1
|
Aldahan Z, Kim J, Yoon CY, Seo YJ, Park KH. Preliminary Analysis of Drug-Induced Ototoxicity in South Korea: Trends From a National Sample Dataset. J Audiol Otol 2025; 29:110-116. [PMID: 40296472 DOI: 10.7874/jao.2024.00493] [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: 09/25/2024] [Accepted: 11/04/2024] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND AND OBJECTIVES Certain medications are associated with ototoxicity. This study assesses drug-induced ototoxicity in South Korea by analyzing the Korean national health data. SUBJECTS AND METHODS Hospital records of National Health Insurance members from 2009 to 2016 were reviewed. Data were compared between patients with and without hearing loss (HL). Individuals with HL were identified as having a primary diagnosis code for sensorineural HL or another type of HL in at least one outpatient or inpatient record according to the International Classification of Diseases-10. RESULTS The members in the HL group increased slightly from 0.8% to 1.0% relative to the total sample, compared with 99.2% to 99.0% among the controls. The proportion of males in the HL group ranged from 45.6% to 47.6%, compared with 48.4% to 48.8% among the controls. The proportion of those aged ≥65 years in the HL group increased from 34.1% to 41.4%, compared with 10.6% to 13.3% among the controls. Hypertension prevalence (24.7%-25.7%) in the HL group was higher than that in the control group (12%-12.6%). Diabetes prevalence in the HL group was 10.6%-12.3%, compared with 4.4%-5.9% among the controls. The use of proton pump inhibitor components increased, particularly esomeprazole magnesium trihydrate and rabeprazole sodium, whereas the usage of pantoprazole sodium sesquihydrate and revaprazan was high initially but declined subsequently. The usage of painkillers such as acetaminophen, loxoprofen sodium, and ibuprofen remained high, and antibiotics such as cephalosporins indicated the highest usage. However, the use of penicillin antibiotics such as amoxicillin decreased significantly. Anticancer agents showed relatively low usage compared with other drug categories, whereas antihistamines showed extremely high usage across all years, with a continual increase. CONCLUSIONS These correlations and the underlying mechanisms necessitate further investigation, as several medicines have been linked to an increased risk of HL.
Collapse
Affiliation(s)
- Zahra Aldahan
- Department of Otolaryngology-Head & Neck Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jiwon Kim
- Research Institute of Hearing Enhancement, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Chul Young Yoon
- Research Institute of Hearing Enhancement, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Young Joon Seo
- Research Institute of Hearing Enhancement, Yonsei University Wonju College of Medicine, Wonju, Korea
- Department of Otorhinolaryngology, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Kyoung Ho Park
- Department of Otolaryngology-Head & Neck Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
2
|
de Dios R, Gadar K, Proctor CR, Maslova E, Han J, Soliman MAN, Krawiel D, Dunbar EL, Singh B, Peros S, Killelea T, Warnke AL, Haugland MM, Bolt EL, Lentz CS, Rudolph CJ, McCarthy RR. Saccharin disrupts bacterial cell envelope stability and interferes with DNA replication dynamics. EMBO Mol Med 2025:10.1038/s44321-025-00219-1. [PMID: 40169895 DOI: 10.1038/s44321-025-00219-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 03/04/2025] [Accepted: 03/06/2025] [Indexed: 04/03/2025] Open
Abstract
Saccharin has been part of the human diet for over 100 years, and there is a comprehensive body of evidence demonstrating that it can influence the gut microbiome, ultimately impacting human health. However, the precise mechanisms through which saccharin can impact bacteria have remained elusive. In this work, we demonstrate that saccharin inhibits cell division, leading to cell filamentation with altered DNA synthesis dynamics. We show that these effects on the cell are superseded by the formation of bulges emerging from the cell envelope, which ultimately trigger cell lysis. We demonstrate that saccharin can inhibit the growth of both Gram-negative and Gram-positive bacteria as well as disrupt key phenotypes linked to host colonisation, such as motility and biofilm formation. In addition, we test its potential to disrupt established biofilms (single-species as well as polymicrobial) and its capacity to re-sensitise multidrug-resistant pathogens to last-resort antibiotics. Finally, we present in vitro and ex vivo evidence of the versatility of saccharin as a potential antimicrobial by integrating it into an effective hydrogel wound dressing.
Collapse
Affiliation(s)
- Rubén de Dios
- Antimicrobial Innovations Centre, Division of Biosciences, Department of Life Sciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, UB8 3PH, UK
| | - Kavita Gadar
- Antimicrobial Innovations Centre, Division of Biosciences, Department of Life Sciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, UB8 3PH, UK
| | - Chris R Proctor
- Antimicrobial Innovations Centre, Division of Biosciences, Department of Life Sciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, UB8 3PH, UK
| | - Evgenia Maslova
- Antimicrobial Innovations Centre, Division of Biosciences, Department of Life Sciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, UB8 3PH, UK
| | - Jie Han
- Antimicrobial Innovations Centre, Division of Biosciences, Department of Life Sciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, UB8 3PH, UK
| | - Mohamed A N Soliman
- Antimicrobial Innovations Centre, Division of Biosciences, Department of Life Sciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, UB8 3PH, UK
| | - Dominika Krawiel
- Antimicrobial Innovations Centre, Division of Biosciences, Department of Life Sciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, UB8 3PH, UK
| | - Emma L Dunbar
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706-1544, USA
| | - Bhupender Singh
- Research Group for Host-Microbe Interactions, Department of Medical Biology and Centre for New Antibacterial Strategies (CANS), UiT-The Arctic University of Norway, 9019, Tromsø, Norway
| | - Stelinda Peros
- Division of Biosciences, Department of Life Sciences, Centre for Genome Engineering and Maintenance, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, UB8 3PH, UK
| | - Tom Killelea
- School of Life Sciences, Faculty of Medicine & Health Sciences, Queens Medical Centre, University of Nottingham, Nottingham, NG7 2UH, UK
| | - Anna-Luisa Warnke
- Department of Chemistry, UiT-The Arctic University of Norway, 9037, Tromsø, Norway
| | - Marius M Haugland
- Department of Chemistry, UiT-The Arctic University of Norway, 9037, Tromsø, Norway
| | - Edward L Bolt
- School of Life Sciences, Faculty of Medicine & Health Sciences, Queens Medical Centre, University of Nottingham, Nottingham, NG7 2UH, UK
| | - Christian S Lentz
- Research Group for Host-Microbe Interactions, Department of Medical Biology and Centre for New Antibacterial Strategies (CANS), UiT-The Arctic University of Norway, 9019, Tromsø, Norway
| | - Christian J Rudolph
- Division of Biosciences, Department of Life Sciences, Centre for Genome Engineering and Maintenance, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, UB8 3PH, UK
| | - Ronan R McCarthy
- Antimicrobial Innovations Centre, Division of Biosciences, Department of Life Sciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, UB8 3PH, UK.
| |
Collapse
|
3
|
Fruchard L, Babosan A, Carvalho A, Lang M, Li B, Duchateau M, Giai Gianetto Q, Matondo M, Bonhomme F, Hatin I, Arbes H, Fabret C, Corler E, Sanchez G, Marchand V, Motorin Y, Namy O, de Crécy-Lagard V, Mazel D, Baharoglu Z. Aminoglycoside tolerance in Vibrio cholerae engages translational reprogramming associated with queuosine tRNA modification. eLife 2025; 13:RP96317. [PMID: 39761105 DOI: 10.7554/elife.96317] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2025] Open
Abstract
Tgt is the enzyme modifying the guanine (G) in tRNAs with GUN anticodon to queuosine (Q). tgt is required for optimal growth of Vibrio cholerae in the presence of sub-lethal aminoglycoside concentrations. We further explored here the role of the Q34 in the efficiency of codon decoding upon tobramycin exposure. We characterized its impact on the overall bacterial proteome, and elucidated the molecular mechanisms underlying the effects of Q34 modification in antibiotic translational stress response. Using molecular reporters, we showed that Q34 impacts the efficiency of decoding at tyrosine TAT and TAC codons. Proteomics analyses revealed that the anti-SoxR factor RsxA is better translated in the absence of tgt. RsxA displays a codon bias toward tyrosine TAT and overabundance of RsxA leads to decreased expression of genes belonging to SoxR oxidative stress regulon. We also identified conditions that regulate tgt expression. We propose that regulation of Q34 modification in response to environmental cues leads to translational reprogramming of transcripts bearing a biased tyrosine codon usage. In silico analysis further identified candidate genes which could be subject to such translational regulation, among which DNA repair factors. Such transcripts, fitting the definition of modification tunable transcripts, are central in the bacterial response to antibiotics.
Collapse
Affiliation(s)
- Louna Fruchard
- Institut Pasteur, Université Paris Cité, Unité Plasticité du Génome Bactérien, Paris, France
- Sorbonne Université, Collège Doctoral, Paris, France
| | - Anamaria Babosan
- Institut Pasteur, Université Paris Cité, Unité Plasticité du Génome Bactérien, Paris, France
| | - Andre Carvalho
- Institut Pasteur, Université Paris Cité, Unité Plasticité du Génome Bactérien, Paris, France
| | - Manon Lang
- Institut Pasteur, Université Paris Cité, Unité Plasticité du Génome Bactérien, Paris, France
| | - Blaise Li
- Institut Pasteur, Université Paris Cité, Bioinformatics and Biostatistics Hub, Paris, France
| | - Magalie Duchateau
- Institut Pasteur, Université Paris Cité, Proteomics Platform, Mass Spectrometry for Biology Unit, Paris, France
| | - Quentin Giai Gianetto
- Institut Pasteur, Université Paris Cité, Proteomics Platform, Mass Spectrometry for Biology Unit, Paris, France
- Institut Pasteur, Université Paris Cité, Department of Computation Biology, Bioinformatics and Biostatistics Hub, Paris, France
| | - Mariette Matondo
- Institut Pasteur, Université Paris Cité, Proteomics Platform, Mass Spectrometry for Biology Unit, Paris, France
| | - Frederic Bonhomme
- Institut Pasteur, Université Paris cité, Epigenetic Chemical Biology Unit, Paris, France
| | - Isabelle Hatin
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif sur Yvette, France
| | - Hugo Arbes
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif sur Yvette, France
| | - Céline Fabret
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif sur Yvette, France
| | - Enora Corler
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif sur Yvette, France
| | - Guillaume Sanchez
- Université de Lorraine, CNRS, Inserm, UAR2008/US40 IBSLor, Epitranscriptomics and RNA Sequencing Core Facility and UMR7365 IMoPA, Nancy, France
| | - Virginie Marchand
- Université de Lorraine, CNRS, Inserm, UAR2008/US40 IBSLor, Epitranscriptomics and RNA Sequencing Core Facility and UMR7365 IMoPA, Nancy, France
| | - Yuri Motorin
- Université de Lorraine, CNRS, Inserm, UAR2008/US40 IBSLor, Epitranscriptomics and RNA Sequencing Core Facility and UMR7365 IMoPA, Nancy, France
| | - Olivier Namy
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif sur Yvette, France
| | - Valérie de Crécy-Lagard
- Department of Microbiology and Cell Science, University of Florida, Gainesville, United States
- University of Florida Genetics Institute, Gainesville, United States
| | - Didier Mazel
- Institut Pasteur, Université Paris Cité, Unité Plasticité du Génome Bactérien, Paris, France
| | - Zeynep Baharoglu
- Institut Pasteur, Université Paris Cité, Unité Plasticité du Génome Bactérien, Paris, France
| |
Collapse
|
4
|
Hu XH, Li Y, Wu T, Ma J, Wu Y, Wilhelm MJ, Dai HL. Effect of Exogenous Glucose on Molecular Transport through Bacterial Membranes Studied by Second Harmonic Light Scattering. J Phys Chem B 2024; 128:12379-12388. [PMID: 39659191 DOI: 10.1021/acs.jpcb.4c06134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2024]
Abstract
Recent studies revealed that exogenous glucose increases the efficacy of aminoglycosides in eliminating bacterial persister cells. It was speculated that this increased antimicrobial efficacy is induced by glucose-facilitated uptake of the antibiotics. Here, we examine this hypothesis by using second-harmonic light scattering to time-resolve the transport of an antimicrobial quaternary ammonium compound (QAC), malachite green, across the membranes of living Escherichia coli (E. coli) in the presence of glucose. The results show that when the glucose concentration increased from 0 to 100 μM, the QAC transport rate constant across the cytoplasmic membrane of E. coli increased by 3.6 times. Further increase of glucose concentration into the millimolar range, however, does not further enhance the transport rate constant. Conversely, a study of QAC transport across the protein-free membrane of liposomes (constructed from the polar lipid extract of E. coli) indicates that the glucose-induced enhancement in membrane transport in E. coli is mediated by protein transporters. Cell viability experiments show that low concentration of exogenous glucose enhances the QAC efficacy in eliminating E. coli. The loss of viability of the bacterial cells in the presence of the QAC at minimum inhibitory concentration increased dramatically when glucose concentration increased from 0 to 100 μM but increasing the glucose concentration up to 1 mM did not further enhance the antimicrobial efficacy. This behavior coincides with the observed increase in QAC transport rate constant as a function of glucose concentration. These observations reveal a novel antimicrobial strategy in which low concentration glucose enhances the uptake of antimicrobials into the bacteria, thereby improving antimicrobial efficacy.
Collapse
Affiliation(s)
- Xiao-Hua Hu
- Department of Chemistry and Institute for Membranes and Interfaces, Temple University, 1901 North 13th Street, Philadelphia, Pennsylvania 19122, United States
| | - Yujie Li
- Department of Chemistry and Institute for Membranes and Interfaces, Temple University, 1901 North 13th Street, Philadelphia, Pennsylvania 19122, United States
| | - Tong Wu
- Department of Chemistry and Institute for Membranes and Interfaces, Temple University, 1901 North 13th Street, Philadelphia, Pennsylvania 19122, United States
| | - Jianqiang Ma
- Department of Chemistry and Institute for Membranes and Interfaces, Temple University, 1901 North 13th Street, Philadelphia, Pennsylvania 19122, United States
| | - Yuhao Wu
- Department of Chemistry and Institute for Membranes and Interfaces, Temple University, 1901 North 13th Street, Philadelphia, Pennsylvania 19122, United States
| | - Michael J Wilhelm
- Department of Chemistry and Institute for Membranes and Interfaces, Temple University, 1901 North 13th Street, Philadelphia, Pennsylvania 19122, United States
| | - Hai-Lung Dai
- Department of Chemistry and Institute for Membranes and Interfaces, Temple University, 1901 North 13th Street, Philadelphia, Pennsylvania 19122, United States
| |
Collapse
|
5
|
Story S, Arya DP. A Cell-Based Screening Assay for rRNA-Targeted Drug Discovery. ACS Infect Dis 2024; 10:4194-4207. [PMID: 39530678 DOI: 10.1021/acsinfecdis.4c00446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Worldwide, bacterial antibiotic resistance continues to outpace the level of drug development. One way to counteract this threat to society is to identify novel ways to rapidly screen and identify drug candidates in living cells. Developing fluorescent antibiotics that can enter microorganisms and be displaced by potential antimicrobial compounds is an important but challenging endeavor due to the difficulty in entering bacterial cells. We developed a cell-based assay using a fluorescent aminoglycoside molecule that allows for the rapid and direct characterization of aminoglycoside binding in a population of bacterial cells. The assay involves the accumulation and competitive displacement of a fluorescent aminoglycoside binding probe in Escherichia coli as a Gram-negative bacterial model. The assay was optimized for high signal-to-background ratios, ease of performance for reliable outcomes, and amenability to high-throughput screening. We demonstrate that the fluorescent binding probe shows a decrease in fluorescence with cellular uptake, consistent with RNA binding, and also shows a subsequent increase upon the addition of the positive control neomycin. Fluorescence intensity increase with aminoglycosides was indicative of their relative binding affinities for A-site rRNA, with neomycin having the highest affinity, followed by paromomycin, tobramycin, sisomicin, and netilmicin. Intermediate fluorescence was found with plazomicin, neamine, apramycin, ribostamicin, gentamicin, and amikacin. Weak fluorescence was observed with kanamycin, hygromycin, streptomycin, and spectinomycin. A high degree of sensitivity was observed with aminoglycosides known to be strong binders for the 16S rRNA A-site compared with antibiotics that target other biosynthetic pathways. The quality of the optimized assay was excellent for planktonic cells, with an average Z' factor value of 0.80. In contrast to planktonic cells, established biofilms yielded an average Z' factor of 0.61. The high sensitivity of this cell-based assay in a physiological context demonstrates significant potential for identifying potent new ribosomal binding antibiotics.
Collapse
Affiliation(s)
- Sandra Story
- NUBAD, LLC, Greenville, South Carolina 29605, United States
| | - Dev P Arya
- Laboratory of Medicinal Chemistry, Department of Chemistry, Clemson University, Clemson, South Carolina 29634, United States
- NUBAD, LLC, Greenville, South Carolina 29605, United States
| |
Collapse
|
6
|
Best W, Ferrell M, Boris A, Heydarian N, Panlilio H, Rice CV. Acquisition of Resistance to PEGylated Branched Polyethylenimine Increases Pseudomonas Aeruginosa Susceptibility to Aminoglycosides. ChemMedChem 2024; 19:e202300689. [PMID: 38806411 PMCID: PMC11368615 DOI: 10.1002/cmdc.202300689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 05/24/2024] [Accepted: 05/24/2024] [Indexed: 05/30/2024]
Abstract
PEGylated branched polyethylenimine (PEG-BPEI) has antibacterial and antibiofilm properties. Exposure to PEG-BPEI through serial passage leads to resistant P. aeruginosa strains. The minimum inhibitory concentration (MIC) of 600 Da BPEI and PEGylated 600 Da BPEI (PEG-BPEI) in the wild-type PAO1 strain is 16 μg/ml while, after 15 serial passages, the MIC increased to 1024 μg/mL. An additional 15 rounds of serial passage in the absence of BPEI or PEG-BPEI did not change the 1024 μg/mL MIC. Gentamicin, Neomycin, and Tobramycin, cationic antibiotics that inhibit protein synthesis, have a 16-32 fold reduction of MIC values in PEG350-BPEI resistant strains, suggesting increased permeation. The influx of these antibiotics occurs using a self-mediated uptake mechanism, suggesting changes to the outer membrane Data show that resistance causes changes in genes related to outer membrane lipopolysaccharide (LPS) assembly. Mutations were noted in the gene coding for the polymerase Wzy that participates in the assembly of the O-antigen region. Other mutations were noted with wbpE and wbpI of the Wbp pathway responsible for the enzymatic synthesis of ManNAc(3NAc)A in the LPS of P. aeruginosa. These changes suggest that an altered gene product could lead to PEG-BPEI resistance. Nevertheless, the increased susceptibility to aminoglycosides could prevent the emergence of PEG-BPEI resistant bacterial populations.
Collapse
Affiliation(s)
- William Best
- Department of Chemistry and Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, 101 Stephenson Parkway, Norman, OK 73069
| | - Maya Ferrell
- Department of Chemistry and Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, 101 Stephenson Parkway, Norman, OK 73069
| | - Andrew Boris
- Department of Chemistry and Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, 101 Stephenson Parkway, Norman, OK 73069
| | - Neda Heydarian
- Department of Chemistry and Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, 101 Stephenson Parkway, Norman, OK 73069
| | - Hannah Panlilio
- Department of Chemistry and Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, 101 Stephenson Parkway, Norman, OK 73069
| | - Charles V. Rice
- Department of Chemistry and Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, 101 Stephenson Parkway, Norman, OK 73069
| |
Collapse
|
7
|
Sun T, Zhao H, Hu L, Shao X, Lu Z, Wang Y, Ling P, Li Y, Zeng K, Chen Q. Enhanced optical imaging and fluorescent labeling for visualizing drug molecules within living organisms. Acta Pharm Sin B 2024; 14:2428-2446. [PMID: 38828150 PMCID: PMC11143489 DOI: 10.1016/j.apsb.2024.01.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 01/07/2024] [Accepted: 01/25/2024] [Indexed: 06/05/2024] Open
Abstract
The visualization of drugs in living systems has become key techniques in modern therapeutics. Recent advancements in optical imaging technologies and molecular design strategies have revolutionized drug visualization. At the subcellular level, super-resolution microscopy has allowed exploration of the molecular landscape within individual cells and the cellular response to drugs. Moving beyond subcellular imaging, researchers have integrated multiple modes, like optical near-infrared II imaging, to study the complex spatiotemporal interactions between drugs and their surroundings. By combining these visualization approaches, researchers gain supplementary information on physiological parameters, metabolic activity, and tissue composition, leading to a comprehensive understanding of drug behavior. This review focuses on cutting-edge technologies in drug visualization, particularly fluorescence imaging, and the main types of fluorescent molecules used. Additionally, we discuss current challenges and prospects in targeted drug research, emphasizing the importance of multidisciplinary cooperation in advancing drug visualization. With the integration of advanced imaging technology and molecular design, drug visualization has the potential to redefine our understanding of pharmacology, enabling the analysis of drug micro-dynamics in subcellular environments from new perspectives and deepening pharmacological research to the levels of the cell and organelles.
Collapse
Affiliation(s)
- Ting Sun
- School of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery System, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, China
- Institute of Biochemical and Biotechnological Drugs, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Huanxin Zhao
- School of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery System, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, China
| | - Luyao Hu
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xintian Shao
- School of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery System, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, China
- School of Life Sciences, Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, China
| | - Zhiyuan Lu
- School of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery System, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, China
| | - Yuli Wang
- Tianjin Pharmaceutical DA REN TANG Group Corporation Limited Traditional Chinese Pharmacy Research Institute, Tianjin 300457, China
- Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemistry Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Peixue Ling
- Institute of Biochemical and Biotechnological Drugs, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
- Key Laboratory of Biopharmaceuticals, Postdoctoral Scientific Research Workstation, Shandong Academy of Pharmaceutical Science, Jinan 250098, China
| | - Yubo Li
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Kewu Zeng
- School of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery System, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, China
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Qixin Chen
- School of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery System, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, China
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
| |
Collapse
|
8
|
Squire S, Sebghati S, Hammond MC. Cytoplasmic Accumulation and Permeability of Antibiotics in Gram Positive and Gram Negative Bacteria Visualized in Real-Time via a Fluorogenic Tagging Strategy. ACS Chem Biol 2024; 19:3-8. [PMID: 38096425 PMCID: PMC10805102 DOI: 10.1021/acschembio.3c00510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 12/06/2023] [Accepted: 12/08/2023] [Indexed: 01/23/2024]
Abstract
In this study, we describe the first real-time live cell assay for compound accumulation and permeability in both Gram positive and Gram negative bacteria. The assay utilizes a novel fluorogenic tagging strategy that permits direct visualization of compound accumulation dynamics in the cytoplasm of live cells, unobscured by washing or other processing steps. Quantitative differences could be reproducibly measured by flow cytometry at compound concentrations below the limit of detection for MS-based approaches. We establish the fluorogenic assay in E. coli and B. subtilis and compare the intracellular accumulation of two antibiotics, ciprofloxacin and ampicillin, with related pharmacophores in these bacteria.
Collapse
Affiliation(s)
- Scott
O. Squire
- Department
of Chemistry, University of Utah, Salt Lake City, Utah 84112, United States
- Henry
Eyring Center for Cell & Genome Science, University of Utah, Salt Lake
City, Utah 84112, United States
| | - Sepehr Sebghati
- Department
of Chemistry, University of Utah, Salt Lake City, Utah 84112, United States
- Henry
Eyring Center for Cell & Genome Science, University of Utah, Salt Lake
City, Utah 84112, United States
| | - Ming C. Hammond
- Department
of Chemistry, University of Utah, Salt Lake City, Utah 84112, United States
- Henry
Eyring Center for Cell & Genome Science, University of Utah, Salt Lake
City, Utah 84112, United States
| |
Collapse
|
9
|
Lang M, Carvalho A, Baharoglu Z, Mazel D. Aminoglycoside uptake, stress, and potentiation in Gram-negative bacteria: new therapies with old molecules. Microbiol Mol Biol Rev 2023; 87:e0003622. [PMID: 38047635 PMCID: PMC10732077 DOI: 10.1128/mmbr.00036-22] [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] [Indexed: 12/05/2023] Open
Abstract
SUMMARYAminoglycosides (AGs) are long-known molecules successfully used against Gram-negative pathogens. While their use declined with the discovery of new antibiotics, they are now classified as critically important molecules because of their effectiveness against multidrug-resistant bacteria. While they can efficiently cross the Gram-negative envelope, the mechanism of AG entry is still incompletely understood, although this comprehension is essential for the development of new therapies in the face of the alarming increase in antibiotic resistance. Increasing antibiotic uptake in bacteria is one strategy to enhance effective treatments. This review aims, first, to consolidate old and recent knowledge about AG uptake; second, to explore the connection between AG-dependent bacterial stress and drug uptake; and finally, to present new strategies of potentiation of AG uptake for more efficient antibiotic therapies. In particular, we emphasize on the connection between sugar transport and AG potentiation.
Collapse
Affiliation(s)
- Manon Lang
- Institut Pasteur, Université Paris Cité, CNRS UMR3525, Unité Plasticité du Génome Bactérien, Paris, France
| | - André Carvalho
- Institut Pasteur, Université Paris Cité, CNRS UMR3525, Unité Plasticité du Génome Bactérien, Paris, France
| | - Zeynep Baharoglu
- Institut Pasteur, Université Paris Cité, CNRS UMR3525, Unité Plasticité du Génome Bactérien, Paris, France
| | - Didier Mazel
- Institut Pasteur, Université Paris Cité, CNRS UMR3525, Unité Plasticité du Génome Bactérien, Paris, France
| |
Collapse
|
10
|
Krin E, Carvalho A, Lang M, Babosan A, Mazel D, Baharoglu Z. RavA-ViaA antibiotic response is linked to Cpx and Zra2 envelope stress systems in Vibrio cholerae. Microbiol Spectr 2023; 11:e0173023. [PMID: 37861314 PMCID: PMC10848872 DOI: 10.1128/spectrum.01730-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 09/08/2023] [Indexed: 10/21/2023] Open
Abstract
IMPORTANCE The RavA-ViaA complex was previously found to sensitize Escherichia coli to aminoglycosides (AGs) in anaerobic conditions, but the mechanism is unknown. AGs are antibiotics known for their high efficiency against Gram-negative bacteria. In order to elucidate how the expression of the ravA-viaA genes increases bacterial susceptibility to aminoglycosides, we aimed at identifying partner functions necessary for increased tolerance in the absence of RavA-ViaA, in Vibrio cholerae. We show that membrane stress response systems Cpx and Zra2 are required in the absence of RavA-ViaA, for the tolerance to AGs and for outer membrane integrity. In the absence of these systems, the ∆ravvia strain's membrane becomes permeable to external agents such as the antibiotic vancomycin.
Collapse
Affiliation(s)
- Evelyne Krin
- Institut Pasteur, Université Paris Cité, CNRS UMR3525, Unité Plasticité du Génome Bactérien, Paris, France
| | - André Carvalho
- Institut Pasteur, Université Paris Cité, CNRS UMR3525, Unité Plasticité du Génome Bactérien, Paris, France
- Sorbonne Université, Collège doctoral, Paris, France
| | - Manon Lang
- Institut Pasteur, Université Paris Cité, CNRS UMR3525, Unité Plasticité du Génome Bactérien, Paris, France
- Sorbonne Université, Collège doctoral, Paris, France
| | - Anamaria Babosan
- Institut Pasteur, Université Paris Cité, CNRS UMR3525, Unité Plasticité du Génome Bactérien, Paris, France
| | - Didier Mazel
- Institut Pasteur, Université Paris Cité, CNRS UMR3525, Unité Plasticité du Génome Bactérien, Paris, France
| | - Zeynep Baharoglu
- Institut Pasteur, Université Paris Cité, CNRS UMR3525, Unité Plasticité du Génome Bactérien, Paris, France
| |
Collapse
|
11
|
Pierlé SA, Lang M, López-Igual R, Krin E, Fourmy D, Kennedy SP, Val ME, Baharoglu Z, Mazel D. Identification of the active mechanism of aminoglycoside entry in V. cholerae through characterization of sRNA ctrR, regulating carbohydrate utilization and transport. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.19.549712. [PMID: 37502966 PMCID: PMC10370196 DOI: 10.1101/2023.07.19.549712] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
The possible active entry of aminoglycosides in bacterial cells has been debated since the development of this antibiotic family. Here we report the identification of their active transport mechanism in Vibrio species. We combined genome-wide transcriptional analysis and fitness screens to identify alterations driven by treatment of V. cholerae with sub-minimum inhibitory concentrations (sub-MIC) of the aminoglycoside tobramycin. RNA-seq data showed downregulation of the small non-coding RNA ncRNA586 during such treatment, while Tn-seq revealed that inactivation of this sRNA was associated with improved fitness in the presence of tobramycin. This sRNA is located near sugar transport genes and previous work on a homologous region in Vibrio tasmaniensis suggested that this sRNA stabilizes gene transcripts for carbohydrate transport and utilization, as well as phage receptors. The role for ncRNA586, hereafter named ctrR, in the transport of both carbohydrates and aminoglycosides, was further investigated. Flow cytometry on cells treated with a fluorescent aminoglycoside confirmed the role of ctrR and of carbohydrate transporters in differential aminoglycoside entry. Despite sequence diversity, ctrR showed functional conservation across the Vibrionales. This system in directly modulated by carbon sources, suggesting regulation by carbon catabolite repression, a widely conserved mechanism in Gram-negative bacteria, priming future research on aminoglycoside uptake by sugar transporters in other bacterial species.
Collapse
Affiliation(s)
- Sebastian A. Pierlé
- Institut Pasteur, Université Paris Cité, CNRS UMR3525, Unité Plasticité du Génome Bactérien, F-75015 Paris, France
| | - Manon Lang
- Institut Pasteur, Université Paris Cité, CNRS UMR3525, Unité Plasticité du Génome Bactérien, F-75015 Paris, France
| | - Rocío López-Igual
- Institut Pasteur, Université Paris Cité, CNRS UMR3525, Unité Plasticité du Génome Bactérien, F-75015 Paris, France
| | - Evelyne Krin
- Institut Pasteur, Université Paris Cité, CNRS UMR3525, Unité Plasticité du Génome Bactérien, F-75015 Paris, France
| | - Dominique Fourmy
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198, Gif-sur-Yvette, France
| | - Sean P. Kennedy
- Institut Pasteur, Université Paris Cité, USR 3756 CNRS, Department of Computational Biology, 75015 Paris, France
| | - Marie-Eve Val
- Institut Pasteur, Université Paris Cité, CNRS UMR3525, Unité Plasticité du Génome Bactérien, F-75015 Paris, France
| | - Zeynep Baharoglu
- Institut Pasteur, Université Paris Cité, CNRS UMR3525, Unité Plasticité du Génome Bactérien, F-75015 Paris, France
| | - Didier Mazel
- Institut Pasteur, Université Paris Cité, CNRS UMR3525, Unité Plasticité du Génome Bactérien, F-75015 Paris, France
| |
Collapse
|
12
|
Wongso H, Hendra R, Nugraha AS, Ritawidya R, Saptiama I, Kusumaningrum CE. Microbial metabolites diversity and their potential as molecular template for the discovery of new fluorescent and radiopharmaceutical probes. Trends Analyt Chem 2022. [DOI: 10.1016/j.trac.2022.116900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
13
|
Wijers CDM, Pham L, Douglass MV, Skaar EP, Palmer LD, Noto MJ. Gram-negative bacteria act as a reservoir for aminoglycoside antibiotics that interact with host factors to enhance bacterial killing in a mouse model of pneumonia. FEMS MICROBES 2022; 3:xtac016. [PMID: 35909464 PMCID: PMC9326624 DOI: 10.1093/femsmc/xtac016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 05/04/2022] [Accepted: 05/11/2022] [Indexed: 01/28/2023] Open
Abstract
In vitro exposure of multiple Gram-negative bacteria to an aminoglycoside (AG) antibiotic has previously been demonstrated to result in bacterial alterations that interact with host factors to suppress Gram-negative pneumonia. However, the mechanisms resulting in suppression are not known. Here, the hypothesis that Gram-negative bacteria bind and retain AGs, which are introduced into the lung and interact with host defenses to affect bacterial killing, was tested. Following in vitro exposure of one of several, pathogenic Gram-negative bacteria to the AG antibiotics kanamycin or gentamicin, AGs were detected in bacterial cell pellets (up to 208 μg/mL). Using inhibitors of AG binding and internalization, the bacterial outer membrane was implicated as the predominant kanamycin and gentamicin reservoir. Following intranasal administration of gentamicin-bound bacteria or gentamicin solution at the time of infection with live, AG-naïve bacteria, gentamicin was detected in the lungs of infected mice (up to 8 μg/g). Co-inoculation with gentamicin-bound bacteria resulted in killing of AG-naïve bacteria by up to 3-log10, mirroring the effects of intranasal gentamicin treatment. In vitro killing of AG-naïve bacteria mediated by kanamycin-bound bacteria required the presence of detergents or pulmonary surfactant, suggesting that increased bacterial killing inside the murine lung is facilitated by the detergent component of pulmonary surfactant. These findings demonstrate that Gram-negative bacteria bind and retain AGs that can interact with host-derived pulmonary surfactant to enhance bacterial killing in the lung. This may help explain why AGs appear to have unique efficacy in the lung and might expand their clinical utility.
Collapse
Affiliation(s)
- Christiaan D M Wijers
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, 1161 21st Avenue South, Nashville, TN 37232, United States
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, 1161 21st Avenue South, Nashville, TN 37232, United States
| | - Ly Pham
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, 1161 21st Avenue South, Nashville, TN 37232, United States
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, 1161 21st Avenue South, Nashville, TN 37232, United States
| | - Martin V Douglass
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, 1161 21st Avenue South, Nashville, TN 37232, United States
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, 1161 21st Avenue South, Nashville, TN 37232, United States
| | - Eric P Skaar
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, 1161 21st Avenue South, Nashville, TN 37232, United States
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, 1161 21st Avenue South, Nashville, TN 37232, United States
| | - Lauren D Palmer
- Department of Microbiology and Immunology, University of Illinois Chicago, 835 South Wolcott Avenue, Chicago, IL 60612, United States
| | - Michael J Noto
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, 1161 21st Avenue South, Nashville, TN 37232, United States
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, 1161 21st Avenue South, Nashville, TN 37232, United States
- Department of Medicine, Vanderbilt University Medical Center, 1161 21st Avenue South, Nashville, TN 37232, United States
| |
Collapse
|
14
|
Lang M, Krin E, Korlowski C, Sismeiro O, Varet H, Coppée JY, Mazel D, Baharoglu Z. Sleeping ribosomes: Bacterial signaling triggers RaiA mediated persistence to aminoglycosides. iScience 2021; 24:103128. [PMID: 34611612 PMCID: PMC8476650 DOI: 10.1016/j.isci.2021.103128] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 09/07/2021] [Accepted: 09/09/2021] [Indexed: 12/19/2022] Open
Abstract
Indole is a molecule proposed to be involved in bacterial signaling. We find that indole secretion is induced by sublethal tobramycin concentrations and increases persistence to aminoglycosides in V. cholerae. Indole transcriptomics showed increased expression of raiA, a ribosome associated factor. Deletion of raiA abolishes the appearance of indole dependent persisters to aminoglycosides, although its overexpression leads to 100-fold increase of persisters, and a reduction in lag phase, evocative of increased active 70S ribosome content, confirmed by sucrose gradient analysis. We propose that, under stress conditions, RaiA-bound inactive 70S ribosomes are stored as “sleeping ribosomes”, and are rapidly reactivated upon stress relief. Our results point to an active process of persister formation through ribosome protection during translational stress (e.g., aminoglycoside treatment) and reactivation upon antibiotic removal. Translation is a universal process, and these results could help elucidate a mechanism of persistence formation in a controlled, thus inducible way. Indole is produced under sub-MIC tobramycin stress in V. cholerae and upregulates raiA RaiA is involved in indole-dependent formation of aminoglycoside specific persisters RaiA overexpression allows faster growth restart and increases 70S ribosome content RaiA-bound inactive 70S ribosomes form intact and reactivable sleeping ribosome pools
Collapse
Affiliation(s)
- Manon Lang
- Département Génomes et Génétique, Institut Pasteur, UMR3525, CNRS, Unité Plasticité du Génome Bactérien, 75015 Paris, France
- Sorbonne Université, Collège Doctoral, 75005 Paris, France
| | - Evelyne Krin
- Département Génomes et Génétique, Institut Pasteur, UMR3525, CNRS, Unité Plasticité du Génome Bactérien, 75015 Paris, France
| | - Chloé Korlowski
- Département Génomes et Génétique, Institut Pasteur, UMR3525, CNRS, Unité Plasticité du Génome Bactérien, 75015 Paris, France
| | - Odile Sismeiro
- Biomics Technological Platform, Center for Technological Resources and Research, Institut Pasteur, 75015 Paris, France
| | - Hugo Varet
- Biomics Technological Platform, Center for Technological Resources and Research, Institut Pasteur, 75015 Paris, France
- Bioinformatics and Biostatistics Hub, Department of Computational Biology, USR 3756 CNRS, Institut Pasteur, 75015 Paris, France
| | - Jean-Yves Coppée
- Biomics Technological Platform, Center for Technological Resources and Research, Institut Pasteur, 75015 Paris, France
| | - Didier Mazel
- Département Génomes et Génétique, Institut Pasteur, UMR3525, CNRS, Unité Plasticité du Génome Bactérien, 75015 Paris, France
- Corresponding author
| | - Zeynep Baharoglu
- Département Génomes et Génétique, Institut Pasteur, UMR3525, CNRS, Unité Plasticité du Génome Bactérien, 75015 Paris, France
- Corresponding author
| |
Collapse
|
15
|
Carvalho A, Mazel D, Baharoglu Z. Deficiency in cytosine DNA methylation leads to high chaperonin expression and tolerance to aminoglycosides in Vibrio cholerae. PLoS Genet 2021; 17:e1009748. [PMID: 34669693 PMCID: PMC8559950 DOI: 10.1371/journal.pgen.1009748] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 11/01/2021] [Accepted: 10/04/2021] [Indexed: 12/11/2022] Open
Abstract
Antibiotic resistance has become a major global issue. Understanding the molecular mechanisms underlying microbial adaptation to antibiotics is of keen importance to fight Antimicrobial Resistance (AMR). Aminoglycosides are a class of antibiotics that target the small subunit of the bacterial ribosome, disrupting translational fidelity and increasing the levels of misfolded proteins in the cell. In this work, we investigated the role of VchM, a DNA methyltransferase, in the response of the human pathogen Vibrio cholerae to aminoglycosides. VchM is a V. cholerae specific orphan m5C DNA methyltransferase that generates cytosine methylation at 5'-RCCGGY-3' motifs. We show that deletion of vchM, although causing a growth defect in absence of stress, allows V. cholerae cells to cope with aminoglycoside stress at both sub-lethal and lethal concentrations of these antibiotics. Through transcriptomic and genetic approaches, we show that groESL-2 (a specific set of chaperonin-encoding genes located on the second chromosome of V. cholerae), are upregulated in cells lacking vchM and are needed for the tolerance of vchM mutant to lethal aminoglycoside treatment, likely by fighting aminoglycoside-induced misfolded proteins. Interestingly, preventing VchM methylation of the four RCCGGY sites located in groESL-2 region, leads to a higher expression of these genes in WT cells, showing that the expression of these chaperonins is modulated in V. cholerae by DNA methylation.
Collapse
Affiliation(s)
- André Carvalho
- Département Génomes et Génétique, Institut Pasteur, UMR3525, CNRS, Unité Plasticité du Génome Bactérien, Paris, France
- Sorbonne Université, Collège doctoral, F-75005 Paris, France
| | - Didier Mazel
- Département Génomes et Génétique, Institut Pasteur, UMR3525, CNRS, Unité Plasticité du Génome Bactérien, Paris, France
| | - Zeynep Baharoglu
- Département Génomes et Génétique, Institut Pasteur, UMR3525, CNRS, Unité Plasticité du Génome Bactérien, Paris, France
| |
Collapse
|
16
|
Array-based microbial identification upon extracellular aminoglycoside residue sensing. Anal Bioanal Chem 2021; 413:4689-4696. [PMID: 33893514 DOI: 10.1007/s00216-021-03346-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 04/10/2021] [Accepted: 04/13/2021] [Indexed: 10/21/2022]
Abstract
Sensitive and rapid identification of pathogenic microorganisms is of great importance for clinical diagnosis and treatment. In this study, we developed an ultrasensitive colorimetric sensor array (CSA) based on the interactions between aminoglycoside antibiotics (AMGs) and Ag nanoparticles decorated with β-cyclodextrin (AgNPs@β-CD) to discriminate microorganisms quickly and accurately. Microorganisms can absorb different amounts of AMGs after incubation. Upon the addition of AgNPs@β-CD, the corresponding extracellular AMG residues will bind to AgNPs@β-CD, leading to color changes due to the modifications in localized surface plasmon resonance. The array was developed using 4 AMGs as sensing elements and AgNPs@β-CD as the colorimetric probe to generate a unique colorimetric response pattern for each microorganism. Standard chemometric methods indicated excellent discrimination among 20 microorganisms at low concentrations of 2 × 106 CFU/mL. Therefore, this ultrasensitive CSA can be used for microbial discrimination portably and efficiently. Importantly, the concentration of microbial discrimination by our array is much lower than that of prior CSAs. This method of extracellular residue sensing also provided a new strategy to improve the sensitivity of conventional CSA in the discrimination of microorganisms, to measure the amount of intercellular uptake of AMGs by microorganisms, and to screen drugs that can easily be accumulated by the pathogenic microorganisms.
Collapse
|
17
|
Hira J, Uddin MJ, Haugland MM, Lentz CS. From Differential Stains to Next Generation Physiology: Chemical Probes to Visualize Bacterial Cell Structure and Physiology. Molecules 2020; 25:E4949. [PMID: 33114655 PMCID: PMC7663024 DOI: 10.3390/molecules25214949] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/21/2020] [Accepted: 10/23/2020] [Indexed: 12/16/2022] Open
Abstract
Chemical probes have been instrumental in microbiology since its birth as a discipline in the 19th century when chemical dyes were used to visualize structural features of bacterial cells for the first time. In this review article we will illustrate the evolving design of chemical probes in modern chemical biology and their diverse applications in bacterial imaging and phenotypic analysis. We will introduce and discuss a variety of different probe types including fluorogenic substrates and activity-based probes that visualize metabolic and specific enzyme activities, metabolic labeling strategies to visualize structural features of bacterial cells, antibiotic-based probes as well as fluorescent conjugates to probe biomolecular uptake pathways.
Collapse
Affiliation(s)
- Jonathan Hira
- Research Group for Host-Microbe Interactions, Department of Medical Biology and Centre for New Antibacterial Strategies (CANS), UiT—The Arctic University of Norway, 9019 Tromsø, Norway; (J.H.); (M.J.U.)
| | - Md. Jalal Uddin
- Research Group for Host-Microbe Interactions, Department of Medical Biology and Centre for New Antibacterial Strategies (CANS), UiT—The Arctic University of Norway, 9019 Tromsø, Norway; (J.H.); (M.J.U.)
| | - Marius M. Haugland
- Department of Chemistry and Centre for New Antibacterial Strategies (CANS), UiT—The Arctic University of Norway, 9019 Tromsø, Norway;
| | - Christian S. Lentz
- Research Group for Host-Microbe Interactions, Department of Medical Biology and Centre for New Antibacterial Strategies (CANS), UiT—The Arctic University of Norway, 9019 Tromsø, Norway; (J.H.); (M.J.U.)
| |
Collapse
|