] [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: 05/23/2024] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 11/05/2024]
Abstract
The ability to reliably detect enrofloxacin in animal-derived food products has important health implications. In the present study, a nanobody-horseradish peroxidase fusion specific for ENR was generated to enable a sensitive and rapid competitive ELISA suitable for detecting enrofloxacin in samples of milk and animal tissue. An enrofloxacin hapten generated via the glutaraldehyde method was initially used to immunize an adult Bactrian camel as a means of constructing a phage library. Enrofloxacin-specific nanobodies were then selected through three rounds of biopanning, and HRP-fused versions of these nanobodies were then expressed. Lastly, these nanobodies were used to develop a sensitive cELISA for enrofloxacin detection in milk and animal tissues, with the resultant assay exhibiting an IC50 of 37.41 ng/mL and a linear detection range (IC20-IC80) of 10.89 to 244.34 ng/mL. The limit of detection for this cELISA was 6.48 ng/mL, with 4.66 % cross-reactivity with ciprofloxacin, and recovery rates that ranged from84.99 % to 107.72 % together with an RSD below 10.70 %.
Collapse
Affiliation(s)
- Ming Yang
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, Sichuan, China
| | - Qiang Xu
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, Sichuan, China
| | - Kui Gu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Renqiao Wen
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, Sichuan, China
| | - Changyu Zhou
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, Sichuan, China
| | - Yu Zhao
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, Sichuan, China
| | - Boyan Guo
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, Sichuan, China
| | - Wei Xu
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, Sichuan, China
| | - Yuanyuan Zhang
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, Sichuan, China
| | - Cui Li
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, Sichuan, China
| | - Changwei Lei
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, Sichuan, China.
| | - Hongning Wang
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
31
|
An JU, Lee J, Cho S, Song H. Comparative Genomic Analysis of Livestock-Derived Campylobacter jejuni: Antimicrobial Resistance, Virulence, Mobile Genetic Elements, and Genetic Relatedness. J Microbiol Biotechnol 2025; 35:e2411044. [PMID: 39947677 PMCID: PMC11876010 DOI: 10.4014/jmb.2411.11044] [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: 11/16/2024] [Revised: 12/06/2024] [Accepted: 12/11/2024] [Indexed: 03/06/2025]
Abstract
Campylobacter jejuni is a major cause of foodborne illnesses, and its increasing antimicrobial resistance (AMR) poses serious public health risks. Owing to their high genetic diversity and frequent intraspecific recombination, understanding the virulence traits of Campylobacter remains challenging. We elucidated the resistance and virulence mechanisms of C. jejuni in livestock using comparative genomic and phenotypic analyses. We analyzed C. jejuni strains isolated from chicken meat, chicken slaughterhouses, and dairy cattle farms. High resistance rates were observed for nalidixic acid, ciprofloxacin, and tetracycline. The chicken-derived strains showed significantly higher tetracycline resistance and marginally higher nalidixic acid resistance, whereas the cattle-derived strains showed marginally higher ciprofloxacin resistance. The key AMR determinants included gyrA and tet(O), which were correlated with resistance phenotypes. Ten virulence factor families were identified with prevalences exceeding 90%. Biofilm formation was observed in 31.9% of strains and correlated with flagella-associated virulence factors. Eighteen plasmid types were detected, primarily in the pTet family, which carried various AMR genes and components of the Type IV secretion system, potentially facilitating the co-transfer of resistance and virulence traits. Conjugation experiments confirmed the horizontal transfer of two pTet plasmid types into the wild-type C. jejuni strain. Further, our analyses revealed over 95% genetic similarity with European C. jejuni strains in a public database-supporting the hypothesis of zoonotic transmission via global food chains-and the zoonotic risks of livestock-derived Campylobacter jejuni. These findings emphasize the need for extended global surveillance to mitigate the risk of zoonotic transmission.
Collapse
Affiliation(s)
- Jae-Uk An
- College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Republic of Korea
| | - Junbum Lee
- College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Republic of Korea
| | - Seongbeom Cho
- College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Republic of Korea
| | - Hyokeun Song
- College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
32
|
Khafagy ES, Soliman GA, Abdel-Kader MS, Bendary MM, Hegazy WAH, Askoura M. Enoxacin adversely affects Salmonella enterica virulence and host pathogenesis through interference with type III secretion system type II (T3SS-II) and disruption of translocation of Salmonella Pathogenicity Island-2 (SPI2) effectors. J Microbiol 2025; 63:e2410015. [PMID: 40044131 DOI: 10.71150/jm.2410015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 01/06/2025] [Indexed: 05/13/2025]
Abstract
Salmonella enterica is a clinically significant oro-fecal pathogen that causes a wide variety of illnesses and can lead to epidemics. S. enterica expresses a lot of virulence factors that enhance its pathogenesis in host. For instance, S. enterica employs a type three secretion system (T3SS) to translocate a wide array of effector proteins that could change the surrounding niche ensuring suitable conditions for the thrive of Salmonella infection. Many antimicrobials have been recently introduced to overcome the annoying bacterial resistance to antibiotics. Enoxacin is member of the second-generation quinolones that possesses a considerable activity against S. enterica. The present study aimed to evaluate the effect of enoxacin at sub-minimum inhibitory concentration (sub-MIC) on S. enterica virulence capability and pathogenesis in host. Enoxacin at sub-MIC significantly diminished both Salmonella invasion and intracellular replication within the host cells. The observed inhibitory effect of enoxacin on S. enterica internalization could be attributed to its ability to interfere with translocation of the T3SS effector proteins. These results were further confirmed by the finding that enoxacin at sub-MIC down-regulated the expression of the genes encoding for T3SS-type II (T3SS-II). Moreover, enoxacin at sub-MIC lessened bacterial adhesion to abiotic surface and biofilm formation which indicates a potential anti-virulence activity. Importantly, in vivo results showed a significant ability of enoxacin to protect mice against S. enterica infection and decreased bacterial colonization within animal tissues. In nutshell, current findings shed light on an additional mechanism of enoxacin at sub-MIC by interfering with Salmonella intracellular replication. The outcomes presented herein could be further invested in conquering bacterial resistance and open the door for additional effective clinical applications.
Collapse
Affiliation(s)
- El-Sayed Khafagy
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam bin Abdulaziz University, Al-kharj 11942, Saudi Arabia
| | - Gamal A Soliman
- Department of Pharmacology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Maged S Abdel-Kader
- Department of Pharmacology, College of Veterinary Medicine, Cairo University, Giza 12211, Egypt
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Mahmoud M Bendary
- Department of Pharmacognosy, Faculty of Pharmacy, Alexandria University, Alexandria 21215, Egypt
| | - Wael A H Hegazy
- Department of Microbiology and Immunology, Faculty of Pharmacy, Port Said University, Port Said 42511, Egypt
- Department of Microbiology and Immunology, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Momen Askoura
- Department of Microbiology and Immunology, Faculty of Pharmacy, Port Said University, Port Said 42511, Egypt
| |
Collapse
|
33
|
Teichmann L, Pasman R, Luitwieler S, Varriale C, Bengtsson-Palme J, Ter Kuile B. Adaptation of Escherichia coli to ciprofloxacin and enrofloxacin: Differential proteomics of the SOS response and RecA-independent mechanisms. Int J Antimicrob Agents 2025; 65:107420. [PMID: 39742892 DOI: 10.1016/j.ijantimicag.2024.107420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/23/2024] [Accepted: 12/16/2024] [Indexed: 01/04/2025]
Abstract
OBJECTIVE Antibiotic resistance is a growing global healthcare challenge, treatment of bacterial infections with fluoroquinolones being no exception. These antibiotics can induce genetic instability through several mechanisms, one of the most significant being the activation of the SOS response. During exposure to sublethal concentration, this stress response increases mutation rates, accelerating resistance evolution. METHODS To explore the role of the SOS response in fluoroquinolone adaptation, we induced de novo resistance by exposure to step-wise increasing concentrations Escherichia coli wild-type (MG1655) and a ΔrecA mutant strain, which is deficient in SOS activation. Both strains were exposed to stepwise increasing concentrations of ciprofloxacin and enrofloxacin - two fluoroquinolones that differ only by a single methyl group. RESULTS Development of resistance against both fluoroquinolones was severely hampered in the ΔrecA mutant. While these antibiotics are often assumed to elicit similar cellular responses, our data revealed distinct genomic and adaptive differences. Building on these findings, we performed a comparative proteomics analysis to investigate how E. coli adapts to ciprofloxacin and enrofloxacin at the protein level. CONCLUSIONS The results demonstrate that the slight structural variation between ciprofloxacin and enrofloxacin leads to unique proteomic adaptations. These findings suggest that even subtle chemical differences can lead to distinct adaptive trajectories and illustrate the flexibility of cellular stress responses.
Collapse
Affiliation(s)
- Lisa Teichmann
- University of Amsterdam, Swammerdam Institute of Life Sciences, Molecular Biology and Microbial Food Safety, Amsterdam, The Netherlands
| | - Raymond Pasman
- University of Amsterdam, Swammerdam Institute of Life Sciences, Molecular Biology and Microbial Food Safety, Amsterdam, The Netherlands
| | - Sam Luitwieler
- University of Amsterdam, Swammerdam Institute of Life Sciences, Molecular Biology and Microbial Food Safety, Amsterdam, The Netherlands
| | - Chiara Varriale
- University of Amsterdam, Swammerdam Institute of Life Sciences, Molecular Biology and Microbial Food Safety, Amsterdam, The Netherlands
| | - Johan Bengtsson-Palme
- Department of Life Sciences, SciLifeLab, Division of Systems and Synthetic Biology, Chalmers University of Technology, Gothenburg, Sweden; Department of Infectious Diseases, University of Gothenburg, Institute of Biomedicine, Gothenburg, Sweden; Centre for Antibiotic Resistance Research (CARe), Gothenburg, Sweden
| | - Benno Ter Kuile
- University of Amsterdam, Swammerdam Institute of Life Sciences, Molecular Biology and Microbial Food Safety, Amsterdam, The Netherlands.
| |
Collapse
|
34
|
Verbel-Olarte MI, Serna-Galvis EA, Jimenez-Lopez DM, Jojoa-Sierra SD, Porras J, Pulgarin C, Torres-Palma RA. First evidence that antibiotic-resistant bacteria are inactivated by chemical species produced through the solar photosensitization of ciprofloxacin in water. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 963:178442. [PMID: 39827635 DOI: 10.1016/j.scitotenv.2025.178442] [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: 09/07/2024] [Revised: 12/30/2024] [Accepted: 01/07/2025] [Indexed: 01/22/2025]
Abstract
For the first time, using a chemical pollutant (an antibiotic) as a photosensitizer to improve the elimination of a microbiological contaminant of emerging concern (antibiotic-resistant bacteria) is presented. The effect of ciprofloxacin (CIP) on the inactivation of three light-promoted antibiotic-resistant bacteria (ARB) was evaluated. Ciprofloxacin-resistant Escherichia coli, ciprofloxacin-resistant Staphylococcus aureus, and carbapenem-resistant Klebsiella pneumoniae. Firstly, the photosensitizing effect of CIP on E. coli inactivation was studied. Irradiated CIP (1 ppm) induced superoxide anion radical formation (confirmed through EPR analyses), and the combination of these reactive oxygen species (ROS) with ongoing solar radiation exposure enhanced bacterial inactivation. CIP enhanced the disinfection of antibiotic-resistant E. coli (by 1.84 log units at 120 min of irradiation) and improved the inactivation of K. pneumoniae (by 3.48 log units at 135 min)-both Gram-negative bacteria. Conversely, the photo-inactivation of the Gram-positive bacteria S. aureus did not significantly change (just a slight reduction of 0.42 log units at 120 min) by the presence of CIP. Showing the bacterial structure influences the disinfection process. Another critical factor was antibiotic concentration. A high CIP concentration (10 ppm) induced an interfering screen effect, while a low concentration promoted bacteria inactivation via photosensitization (in Gram-negative bacteria). Interestingly, no photosensitizing effect was observed when CIP was replaced by levofloxacin (LEV, another fluoroquinolone antibiotic), indicating a strong dependence on antibiotic structure. Additionally, the effect of the light source on photosensitized inactivation was evaluated, substituting sunlight with UVC irradiation. Under UVC light, CIP worsened ARB photo-inactivation, suggesting disinfection was mainly due to direct light action on microorganisms rather than photosensitization. Finally, the influence of water components on sunlight-photosensitized disinfection was examined using simulated urine and freshwater. The ARB inactivation decreased as matrix complexity increased. Thus, the effectiveness order was Milli-Q water > freshwater > urine.
Collapse
Affiliation(s)
- Martha I Verbel-Olarte
- Grupo de Investigación en Remediación Ambiental y Biocatálisis (GIRAB), Instituto de Química, Facultad de Ciencias Exactas y Naturales, Universidad de Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia
| | - Efraím A Serna-Galvis
- Grupo de Investigación en Remediación Ambiental y Biocatálisis (GIRAB), Instituto de Química, Facultad de Ciencias Exactas y Naturales, Universidad de Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia; Grupo de Catalizadores y Adsorbentes (CATALAD), Instituto de Química, Facultad de Ciencias Exactas y Naturales, Universidad de Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia
| | - David M Jimenez-Lopez
- Grupo de Investigaciones Biomédicas Uniremington, Facultad de Ciencias de la Salud, Corporación Universitaria Remington (Uniremington), Calle 51 No. 51-27, Medellín, Colombia
| | - Sindy D Jojoa-Sierra
- Grupo de Investigación en Remediación Ambiental y Biocatálisis (GIRAB), Instituto de Química, Facultad de Ciencias Exactas y Naturales, Universidad de Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia; Grupo de Investigación en Agua y Salud Ambiental, Departamento de Ingeniería Química y Tecnologías del Medio Ambiente, Universidad de Zaragoza, Calle María de Luna 3, 50018, Zaragoza, España
| | - Jazmín Porras
- Grupo de Investigaciones Biomédicas Uniremington, Facultad de Ciencias de la Salud, Corporación Universitaria Remington (Uniremington), Calle 51 No. 51-27, Medellín, Colombia.
| | - Cesar Pulgarin
- Grupo de Investigación en Remediación Ambiental y Biocatálisis (GIRAB), Instituto de Química, Facultad de Ciencias Exactas y Naturales, Universidad de Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia; Institute of Chemical Science and Engineering, Swiss Federal Institute of Technology (EPFL), Station 6, CH-1015 Lausanne, Switzerland; Colombian Academy of Exact, Physical and Natural Sciences, Carrera 28 A No. 39A-63, Bogotá, Colombia
| | - Ricardo A Torres-Palma
- Grupo de Investigación en Remediación Ambiental y Biocatálisis (GIRAB), Instituto de Química, Facultad de Ciencias Exactas y Naturales, Universidad de Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia.
| |
Collapse
|
35
|
Bush NG, Diez-Santos I, Sankara Krishna P, Clavijo B, Maxwell A. Insights into antibiotic resistance promoted by quinolone exposure. Antimicrob Agents Chemother 2025; 69:e0099724. [PMID: 39589140 PMCID: PMC11784200 DOI: 10.1128/aac.00997-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 10/23/2024] [Indexed: 11/27/2024] Open
Abstract
Quinolone-induced antibiotic resistance (QIAR) refers to the phenomenon by which bacteria exposed to sublethal levels of quinolones acquire resistance to non-quinolone antibiotics. We have explored this in Escherichia coli MG1655 using a variety of compounds and bacteria carrying a quinolone-resistance mutation in gyrase, mutations affecting the SOS response, and mutations in error-prone polymerases. The nature of the antibiotic-resistance mutations was determined by whole-genome sequencing. Exposure to low levels of most quinolones tested led to mutations conferring resistance to chloramphenicol, ampicillin, kanamycin, and tetracycline. The mutations included point mutations and deletions and could mostly be correlated with the resistance phenotype. QIAR depended upon DNA gyrase and involved the SOS response but was not dependent on error-prone polymerases. Only moxifloxacin, among the quinolones tested, did not display a significant QIAR effect. We speculate that the lack of QIAR with moxifloxacin may be attributable to it acting via a different mechanism. In addition to the concerns about antimicrobial resistance to quinolones and other compounds, QIAR presents an additional challenge in relation to the usage of quinolone antibacterials.
Collapse
Affiliation(s)
- Natassja G. Bush
- Department of Biological Chemistry, John Innes Centre, Norwich Research Park, Norwich, United Kingdom
- School of Biological Sciences, University of East Anglia School of Biological Sciences, Norwich, United Kingdom
| | - Isabel Diez-Santos
- Department of Biological Chemistry, John Innes Centre, Norwich Research Park, Norwich, United Kingdom
- School of Biological Sciences, University of East Anglia School of Biological Sciences, Norwich, United Kingdom
| | - Pilla Sankara Krishna
- Department of Biological Chemistry, John Innes Centre, Norwich Research Park, Norwich, United Kingdom
| | - Bernardo Clavijo
- Earlham Institute, Norwich Research Park, Norwich, United Kingdom
| | - Anthony Maxwell
- Department of Biological Chemistry, John Innes Centre, Norwich Research Park, Norwich, United Kingdom
- Department of Molecular Microbiology, John Innes Centre, Norwich Research Park, Norwich, United Kingdom
| |
Collapse
|
36
|
Duman E, Müller-Deubert S, Pattappa G, Stratos I, Sieber SA, Clausen-Schaumann H, Sarafian V, Shukunami C, Rudert M, Docheva D. Fluoroquinolone-Mediated Tendinopathy and Tendon Rupture. Pharmaceuticals (Basel) 2025; 18:184. [PMID: 40005998 PMCID: PMC11858458 DOI: 10.3390/ph18020184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/23/2025] [Accepted: 01/25/2025] [Indexed: 02/27/2025] Open
Abstract
The fluoroquinolone (FQ) class of antibiotics includes the world's most prescribed antibiotics such as ciprofloxacin, levofloxacin, and ofloxacin that are known for their low bacterial resistance. This is despite their potential to trigger severe side effects, such as myopathy, hearing loss, tendinopathy, and tendon rupture. Thus, healthcare organizations around the world have recommended limiting the prescription of FQs. Tendinopathy is a common name for maladies that cause pain and degeneration in the tendon tissue, which can result in tendon rupture. Whilst there are several identified effects of FQ on tendons, the exact molecular mechanisms behind FQ-mediated tendon rupture are unclear. Previous research studies indicated that FQ-mediated tendinopathy and tendon rupture can be induced by changes in gene expression, metabolism, and function of tendon resident cells, thus leading to alterations in the extracellular matrix. Hence, this review begins with an update on FQs, their mode of action, and their known side effects, as well as summary information on tendon tissue structure and cellular content. Next, how FQs affect the tendon tissue and trigger tendinopathy and tendon rupture is explored in detail. Lastly, possible preventative measures and promising areas for future research are also discussed. Specifically, follow-up studies should focus on understanding the FQ-mediated tendon changes in a more complex manner and integrating in vitro with in vivo models. With respect to in vitro systems, the field should move towards three-dimensional models that reflect the cellular diversity found in the tissue.
Collapse
Affiliation(s)
- Ezgi Duman
- Department of Musculoskeletal Tissue Regeneration, Orthopaedic Hospital König-Ludwig-Haus, University of Würzburg, 97070 Würzburg, Germany; (S.M.-D.); (G.P.)
| | - Sigrid Müller-Deubert
- Department of Musculoskeletal Tissue Regeneration, Orthopaedic Hospital König-Ludwig-Haus, University of Würzburg, 97070 Würzburg, Germany; (S.M.-D.); (G.P.)
| | - Girish Pattappa
- Department of Musculoskeletal Tissue Regeneration, Orthopaedic Hospital König-Ludwig-Haus, University of Würzburg, 97070 Würzburg, Germany; (S.M.-D.); (G.P.)
| | - Ioannis Stratos
- Department of Orthopaedics, Orthopaedic Hospital König-Ludwig-Haus, University of Würzburg, 97070 Würzburg, Germany; (I.S.); (M.R.)
| | - Stephan A. Sieber
- Center for Functional Protein Assemblies, Department of Bioscience, TUM School of Natural Sciences, Technical University of Munich, 85748 Garching, Germany;
| | - Hauke Clausen-Schaumann
- Center for Applied Tissue Engineering and Regenerative Medicine (CANTER), University of Applied Sciences, 80335 Munich, Germany;
| | - Victoria Sarafian
- Department of Medical Biology, Medical University of Plovdiv, 4000 Plovdiv, Bulgaria;
- Research Institute, Medical University of Plovdiv, 4000 Plovdiv, Bulgaria
| | - Chisa Shukunami
- Department of Molecular Biology and Biochemistry, Division of Dental Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan;
| | - Maximilian Rudert
- Department of Orthopaedics, Orthopaedic Hospital König-Ludwig-Haus, University of Würzburg, 97070 Würzburg, Germany; (I.S.); (M.R.)
| | - Denitsa Docheva
- Department of Musculoskeletal Tissue Regeneration, Orthopaedic Hospital König-Ludwig-Haus, University of Würzburg, 97070 Würzburg, Germany; (S.M.-D.); (G.P.)
| |
Collapse
|
37
|
Teichmann L, Luitwieler S, Bengtsson-Palme J, Ter Kuile B. Fluoroquinolone-specific resistance trajectories in E. coli and their dependence on the SOS-response. BMC Microbiol 2025; 25:37. [PMID: 39838279 PMCID: PMC11748515 DOI: 10.1186/s12866-025-03771-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 01/13/2025] [Indexed: 01/23/2025] Open
Abstract
BACKGROUND Fluoroquinolones are indispensable antibiotics used in treating bacterial infections in both human and veterinary medicine. However, resistance to these drugs presents a growing challenge. The SOS response, a DNA repair pathway activated by DNA damage, is known to influence resistance development, yet its role in fluoroquinolone resistance is not fully understood. This study aims to unfold the mechanisms of fluoroquinolone resistance by investigating the impact of the SOS response on bacterial adaptation. RESULTS We exposed Escherichia coli to four fluoroquinolones-ciprofloxacin, enrofloxacin, levofloxacin, and moxifloxacin. Using a recA knockout mutant, deficient in the SOS response, as a control, we assessed how the presence or absence of this pathway affects resistance development. Our findings demonstrated that the rate of resistance evolution varied between the different fluoroquinolones. Ciprofloxacin, enrofloxacin, and moxifloxacin exposures led to the most evident reliance on the SOS response for resistance, whereas levofloxacin exposed cultures showed less dependency. Whole genome analysis indicated distinct genetic changes associated with each fluoroquinolone, highlighting potential different pathways and mechanisms involved in resistance. CONCLUSIONS This study shows that the SOS response plays a crucial role in resistance development to certain fluoroquinolones, with varying dependencies per drug. The characteristic impact of fluoroquinolones on resistance mechanisms emphasizes the need to consider the unique properties of each antibiotic in resistance studies and treatment strategies. These findings are essential for improving antibiotic stewardship and developing more effective, tailored interventions to combat resistance.
Collapse
Affiliation(s)
- Lisa Teichmann
- University of Amsterdam, Swammerdam Institute of Life Sciences, Molecular Biology and Microbial Food Safety, Amsterdam, The Netherlands
| | - Sam Luitwieler
- University of Amsterdam, Swammerdam Institute of Life Sciences, Molecular Biology and Microbial Food Safety, Amsterdam, The Netherlands
| | - Johan Bengtsson-Palme
- Department of Life Sciences, SciLifeLab, Division of Systems and Synthetic Biology, Chalmers University of Technology, Gothenburg, Sweden
- Institute of Biomedicine, Department of Infectious Diseases, University of Gothenburg, Gothenburg, Sweden
- Centre for Antibiotic Resistance Research (CARe), Gothenburg, Sweden
| | - Benno Ter Kuile
- University of Amsterdam, Swammerdam Institute of Life Sciences, Molecular Biology and Microbial Food Safety, Amsterdam, The Netherlands.
| |
Collapse
|
38
|
Macesic N, Uhlemann AC, Peleg AY. Multidrug-resistant Gram-negative bacterial infections. Lancet 2025; 405:257-272. [PMID: 39826970 DOI: 10.1016/s0140-6736(24)02081-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/03/2024] [Accepted: 09/18/2024] [Indexed: 01/22/2025]
Abstract
Multidrug-resistant Gram-negative bacterial infections cause significant morbidity and mortality globally. These pathogens easily acquire antimicrobial resistance (AMR), further highlighting their clinical significance. Third-generation cephalosporin-resistant and carbapenem-resistant Enterobacterales (eg, Escherichia coli and Klebsiella spp), multidrug-resistant Pseudomonas aeruginosa, and carbapenem-resistant Acinetobacter baumannii are the most problematic and have been identified as priority pathogens. In response, several new diagnostic technologies aimed at rapidly detecting AMR have been developed, including biochemical, molecular, genomic, and proteomic techniques. The last decade has also seen the licensing of multiple antibiotics that have changed the treatment landscape for these challenging infections.
Collapse
Affiliation(s)
- Nenad Macesic
- Department of Infectious Diseases, The Alfred Hospital and School of Translational Medicine, Monash University, Melbourne, VIC, Australia; Centre to Impact AMR, Monash University, Melbourne, VIC, Australia
| | - Anne-Catrin Uhlemann
- Department of Medicine, Division of Infectious Diseases, Columbia University Irving Medical Center, New York, NY, USA
| | - Anton Y Peleg
- Department of Infectious Diseases, The Alfred Hospital and School of Translational Medicine, Monash University, Melbourne, VIC, Australia; Centre to Impact AMR, Monash University, Melbourne, VIC, Australia; Infection Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia.
| |
Collapse
|
39
|
Chen PY, Ho MW, Lu PL, Tang HJ, Sy CL, Wang JT. Comparative In vitro antibacterial activity of nemonoxacin and other fluoroquinolones in correlation with resistant mechanisms in contemporary methicillin-resistant Staphylococcus aureus blood isolates in Taiwan. Ann Clin Microbiol Antimicrob 2025; 24:5. [PMID: 39825371 PMCID: PMC11742215 DOI: 10.1186/s12941-024-00772-6] [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: 07/07/2024] [Accepted: 12/30/2024] [Indexed: 01/20/2025] Open
Abstract
BACKGROUND Nemonoxacin is a new quinolone with an antibacterial efficacy against methicillin-resistant Staphylococcus aureus (MRSA). Certain sequence types (STs) have been emerging in Taiwan, including fluoroquinolone-resistant ST8/USA300. It's an urgent need to determine nemonoxacin susceptibility against ST8/USA300 and other emerging lineages, if any. Additionally, molecular characterization of nemonoxacin resistance among different lineages has yet to be defined. METHODS Non-duplicated MRSA blood isolates from five hospitals during 2019-2020 were collected and genotyped by pulsed-field gel electrophoresis, and further correlated to their STs. Antimicrobial susceptibility testing for all antibiotics was performing by using Sensititre standard panel, except nemonoxacin by using agar dilution method. Selected isolates with nemonoxacin MICs ≥ 0.5 mg/mL were sequenced for quinolone resistance-determining regions (QRDRs). RESULTS Overall, 915 MRSA isolates belonged to four major lineages, ST8 (34.2%), ST59 (23.5%), ST239 (13.9%), and clonal complex 45 (13.7%). Two-thirds of tested isolates were non-susceptible to moxifloxacin, especially ST8/USA300 and ST239. Of them, proportions of nemonoxacin non-susceptibility by a tentative clinical breakpoint (tCBP) of 1 µg/mL among four major lineages appeared to be different (P = 0.06) and highest in ST239 (22.2%), followed by ST8/USA300 (13.5%). Among 89 isolates sequenced, 44.1% of ST8 and all ST239 isolates had ≥ 3 amino acid substitutions (AAS) in gyrA/parC (group A) or 2 AAS in gyrA/parC with additional AAS in gyrB/parE (group B). Compared to other AAS patterns, isolates in group A had the greatest non-susceptible proportions to nemonoxacin (86.9%; overall/pair-wised comparisons, P < 0.05). CONCLUSIONS Our study confirmed ST8/USA300 MRSA has disseminated in Taiwan. Using a tCBP defined by a higher parenteral daily dosage, nemonoxacin retained potency against moxifloxacin non-susceptible isolates. Patterns of AAS in QRDRs among different lineages may contribute to difference of nemonoxacin susceptibility.
Collapse
Affiliation(s)
- Pao -Yu Chen
- Division of Infectious Diseases, Department of Internal Medicine, National Taiwan University Hospital, 7 Chung-Shan South Road, Taipei, 100, Taiwan
| | - Mao-Wang Ho
- Division of Infectious Diseases, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
- China Medical University, Taichung, Taiwan
| | - Po-Liang Lu
- Division of Infectious Diseases, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- School of Post-Baccalaureate Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Center for Liquid Biopsy and Cohort Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hung-Jen Tang
- Division of Infectious Diseases, Department of Internal Medicine, Chi Mei Medical Center, Tainan, Taiwan
| | - Cheng Len Sy
- Division of Infectious Diseases, Department of Internal Medicine, Kaohsiung Veteran General Hospital, Kaohsiung, Taiwan
| | - Jann-Tay Wang
- Division of Infectious Diseases, Department of Internal Medicine, National Taiwan University Hospital, 7 Chung-Shan South Road, Taipei, 100, Taiwan.
- National Institutes of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Taiwan.
| |
Collapse
|
40
|
Aragaw WW, Gebresilase TT, Negatu DA, Dartois V, Dick T. Multidrug tolerance conferred by loss-of-function mutations in anti-sigma factor RshA of Mycobacterium abscessus. Antimicrob Agents Chemother 2024; 68:e0105124. [PMID: 39470195 PMCID: PMC11619451 DOI: 10.1128/aac.01051-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 09/29/2024] [Indexed: 10/30/2024] Open
Abstract
Low-level drug resistance in noncanonical pathways can constitute steppingstones toward acquisition of high-level on-target resistance mutations in the clinic. To capture these intermediate steps in Mycobacterium abscessus (Mab), we performed classic mutant selection experiments with moxifloxacin at twofold its minimum inhibitory concentration (MIC) on solid medium. We found that low-level resistance emerged reproducibly as loss-of-function mutations in RshA (MAB_3542c), an anti-sigma factor that negatively regulates activity of SigH, which orchestrates a response to oxidative stress in mycobacteria. Since oxidative stress is generated in response to many antibiotics, we went on to show that deletion of rshA confers low to moderate resistance-by measure of MIC-to a dozen agents recommended or evaluated for the treatment of Mab pulmonary infections. Interestingly, this moderate resistance was associated with a wide range of drug tolerance, up to 1,000-fold increased survival of a ΔrshA Mab mutant upon exposure to several β-lactams and DNA gyrase inhibitors. Consistent with the putative involvement of the SigH regulon, we showed that addition of the transcription inhibitor rifabutin (RBT) abrogated the high-tolerance phenotype of ΔrshA to representatives of the β-lactam and DNA gyrase inhibitor classes. In a survey of 10,000 whole Mab genome sequences, we identified several loss-of-function mutations in rshA as well as non-synonymous polymorphisms in two cysteine residues critical for interactions with SigH. Thus, the multidrug multiform resistance phenotype we have uncovered may not only constitute a step toward canonical resistance acquisition during treatment but also contribute directly to treatment failure.
Collapse
Affiliation(s)
- Wassihun Wedajo Aragaw
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, New Jersey, USA
| | - Tewodros T. Gebresilase
- Armauer Hansen Research Institute (AHRI), Addis Ababa, Ethiopia
- Institute of Biotechnology, Addis Ababa University, Addis Ababa, Ethiopia
| | - Dereje A. Negatu
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, New Jersey, USA
- Center for Innovative Drug Development and Therapeutic Trials for Africa (CDT-Africa), Addis Ababa University, Addis Ababa, Ethiopia
| | - Véronique Dartois
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, New Jersey, USA
- Department of Medical Sciences, Hackensack Meridian School of Medicine, Nutley, New Jersey, USA
| | - Thomas Dick
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, New Jersey, USA
- Department of Medical Sciences, Hackensack Meridian School of Medicine, Nutley, New Jersey, USA
- Department of Microbiology and Immunology, Georgetown University, Washington, DC, USA
| |
Collapse
|
41
|
Airola C, Severino A, Spinelli I, Gasbarrini A, Cammarota G, Ianiro G, Ponziani FR. "Pleiotropic" Effects of Antibiotics: New Modulators in Human Diseases. Antibiotics (Basel) 2024; 13:1176. [PMID: 39766566 PMCID: PMC11727521 DOI: 10.3390/antibiotics13121176] [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: 10/23/2024] [Revised: 11/22/2024] [Accepted: 11/29/2024] [Indexed: 01/15/2025] Open
Abstract
Antibiotics, widely used medications that have significantly increased life expectancy, possess a broad range of effects beyond their primary antibacterial activity. While some are recognized as adverse events, others have demonstrated unexpected benefits. These adjunctive effects, which have been defined as "pleiotropic" in the case of other pharmacological classes, include immunomodulatory properties and the modulation of the microbiota. Specifically, macrolides, tetracyclines, and fluoroquinolones have been shown to modulate the immune system in both acute and chronic conditions, including autoimmune disorders (e.g., rheumatoid arthritis, spondyloarthritis) and chronic inflammatory pulmonary diseases (e.g., asthma, chronic obstructive pulmonary disease). Azithromycin, in particular, is recommended for the long-term treatment of chronic inflammatory pulmonary diseases due to its well-established immunomodulatory effects. Furthermore, antibiotics influence the human microbiota. Rifaximin, for example, exerts a eubiotic effect that enhances the balance between the gut microbiota and the host immune cells and epithelial cells. These pleiotropic effects offer new therapeutic opportunities by interacting with human cells, signaling molecules, and bacteria involved in non-infectious diseases like spondyloarthritis and inflammatory bowel diseases. The aim of this review is to explore the pleiotropic potential of antibiotics, from molecular and cellular evidence to their clinical application, in order to optimize their use. Understanding these effects is essential to ensure careful use, particularly in consideration of the threat of antimicrobial resistance.
Collapse
Affiliation(s)
- Carlo Airola
- Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (C.A.); (A.S.); (I.S.); (A.G.); (G.C.); (G.I.)
- Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Andrea Severino
- Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (C.A.); (A.S.); (I.S.); (A.G.); (G.C.); (G.I.)
- Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Irene Spinelli
- Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (C.A.); (A.S.); (I.S.); (A.G.); (G.C.); (G.I.)
- Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Antonio Gasbarrini
- Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (C.A.); (A.S.); (I.S.); (A.G.); (G.C.); (G.I.)
- Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Giovanni Cammarota
- Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (C.A.); (A.S.); (I.S.); (A.G.); (G.C.); (G.I.)
- Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Gianluca Ianiro
- Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (C.A.); (A.S.); (I.S.); (A.G.); (G.C.); (G.I.)
- Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Francesca Romana Ponziani
- Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (C.A.); (A.S.); (I.S.); (A.G.); (G.C.); (G.I.)
- Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy
| |
Collapse
|
42
|
Mandel T, Iulietto MF, Reinik M, Condoleo R. Exploring frameworks for quantitative risk assessment of antimicrobial resistance along the food chain. EFSA J 2024; 22:e221117. [PMID: 39712910 PMCID: PMC11659737 DOI: 10.2903/j.efsa.2024.e221117] [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] [Indexed: 12/24/2024] Open
Abstract
Campylobacter is one of the most reported causes of bacterial gastroenteritis worldwide. Birds are the predominant reservoirs for thermotolerant Campylobacter, therefore consumption of contaminated and undercooked poultry products represents one of the major transmission routes for campylobacteriosis. In addition to foodborne diseases, another relevant public challenge is the silent pandemic of antimicrobial resistance (AMR), impacting also the food chain. The occurrence of antimicrobial-resistant Campylobacter in broiler meat poses a significant threat to public health. In this context, quantitative microbiological risk assessment (QMRA) might support policy-makers in addressing these challenges. Hence, this project aimed to describe the current status of knowledge on occurrence of AMR Campylobacter at a global scale, with focus in estimating the prevalence of fluoroquinolone-resistant Campylobacter isolated from broiler meat at retail level. Following the drafting of a dedicated protocol, a systematic literature review and meta-analysis were conducted. Based on the extracted data, after the determination of the proportion of AMR isolates, the proportions were compared by Campylobacter species, geographical regions, processing step and sampling matrices. Meta-analysis allowed the estimation of the pooled prevalences of resistant isolates by regions and species, combining the findings from different independent studies in a comparable way. In terms of future perspectives, the outcome of this project will support future risk assessment and provide valuable inputs for estimating consumer exposure to AMR Campylobacter via broiler meat consumption. In conclusion, this report will provide a general overview of activities, preliminary results and research performed during the EU-FORA fellowship programme (2023/2024).
Collapse
Affiliation(s)
- Tiina Mandel
- National Centre for Laboratory Research and Risk Assessment (LABRIS)TartuEstonia
| | | | - Mari Reinik
- National Centre for Laboratory Research and Risk Assessment (LABRIS)TartuEstonia
| | - Roberto Condoleo
- Istituto Zooprofilattico Sperimentale del Lazio e della Toscana (IZSLT)RomeItaly
| |
Collapse
|
43
|
Khanna A, Kumar N, Rana R, Jyoti, Sharma A, Muskan, Kaur H, Bedi PMS. Fluoroquinolones tackling antimicrobial resistance: Rational design, mechanistic insights and comparative analysis of norfloxacin vs ciprofloxacin derivatives. Bioorg Chem 2024; 153:107773. [PMID: 39241583 DOI: 10.1016/j.bioorg.2024.107773] [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: 06/21/2024] [Revised: 08/01/2024] [Accepted: 08/28/2024] [Indexed: 09/09/2024]
Abstract
Antimicrobial resistance poses a global health concern and develops a need to discover novel antimicrobial agents or targets to tackle this problem. Fluoroquinolone (FN), a DNA gyrase and topoisomerase IV inhibitor, has helped to conquer antimicrobial resistance as it provides flexibility to researchers to rationally modify its structure to increase potency and efficacy. This review provides insights into the rational modification of FNs, the causes of resistance to FNs, and the mechanism of action of FNs. Herein, we have explored the latest advancements in antimicrobial activities of FN analogues and the effect of various substitutions with a focus on utilizing the FN nucleus to search for novel potential antimicrobial candidates. Moreover, this review also provides a comparative analysis of two widely prescribed FNs that are ciprofloxacin and norfloxacin, explaining their rationale for their design, structure-activity relationships (SAR), causes of resistance, and mechanistic studies. These insights will prove advantageous for new researchers by aiding them in designing novel and effective FN-based compounds to combat antimicrobial resistance.
Collapse
Affiliation(s)
- Aanchal Khanna
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, Punjab 143005, India
| | - Nitish Kumar
- Sri Sai College of Pharmacy, Badhani, Pathankot, Punjab 145001, India.
| | - Rupali Rana
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, Punjab 143005, India
| | - Jyoti
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, Punjab 143005, India
| | - Anchal Sharma
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, Punjab 143005, India
| | - Muskan
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, Punjab 143005, India
| | | | | |
Collapse
|
44
|
Lungu IA, Oancea OL, Rusu A. In Silico Study of the Potential Inhibitory Effects on Escherichia coli DNA Gyrase of Some Hypothetical Fluoroquinolone-Tetracycline Hybrids. Pharmaceuticals (Basel) 2024; 17:1540. [PMID: 39598450 PMCID: PMC11597511 DOI: 10.3390/ph17111540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 11/12/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND/OBJECTIVES Despite the discovery of antibiotics, bacterial infections persist globally, exacerbated by rising antimicrobial resistance that results in millions of cases, increased healthcare costs, and more extended hospital stays. The urgent need for new antibacterial drugs continues as resistance evolves. Fluoroquinolones and tetracyclines are versatile antibiotics that are effective against various bacterial infections. A hybrid antibiotic combines two or more molecules to enhance antimicrobial effectiveness and combat resistance better than monotherapy. Fluoroquinolones are ideal candidates for hybridization due to their potent bactericidal effects, ease of synthesis, and ability to form combinations with other molecules. METHODS This study explored the mechanisms of action for 40 hypothetical fluoroquinolone-tetracycline hybrids, all of which could be obtained using a simple, eco-friendly synthesis method. Their interaction with Escherichia coli DNA Gyrase and similarity to albicidin were evaluated using the FORECASTER platform. RESULTS Hybrids such as Do-Ba, Mi-Fi, and Te-Ba closely resembled albicidin in physicochemical properties and FITTED Scores, while Te-De surpassed it with a better score. Similar to fluoroquinolones, these hybrids likely inhibit DNA synthesis by binding to enzyme-DNA complexes. CONCLUSIONS These hybrids could offer broad-spectrum activity and help mitigate bacterial resistance, though further in vitro and in vivo studies are needed to validate their potential.
Collapse
Affiliation(s)
- Ioana-Andreea Lungu
- Medicine and Pharmacy Doctoral School, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Targu Mures, Romania
| | - Octavia-Laura Oancea
- Organic Chemistry Department, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Targu Mures, Romania;
| | - Aura Rusu
- Pharmaceutical and Therapeutic Chemistry Department, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Targu Mures, Romania;
| |
Collapse
|
45
|
Fang L, Deng Y, Lakshmanan P, Liu W, Tang X, Zou W, Zhang T, Wang X, Xiao R, Zhang J, Chen X, Su X. Selective increase of antibiotic-resistant denitrifiers drives N 2O production in ciprofloxacin-contaminated soils. JOURNAL OF HAZARDOUS MATERIALS 2024; 479:135673. [PMID: 39217949 DOI: 10.1016/j.jhazmat.2024.135673] [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: 04/01/2024] [Revised: 08/02/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Agricultural systems significantly contribute to global N2O emissions, which is intensified by excessive fertilization and antibiotic residues, attracting global concerns. However, the dynamics and pathways of antibiotics-induced soil N2O production coupled with microbial metabolism remain controversial. Here, we explored the pathways of N2O production in agricultural soils exposed to ciprofloxacin (CIP), and revealed the underlying mechanisms of CIP degradation and the associated microbial metabolisms using 15N-isotope labeling and molecular techniques. CIP exposure significantly increases the total soil N2O production rate. This is attributed to an unexpected shift from heterotrophic and autotrophic nitrification to denitrification and an increased abundance of denitrifiers Methylobacillus members under CIP exposure. The most striking strain M. flagellatus KT is further discovered to harbor N2O-producing genes but lacks a N2O-reducing gene, thereby stimulating denitrification-based N2O production. Moreover, this denitrifying strain is probably capable of utilizing the byproducts of CIP as carbon sources, evidenced by genes associated with CIP resistance and degradation. Molecular docking further shows that CIP is well ordered in the catalytic active site of CotA laccase, thus affirming the potential for this strain to degrade CIP. These findings advance the mechanistic insights into N2O production within terrestrial ecosystems coupled with the organic contaminants degradation.
Collapse
Affiliation(s)
- Linfa Fang
- Interdisciplinary Research Center for Agriculture Green Development in Yangtze River Basin, College of Resources and Environment, Southwest University, Chongqing 400715, China
| | - Yue Deng
- Interdisciplinary Research Center for Agriculture Green Development in Yangtze River Basin, College of Resources and Environment, Southwest University, Chongqing 400715, China
| | - Prakash Lakshmanan
- Interdisciplinary Research Center for Agriculture Green Development in Yangtze River Basin, College of Resources and Environment, Southwest University, Chongqing 400715, China; Guangxi Key Laboratory of Sugarcane Genetic Improvement, Sugarcane Research Institute, Guangxi Academy of Agricultural Sciences, Nanning 530007, China; Key Laboratory of Sugarcane Biotechnology and Genetic Improvement (Guangxi), Ministry of Agriculture and Rural Affairs, Sugarcane Research Institute, Guangxi Academy of Agricultural Sciences, Nanning 530007, China; Queensland Alliance for Agriculture and Food Innovation, University of Queensland, St Lucia, QLD 4067, Australia
| | - Weibing Liu
- Interdisciplinary Research Center for Agriculture Green Development in Yangtze River Basin, College of Resources and Environment, Southwest University, Chongqing 400715, China
| | - Xiufeng Tang
- Key Laboratory of Geographic Information Science (Ministry of Education), School of Geographic Sciences, East China Normal University, Shanghai, China
| | - Wenxin Zou
- Interdisciplinary Research Center for Agriculture Green Development in Yangtze River Basin, College of Resources and Environment, Southwest University, Chongqing 400715, China
| | - Tong Zhang
- Interdisciplinary Research Center for Agriculture Green Development in Yangtze River Basin, College of Resources and Environment, Southwest University, Chongqing 400715, China
| | - Xiaozhong Wang
- Interdisciplinary Research Center for Agriculture Green Development in Yangtze River Basin, College of Resources and Environment, Southwest University, Chongqing 400715, China
| | - Ran Xiao
- Interdisciplinary Research Center for Agriculture Green Development in Yangtze River Basin, College of Resources and Environment, Southwest University, Chongqing 400715, China
| | - Jinbo Zhang
- School of Geography, Nanjing Normal University, Nanjing 210023, China; Liebig Centre for Agroecology and Climate Impact Research, Justus Liebig University, Germany
| | - Xinping Chen
- Interdisciplinary Research Center for Agriculture Green Development in Yangtze River Basin, College of Resources and Environment, Southwest University, Chongqing 400715, China; Key Laboratory of Low-carbon Green Agriculture in Southwestern China, Ministry of Agriculture and Rural Affairs, Chongqing 400715, China
| | - Xiaoxuan Su
- Interdisciplinary Research Center for Agriculture Green Development in Yangtze River Basin, College of Resources and Environment, Southwest University, Chongqing 400715, China.
| |
Collapse
|
46
|
Guo J, Sun D, Li K, Dai Q, Geng S, Yang Y, Mo M, Zhu Z, Shao C, Wang W, Song J, Yang C, Zhang H. Metabolic Labeling and Digital Microfluidic Single-Cell Sequencing for Single Bacterial Genotypic-Phenotypic Analysis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2402177. [PMID: 39077951 DOI: 10.1002/smll.202402177] [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: 03/20/2024] [Revised: 06/23/2024] [Indexed: 07/31/2024]
Abstract
Accurate assessment of phenotypic and genotypic characteristics of bacteria can facilitate comprehensive cataloguing of all the resistance factors for better understanding of antibiotic resistance. However, current methods primarily focus on individual phenotypic or genotypic profiles across different colonies. Here, a Digital microfluidic-based automated assay for whole-genome sequencing of single-antibiotic-resistant bacteria is reported, enabling Genotypic and Phenotypic Analysis of antibiotic-resistant strains (Digital-GPA). Digital-GPA can efficiently isolate and sequence antibiotic-resistant bacteria illuminated by fluorescent D-amino acid (FDAA)-labeling, producing high-quality single-cell amplified genomes (SAGs). This enables identifications of both minor and major mutations, pinpointing substrains with distinctive resistance mechanisms. Digital-GPA can directly process clinical samples to detect and sequence resistant pathogens without bacterial culture, subsequently provide genetic profiles of antibiotic susceptibility, promising to expedite the analysis of hard-to-culture or slow-growing bacteria. Overall, Digital-GPA opens a new avenue for antibiotic resistance analysis by providing accurate and comprehensive molecular profiles of antibiotic resistance at single-cell resolution.
Collapse
Affiliation(s)
- Junnan Guo
- Department of Chemical Biology, College of Chemistry and Chemical Engineering, School of Life Sciences, Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), Xiamen University, Xiamen, 361005, China
| | - Di Sun
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Kunjie Li
- Department of Chemical Biology, College of Chemistry and Chemical Engineering, School of Life Sciences, Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), Xiamen University, Xiamen, 361005, China
| | - Qi Dai
- Department of Chemical Biology, College of Chemistry and Chemical Engineering, School of Life Sciences, Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), Xiamen University, Xiamen, 361005, China
| | - Shichen Geng
- Department of Chemical Biology, College of Chemistry and Chemical Engineering, School of Life Sciences, Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), Xiamen University, Xiamen, 361005, China
| | - Yuanyuan Yang
- Department of Chemical Biology, College of Chemistry and Chemical Engineering, School of Life Sciences, Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), Xiamen University, Xiamen, 361005, China
| | - Mengwu Mo
- Department of Chemical Biology, College of Chemistry and Chemical Engineering, School of Life Sciences, Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), Xiamen University, Xiamen, 361005, China
| | - Zhi Zhu
- Department of Chemical Biology, College of Chemistry and Chemical Engineering, School of Life Sciences, Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), Xiamen University, Xiamen, 361005, China
| | - Chen Shao
- Department of Chemical Biology, College of Chemistry and Chemical Engineering, School of Life Sciences, Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), Xiamen University, Xiamen, 361005, China
| | - Wei Wang
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Jia Song
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Chaoyong Yang
- Department of Chemical Biology, College of Chemistry and Chemical Engineering, School of Life Sciences, Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), Xiamen University, Xiamen, 361005, China
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Huimin Zhang
- Department of Chemical Biology, College of Chemistry and Chemical Engineering, School of Life Sciences, Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), Xiamen University, Xiamen, 361005, China
| |
Collapse
|
47
|
Gedeon A, Yab E, Dinut A, Sadowski E, Capton E, Dreneau A, Petit J, Gioia B, Piveteau C, Djaout K, Lecat E, Wehenkel AM, Gubellini F, Mechaly A, Alzari PM, Deprez B, Baulard A, Aubry A, Willand N, Petrella S. Molecular mechanism of a triazole-containing inhibitor of Mycobacterium tuberculosis DNA gyrase. iScience 2024; 27:110967. [PMID: 39429773 PMCID: PMC11489056 DOI: 10.1016/j.isci.2024.110967] [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: 03/15/2024] [Revised: 07/02/2024] [Accepted: 09/12/2024] [Indexed: 10/22/2024] Open
Abstract
Antimicrobial resistance remains a persistent and pressing public health concern. Here, we describe the synthesis of original triazole-containing inhibitors targeting the DNA gyrase, a well-validated drug target for developing new antibiotics. Our compounds demonstrate potent antibacterial activity against various pathogenic bacteria, with notable potency against Mycobacterium tuberculosis (Mtb). Moreover, one hit, compound 10a, named BDM71403, was shown to be more potent in Mtb than the NBTI of reference, gepotidacin. Mechanistic enzymology assays reveal a competitive interaction of BDM71403 with fluoroquinolones within the Mtb gyrase cleavage core. High-resolution cryo-electron microscopy structural analysis provides detailed insights into the ternary complex formed by the Mtb gyrase, double-stranded DNA, and either BDM71403 or gepotidacin, providing a rational framework to understand the superior in vitro efficacy on Mtb. This study highlights the potential of triazole-based scaffolds as promising gyrase inhibitors, offering new avenues for drug development in the fight against antimicrobial resistance.
Collapse
Affiliation(s)
- Antoine Gedeon
- Institut Pasteur, Université Paris Cité, CNRS UMR 3528, Unité de Microbiologie Structurale, 75015 Paris, France
| | - Emilie Yab
- Institut Pasteur, Université Paris Cité, CNRS UMR 3528, Unité de Microbiologie Structurale, 75015 Paris, France
| | - Aurelia Dinut
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177 - Drugs and Molecules for living Systems, 59000 Lille, France
| | - Elodie Sadowski
- Cimi-Paris, INSERM U1135, Sorbonne Université, AP-HP. Sorbonne Université, Laboratoire de Bactériologie-Hygiène, CNR des Mycobactéries et de la Résistance des Mycobactéries aux Antituberculeux, 75005 Paris, France
| | - Estelle Capton
- Cimi-Paris, INSERM U1135, Sorbonne Université, AP-HP. Sorbonne Université, Laboratoire de Bactériologie-Hygiène, CNR des Mycobactéries et de la Résistance des Mycobactéries aux Antituberculeux, 75005 Paris, France
| | - Aurore Dreneau
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177 - Drugs and Molecules for living Systems, 59000 Lille, France
| | - Julienne Petit
- Institut Pasteur, Université Paris Cité, CNRS UMR 3528, Unité de Microbiologie Structurale, 75015 Paris, France
| | - Bruna Gioia
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177 - Drugs and Molecules for living Systems, 59000 Lille, France
| | - Catherine Piveteau
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177 - Drugs and Molecules for living Systems, 59000 Lille, France
| | - Kamel Djaout
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - CIIL - Center for Infection and Immunity of Lille, 59000 Lille, France
| | - Estelle Lecat
- Institut Pasteur, Université Paris Cité, CNRS UMR 3528, Unité de Microbiologie Structurale, 75015 Paris, France
| | - Anne Marie Wehenkel
- Institut Pasteur, Université Paris Cité, CNRS UMR 3528, Unité de Microbiologie Structurale, 75015 Paris, France
- Institut Pasteur, Université Paris Cité, CNRS UMR 3528, Bacterial Cell Cycle Mechanisms Unit, 75015 Paris, France
| | - Francesca Gubellini
- Institut Pasteur, Université Paris Cité, CNRS UMR 3528, Unité de Microbiologie Structurale, 75015 Paris, France
| | - Ariel Mechaly
- Institut Pasteur, Plate-Forme de Cristallographie, CNRS UMR 3528, 75015 Paris, France
| | - Pedro M. Alzari
- Institut Pasteur, Université Paris Cité, CNRS UMR 3528, Unité de Microbiologie Structurale, 75015 Paris, France
| | - Benoît Deprez
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177 - Drugs and Molecules for living Systems, 59000 Lille, France
| | - Alain Baulard
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - CIIL - Center for Infection and Immunity of Lille, 59000 Lille, France
| | - Alexandra Aubry
- Cimi-Paris, INSERM U1135, Sorbonne Université, AP-HP. Sorbonne Université, Laboratoire de Bactériologie-Hygiène, CNR des Mycobactéries et de la Résistance des Mycobactéries aux Antituberculeux, 75005 Paris, France
| | - Nicolas Willand
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177 - Drugs and Molecules for living Systems, 59000 Lille, France
| | - Stéphanie Petrella
- Institut Pasteur, Université Paris Cité, CNRS UMR 3528, Unité de Microbiologie Structurale, 75015 Paris, France
- Institut Pasteur, Université Paris Cité, CNRS UMR 3528, Bacterial Cell Cycle Mechanisms Unit, 75015 Paris, France
| |
Collapse
|
48
|
Ashwath P, Osiecki P, Weiner D, Via LE, Sarathy JP. Role of DNA Double-Strand Break Formation in Gyrase Inhibitor-Mediated Killing of Nonreplicating Persistent Mycobacterium tuberculosis in Caseum. ACS Infect Dis 2024; 10:3631-3639. [PMID: 39315541 PMCID: PMC11474946 DOI: 10.1021/acsinfecdis.4c00499] [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: 06/14/2024] [Revised: 09/04/2024] [Accepted: 09/16/2024] [Indexed: 09/25/2024]
Abstract
Tuberculosis is the leading cause of mortality by infectious agents worldwide. The necrotic debris, known as caseum, which accumulates in the center of pulmonary lesions and cavities is home to nonreplicating drug-tolerant Mycobacterium tuberculosis that presents a significant hurdle to achieving a fast and durable cure. Fluoroquinolones such as moxifloxacin are highly effective at killing this nonreplicating persistent bacterial population and boosting TB lesion sterilization. Fluoroquinolones target bacterial DNA gyrase, which catalyzes the negative supercoiling of DNA and relaxes supercoils ahead of replication forks. In this study, we investigated the potency of several other classes of gyrase inhibitors against M. tuberculosis in different states of replication. In contrast to fluoroquinolones, many other gyrase inhibitors kill only replicating bacterial cultures but produce negligible cidal activity against M. tuberculosis in ex vivo rabbit caseum. We demonstrate that while these inhibitors are capable of inhibiting M. tuberculosis gyrase DNA supercoiling activity, fluoroquinolones are unique in their ability to cleave double-stranded DNA at low micromolar concentrations. We hypothesize that double-strand break formation is an important driver of gyrase inhibitor-mediated bactericidal potency against nonreplicating persistent M. tuberculosis populations in the host. This study provides general insight into the lesion sterilization potential of different gyrase inhibitor classes and informs the development of more effective chemotherapeutic options against persistent mycobacterial infections.
Collapse
Affiliation(s)
- Priyanka Ashwath
- Center
for Discovery and Innovation, Hackensack
Meridian Health, 111 Ideation Way, Nutley, New Jersey 07110, United States
| | - Paulina Osiecki
- Center
for Discovery and Innovation, Hackensack
Meridian Health, 111 Ideation Way, Nutley, New Jersey 07110, United States
| | - Danielle Weiner
- Tuberculosis
Research Section, Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, 33 North Drive, Bethesda, Maryland 20892, United States
- Tuberculosis
Imaging Program (TBIP), Division of Intramural Research, NIAID, NIH, 33 North Drive, Building 33, Bethesda, Maryland 20892, United States
| | - Laura E. Via
- Tuberculosis
Research Section, Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, 33 North Drive, Bethesda, Maryland 20892, United States
- Tuberculosis
Imaging Program (TBIP), Division of Intramural Research, NIAID, NIH, 33 North Drive, Building 33, Bethesda, Maryland 20892, United States
| | - Jansy P. Sarathy
- Center
for Discovery and Innovation, Hackensack
Meridian Health, 111 Ideation Way, Nutley, New Jersey 07110, United States
- Department
of Medical Sciences, Hackensack Meridian
School of Medicine, 123
Metro Blvd, Nutley 07110 New Jersey, United
States
| |
Collapse
|
49
|
Elhassan E, Omolo CA, Gafar MA, Kiruri LW, Ibrahim UH, Ismail EA, Devnarain N, Govender T. Disease-Inspired Design of Biomimetic Tannic Acid-Based Hybrid Nanocarriers for Enhancing the Treatment of Bacterial-Induced Sepsis. Mol Pharm 2024; 21:4924-4946. [PMID: 39214595 DOI: 10.1021/acs.molpharmaceut.4c00048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
This study explored the development of novel biomimetic tannic acid-based hybrid nanocarriers (HNs) for targeted delivery of ciprofloxacin (CIP-loaded TAH-NPs) against bacterial-induced sepsis. The prepared CIP-loaded TAH-NPs exhibited appropriate physicochemical characteristics and demonstrated biocompatibility and nonhemolytic properties. Computational simulations and microscale thermophoresis studies validated the strong binding affinity of tannic acid (TA) and its nanoformulation to human Toll-like receptor 4, surpassing that of the natural substrate lipopolysaccharide (LPS), suggesting a potential competitive inhibition against LPS-induced inflammatory responses. CIP released from TAH-NPs displayed a sustained release profile over 72 h. The in vitro antibacterial activity studies revealed that CIP-loaded TAH-NPs exhibited enhanced antibacterial efficacy and efflux pump inhibitory activity. Specifically, they showed a 3-fold increase in biofilm eradication activity against MRSA and a 2-fold increase against P. aeruginosa compared to bare CIP. Time-killing assays demonstrated complete bacterial clearance within 8 h of treatment with CIP-loaded TAH-NPs. In vitro DPPH scavenging and anti-inflammatory investigations confirmed the ability of the prepared hybrid nanosystem to neutralize reactive oxygen species (ROS) and modulate LPS-induced inflammatory responses. Collectively, these results suggest that CIP-loaded TAH-NPs may serve as an innovative nanocarrier for the effective and targeted delivery of antibiotics against bacterial-induced sepsis.
Collapse
Affiliation(s)
- Eman Elhassan
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag Durban X54001, South Africa
| | - Calvin A Omolo
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag Durban X54001, South Africa
- Department of Pharmaceutics and Pharmacy Practice, School of Pharmacy and Health Sciences, United States International University-Africa, P.O. Box 14634-00800, Nairobi 00800, Kenya
| | - Mohammed Ali Gafar
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag Durban X54001, South Africa
- Department of Pharmaceutics, Faculty of Pharmacy, University of Khartoum, Khartoum 11111, Sudan
| | - Lucy W Kiruri
- Department of Chemistry, Kenyatta University, P.O. Box 43844, Nairobi 00100, Kenya
| | - Usri H Ibrahim
- Discipline of Human Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban 4300, South Africa
| | - Eman A Ismail
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag Durban X54001, South Africa
| | - Nikita Devnarain
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag Durban X54001, South Africa
| | - Thirumala Govender
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag Durban X54001, South Africa
| |
Collapse
|
50
|
Xu N, Zhang H, Dong J, Yang Y, Liu Y, Zhou S, Zhu X, Ai X. A modified withdrawal time estimation and risk assessment of enrofloxacin in grass carp (Ctenopharyngodon idella) after ad libitum medicated feed based on statistical approaches in natural cultured environments. Vet Res Commun 2024; 48:3049-3060. [PMID: 39052188 DOI: 10.1007/s11259-024-10475-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 07/20/2024] [Indexed: 07/27/2024]
Abstract
Enrofloxacin (EF) is a broad-spectrum and highly efficient antibiotic commonly used for treating diseases in aquatic animals. However, its abuse in aquaculture applications often leads to excess residue in tissues of grass carp (Ctenopharyngodon idella). Hence, this study aimed to estimate the withdrawal time (WT) of EF and its metabolite of ciprofloxacin (CF) administered medicated feed in natural culture environments and conduct a risk assessment. Plasma and tissue samples were gathered at appropriate time points and detected by high-performance liquid chromatography. The data homogeneity was evaluated by Bartlett's test and Cochran's test. The linearity of the regressed line was evaluated by visual inspection and F test. Outliers were estimated on a normal probability scale by plotting the standardized residual versus their cumulative frequency distribution. Finally, the WT was calculated to be 51 days in muscle + skin based on the maximum residue limit of 100 µg/kg. After 51 days, the concentration of EF and CF fell below 10 µg/kg. The estimated daily intake was calculated to be 0.009 µg/kg/d. Hazard quotient was computed to be 0.002, which was far below one. These results suggested that calculated WT of EF could ensure the safety of products from grass carp for humans.
Collapse
Affiliation(s)
- Ning Xu
- Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan, China.
- Hu Bei Province Engineering and Technology Research Center of Aquatic Product Quality and Safety, Wuhan, China.
- Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, No. 8 Wuda Park Road 1, Wuhan, 430223, Hubei Province, China.
| | - Huan Zhang
- Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan, China
- College of Food Science and Engineering, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Jing Dong
- Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan, China
- Hu Bei Province Engineering and Technology Research Center of Aquatic Product Quality and Safety, Wuhan, China
| | - Yibin Yang
- Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan, China
- Hu Bei Province Engineering and Technology Research Center of Aquatic Product Quality and Safety, Wuhan, China
| | - Yongtao Liu
- Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan, China
- Hu Bei Province Engineering and Technology Research Center of Aquatic Product Quality and Safety, Wuhan, China
| | - Shun Zhou
- Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan, China
- Hu Bei Province Engineering and Technology Research Center of Aquatic Product Quality and Safety, Wuhan, China
| | - Xia Zhu
- Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan, China
- Hu Bei Province Engineering and Technology Research Center of Aquatic Product Quality and Safety, Wuhan, China
| | - Xiaohui Ai
- Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan, China.
- Hu Bei Province Engineering and Technology Research Center of Aquatic Product Quality and Safety, Wuhan, China.
- Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, No. 8 Wuda Park Road 1, Wuhan, 430223, Hubei Province, China.
| |
Collapse
|