1
|
Huang X, Hu Z, Shang W, Chen J, Hu Q, Zhou Y, Ding R, Yin J, Li M, Liu H, Dou J, Peng H, Rao Y, Liu L, Wang Y, Tan L, Yang Y, Wu J, Xiao C, Yang Y, Rao X. Beta-Lactam Antibiotics Promote Extracellular Vesicle Production of Staphylococcus aureus Through ROS-Mediated Lipid Metabolic Reprogramming. J Extracell Vesicles 2025; 14:e70077. [PMID: 40314062 PMCID: PMC12046293 DOI: 10.1002/jev2.70077] [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: 11/21/2024] [Accepted: 03/26/2025] [Indexed: 05/03/2025] Open
Abstract
Bacterial extracellular vesicles (EVs) are natural reservoirs of biological active substances. They exhibit promising application in developing bioproducts such as vaccine, drug-delivery system and anticancer agent. However, the low yield of naturally secreted EVs during bacterial growth is a bottleneck factor that restricts EV applications. In this study, we showed that sub-minimum inhibitory concentration (MIC) of β-lactams boosted EV production in various Staphylococcus aureus strains. The expression of penicillin-binding protein (PBP) genes increased after β-lactam treatment, and the inactivation of alternative PBPs promoted EV secretion of S. aureus. We also demonstrated that sub-MIC β-lactams promoted EV production via a reactive oxygen species (ROS)-dependent pathway. Deletion of redundant pbp genes enhanced oxacillin (OXA)-stimulated ROS levels. Transcriptomic and lipidomic analyses revealed that OXA-induced ROS triggered lipid metabolic reprogramming in S. aureus. Particularly, ROS promoted lipid peroxidation (LPO) and increased the biosynthesis of phosphatidic acid (PA) and lipoteichoic acid (LTA) that contributed to EV generation. Furthermore, OXA treatment altered the diversity of EV-loaded proteins. OXA-treated ∆ agr /OXAEVs induced stronger Dengue EDIII-specific antibodies in BALB/c mice than did ∆ agrEVs. Overall, this study provided mechanic insights into β-lactam-promoted EV production in S. aureus, and highlighted the potential strategies to prepare EVs for various applications.
Collapse
Affiliation(s)
- Xiaonan Huang
- Department of MicrobiologyCollege of Basic Medical SciencesKey Laboratory of Microbial Engineering under the Educational Committee in ChongqingArmy Medical UniversityChongqingChina
| | - Zhen Hu
- Department of MicrobiologyCollege of Basic Medical SciencesKey Laboratory of Microbial Engineering under the Educational Committee in ChongqingArmy Medical UniversityChongqingChina
| | - Weilong Shang
- Department of MicrobiologyCollege of Basic Medical SciencesKey Laboratory of Microbial Engineering under the Educational Committee in ChongqingArmy Medical UniversityChongqingChina
| | - Juan Chen
- Department of PharmacyXinqiao HospitalArmy Medical UniversityChongqingChina
| | - Qiwen Hu
- Department of MicrobiologyCollege of Basic Medical SciencesKey Laboratory of Microbial Engineering under the Educational Committee in ChongqingArmy Medical UniversityChongqingChina
| | - Yumin Zhou
- Department of DermatologySouthwest HospitalArmy Medical UniversityChongqingChina
| | - Ruolan Ding
- Department of MicrobiologySchool of MedicineChongqing UniversityChongqingChina
| | - Jing Yin
- Department of NeurologyFirst Affiliated Hospital of Kunming Medical UniversityKunmingChina
| | - Mengyang Li
- Department of MicrobiologySchool of MedicineChongqing UniversityChongqingChina
| | - He Liu
- Department of MicrobiologyCollege of Basic Medical SciencesKey Laboratory of Microbial Engineering under the Educational Committee in ChongqingArmy Medical UniversityChongqingChina
| | - Jianxiong Dou
- Department of MicrobiologyCollege of Basic Medical SciencesKey Laboratory of Microbial Engineering under the Educational Committee in ChongqingArmy Medical UniversityChongqingChina
| | - Huagang Peng
- Department of MicrobiologyCollege of Basic Medical SciencesKey Laboratory of Microbial Engineering under the Educational Committee in ChongqingArmy Medical UniversityChongqingChina
| | - Yifan Rao
- Department of Emergency MedicineXinqiao HospitalArmy Medical UniversityChongqingChina
| | - Lu Liu
- Department of MicrobiologyCollege of Basic Medical SciencesKey Laboratory of Microbial Engineering under the Educational Committee in ChongqingArmy Medical UniversityChongqingChina
| | - Yuting Wang
- Department of MicrobiologyCollege of Basic Medical SciencesKey Laboratory of Microbial Engineering under the Educational Committee in ChongqingArmy Medical UniversityChongqingChina
| | - Li Tan
- Department of MicrobiologyCollege of Basic Medical SciencesKey Laboratory of Microbial Engineering under the Educational Committee in ChongqingArmy Medical UniversityChongqingChina
| | - Yuhua Yang
- Department of MicrobiologyCollege of Basic Medical SciencesKey Laboratory of Microbial Engineering under the Educational Committee in ChongqingArmy Medical UniversityChongqingChina
| | - Jianghong Wu
- Department of MicrobiologyCollege of Basic Medical SciencesKey Laboratory of Microbial Engineering under the Educational Committee in ChongqingArmy Medical UniversityChongqingChina
- Department of Emergency MedicineXinqiao HospitalArmy Medical UniversityChongqingChina
| | - Chuan Xiao
- Department of MicrobiologyCollege of Basic Medical SciencesKey Laboratory of Microbial Engineering under the Educational Committee in ChongqingArmy Medical UniversityChongqingChina
| | - Yi Yang
- Department of MicrobiologyCollege of Basic Medical SciencesKey Laboratory of Microbial Engineering under the Educational Committee in ChongqingArmy Medical UniversityChongqingChina
| | - Xiancai Rao
- Department of MicrobiologyCollege of Basic Medical SciencesKey Laboratory of Microbial Engineering under the Educational Committee in ChongqingArmy Medical UniversityChongqingChina
- Department of MicrobiologySchool of MedicineChongqing UniversityChongqingChina
| |
Collapse
|
2
|
Kawai Y, Errington J. Antibiotic fosmidomycin protects bacteria from cell wall perturbations by antagonizing oxidative damage-mediated cell lysis. Front Microbiol 2025; 16:1560235. [PMID: 40309104 PMCID: PMC12041025 DOI: 10.3389/fmicb.2025.1560235] [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: 01/14/2025] [Accepted: 03/31/2025] [Indexed: 05/02/2025] Open
Abstract
Cell wall peptidoglycan is a defining component of bacterial cells, and its biosynthesis is a major target for medically important antibiotics. Recent studies have revealed that antibiotics can kill cells not only by their direct effects on wall synthesis, but also by downstream perturbations of metabolic homeostasis, leading to oxidative damage-mediated lysis. In this paper, we have investigated the killing effects of various effectors of cell wall inhibition, including an antibiotic inhibitor of isoprenoid synthesis, fosmidomycin, in Bacillus subtilis. We show that oxidative damage largely contributes to the toxic effect (rapid cell lysis) induced by inhibition of peptidoglycan synthesis, but not by inhibition of the isoprenoid synthetic pathway. Remarkably, intermediate concentrations of fosmidomycin, confer resistance to lysis when peptidoglycan synthesis is perturbed. We show that this is because fosmidomycin not only blocks peptidoglycan synthesis, but also impairs the synthesis of menaquinone, which, protects cells from respiratory chain-associated oxidative damage and lysis. Our results provide new insights into the critical involvement of metabolic pathways, such as isoprenoid biosynthesis, on the antibiotic efficacy and evasion by bacteria. This work advances our understanding of bacterial physiology as well as antibiotic activity and resistance.
Collapse
Affiliation(s)
- Yoshikazu Kawai
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | | |
Collapse
|
3
|
Patel Y, Helmann JD. A mutation in RNA polymerase imparts resistance to β-lactams by preventing dysregulation of amino acid and nucleotide metabolism. Cell Rep 2025; 44:115268. [PMID: 39908144 PMCID: PMC11975431 DOI: 10.1016/j.celrep.2025.115268] [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: 05/28/2024] [Revised: 11/19/2024] [Accepted: 01/15/2025] [Indexed: 02/07/2025] Open
Abstract
Resistance to diverse antibiotics can result from mutations in RNA polymerase (RNAP), but the underlying mechanisms remain poorly understood. In this study, we compare two Bacillus subtilis RNAP mutations: one in β' (rpoC G1122D) that increases resistance to cefuroxime (CEF; a model β-lactam) and one in β (rpoB H482Y) that increases sensitivity. CEF resistance is mediated by a decrease in branched-chain amino acid (BCAA), methionine, and pyrimidine pathways. These same pathways are upregulated by CEF, and their derepression increases CEF sensitivity and antibiotic-induced production of reactive oxygen species. The CEF-resistant rpoC G1122D mutant evades these metabolic perturbations, and repression of the BCAA and pyrimidine pathways may function to restrict membrane biogenesis, which is beneficial when cell wall synthesis is impaired. These findings provide a vivid example of how RNAP mutations, which commonly arise in response to diverse selection conditions, can rewire cellular metabolism to enhance fitness.
Collapse
Affiliation(s)
- Yesha Patel
- Department of Microbiology, Cornell University, Ithaca, NY 14853-8101, USA
| | - John D Helmann
- Department of Microbiology, Cornell University, Ithaca, NY 14853-8101, USA.
| |
Collapse
|
4
|
Cross T, Torres F, McGee AP, Aliyu T, Westblade LF, Singh A, Dörr T. Prevalence and mechanisms of high-level carbapenem antibiotic tolerance in clinical isolates of Klebsiella pneumoniae. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.19.639047. [PMID: 40027789 PMCID: PMC11870580 DOI: 10.1101/2025.02.19.639047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Antibiotic tolerance is the ability of bacteria to survive normally lethal doses of antibiotics for extended time periods. Clinically significant Enterobacterales, for example, often exhibit high tolerance to the last-resort antibiotic meropenem. Meropenem tolerance is associated with formation of cell wall-deficient spheroplasts that readily recover to rod shape and normal growth upon removal of the antibiotic. Both the true prevalence of tolerance, and genetic mechanisms underlying it, remain poorly understood. Here, we find that meropenem tolerance is widespread among clinical Enterobacterales. Using forward genetics, we uncover novel tolerance factors in a hypertolerant isolate of the ESKAPE pathogen Klebsiella pneumoniae . We find that multiple mechanisms contribute to tolerance, and that cell envelope stress responses (PhoPQ, Cpx, Rcs and OmpR/EnvZ) collectively promote spheroplast stability and recovery, while the lytic transglycosylase MltB counteracts it. Our data indicate that tolerance is widespread among clinical isolates, and that outer membrane maintenance is a key factor promoting survival of tolerant K. pneumoniae .
Collapse
|
5
|
El-Araby AM, Fisher JF, Mobashery S. Bacterial peptidoglycan as a living polymer. Curr Opin Chem Biol 2025; 84:102562. [PMID: 39700530 PMCID: PMC11788026 DOI: 10.1016/j.cbpa.2024.102562] [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: 10/12/2024] [Revised: 11/24/2024] [Accepted: 11/25/2024] [Indexed: 12/21/2024]
Abstract
The peptidoglycan manifests as a multifaceted component of the bacterial cell wall. Throughout the lifecycle of the bacterium, the peptidoglycan is deconstructed, rebuilt, and remodeled for bacterial cell growth and replication. Degradation products of the peptidoglycan serve as precursors for cell-wall building blocks via recycling processes and as signaling molecules. Cell-wall recycling and de novo cell-wall synthesis converge biochemically at the cytoplasmic compartment. Peptidoglycan biochemistry is finely tuned to maintain the polymer's functions and is intimately connected to antibiotic-resistance mechanisms. Cell-wall-modifying enzymes present a unique opportunity for the discovery of antibiotics and antibiotic adjuvants. The unique chemical template of the peptidoglycan has been a target of numerous chemical biology approaches for investigating its functions and modulation. In this review, we highlight the current perspective on peptidoglycan research. We present recent efforts to understand the peptidoglycan as a functional component of antibiotic resistance, and as a target for antimicrobial therapy.
Collapse
Affiliation(s)
- Amr M El-Araby
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Jed F Fisher
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Shahriar Mobashery
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, 46556, USA.
| |
Collapse
|
6
|
Isaac KP, Krishnamurthy P, Unni SN, Rao SN, Parthasarathy K. Laser Speckle Image analysis for identifying the minimum lethal concentration of ampicillin in Escherichia coli liquid cultures. J Microbiol Methods 2024; 227:107068. [PMID: 39528096 DOI: 10.1016/j.mimet.2024.107068] [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: 06/11/2024] [Revised: 11/05/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Understanding a pathogen's sensitivity to antimicrobial drugs through Minimum Lethal Concentration (MLC) is crucial for effective treatment planning for bactericidal drugs. In this paper, we propose a novel approach using Laser Speckle Imaging (LSI) to determine the MLC of Escherichia coli (E. coli), a common pathogenic bacterial species. LSI enables the capture and analysis of the dynamic changes in speckle patterns caused by alterations in optical scattering and shape alterations of bacterial cells as a response to antibiotic treatments through a label-free approach. The observed speckle pattern changes are correlated with the gold standard method to determine the MLC, representing the lowest concentration at which E. coli is lethally affected. The results demonstrate the potential of LSI as a reliable and rapid method for determining the MLC of E. coli. This method has much potential for antimicrobial research since it provides a quick, non-destructive evaluation of bacterial responses to various bactericidal antibiotic doses without requiring labor-intensive processes like pour plate tests to calculate the MLC.
Collapse
Affiliation(s)
- Kiran Philip Isaac
- Biophotonics Lab, Department of Applied Mechanics and Biomedical Engineering, IIT Madras, Chennai 600036, India
| | - Priya Krishnamurthy
- Biophotonics Lab, Department of Applied Mechanics and Biomedical Engineering, IIT Madras, Chennai 600036, India
| | - Sujatha Narayanan Unni
- Biophotonics Lab, Department of Applied Mechanics and Biomedical Engineering, IIT Madras, Chennai 600036, India.
| | - Sudha Narayani Rao
- Center for Drug Discovery and Development, Sathyabama Institute of Science and Technology, Chennai, India
| | - Krupakar Parthasarathy
- Center for Drug Discovery and Development, Sathyabama Institute of Science and Technology, Chennai, India
| |
Collapse
|
7
|
Li Q, Feng H, Tian Q, Xiang Y, Wang X, He YX, Zhu K. Discovery of antibacterial diketones against gram-positive bacteria. Cell Chem Biol 2024; 31:1874-1884.e6. [PMID: 39089260 DOI: 10.1016/j.chembiol.2024.06.017] [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: 10/23/2023] [Revised: 04/21/2024] [Accepted: 06/28/2024] [Indexed: 08/03/2024]
Abstract
The rapid rise of antibiotic resistance calls for the discovery of new antibiotics with distinct antibacterial mechanisms. New target mining is indispensable for developing antibiotics. Plant-microbial antibiotics are appealing to underexplored sources due to a dearth of comprehensive understanding of antibacterial activity and the excavation of new targets. Here, a series of phloroglucinol derivatives of plant-root-associated Pseudomonas fluorescens were synthesized for structure-activity relationship analysis. Notably, 2,4-diproylphloroglucinol (DPPG) displayed efficient bactericidal activity against a wide range of gram-positive bacteria. Importantly, mechanistic study exhibits that DPPG binds to type II NADH dehydrogenase (NDH-2), an essential enzyme catalyzing the transfer of electrons from NADH to quinones in the electron transport chain (ETC), blocking electron transfer in S. aureus. Last, we validated the efficacy of DPPG in vivo through animal infection models. Our findings not only provide a distinct antibiotic lead to treat multidrug resistant pathogens but also identify a promising antibacterial target.
Collapse
Affiliation(s)
- Qian Li
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Hanzhong Feng
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou 730000, China
| | - Qiong Tian
- State Key Laboratory of Applied Organic Chemistry, Department of Chemistry and School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Yun Xiang
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou 730000, China
| | - Xiaolei Wang
- State Key Laboratory of Applied Organic Chemistry, Department of Chemistry and School of Pharmacy, Lanzhou University, Lanzhou 730000, China.
| | - Yong-Xing He
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou 730000, China; State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China.
| | - Kui Zhu
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
8
|
Keller MR, Soni V, Brown M, Rosch KM, Saleh A, Rhee K, Doerr T. Sugar phosphate-mediated inhibition of peptidoglycan precursor synthesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.13.623475. [PMID: 39605520 PMCID: PMC11601392 DOI: 10.1101/2024.11.13.623475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Antibiotic tolerance, the widespread ability of diverse pathogenic bacteria to sustain viability in the presence of typically bactericidal antibiotics for extended time periods, is an understudied steppingstone towards antibiotic resistance. The Gram-negative pathogen Vibrio cholerae, the causative agent of cholera, is highly tolerant to β-lactam antibiotics. We previously found that the disruption of glycolysis, via deletion of pgi (vc0374, glucose-6-phosphate isomerase), resulted in significant cell wall damage and increased sensitivity towards β-lactam antibiotics. Here, we uncover the mechanism of this resulting damage. We find that glucose causes growth inhibition, partial lysis, and a damaged cell envelope in Δpgi. Supplementation with N-acetylglucosamine, but not other carbon sources (either from upper glycolysis, TCA cycle intermediates, or cell wall precursors) restored growth, re-established antibiotic resistance towards β-lactams, and recovered cellular morphology of a pgi mutant exposed to glucose. Targeted metabolomics revealed the cell wall precursor synthetase enzyme GlmU (vc2762, coding for the bifunctional enzyme that converts glucosamine-1P to UDP-GlcNAc) as a critical bottleneck and mediator of glucose toxicity in Δpgi. In vitro assays of GlmU revealed that sugar phosphates (primarily glucose-1-phosphate) inhibit the acetyltransferase activity of GlmU (likely competitively), resulting in compromised PG and LPS biosynthesis. These findings identify GlmU as a critical branchpoint enzyme between central metabolism and cell envelope integrity and reveal the molecular mechanism of Δpgi glucose toxicity in Vibrio cholerae.
Collapse
Affiliation(s)
- Megan R. Keller
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA
- Cornell Institute of Host-Microbe Interactions and Disease, Cornell University, Ithaca NY 14853, USA
| | - Vijay Soni
- Department of Medicine, Division of Infectious Diseases, Weill Cornell Medicine, New York, NY 10021
| | - Megan Brown
- Department of Medicine, Division of Infectious Diseases, Weill Cornell Medicine, New York, NY 10021
| | - Kelly M. Rosch
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA
- Cornell Institute of Host-Microbe Interactions and Disease, Cornell University, Ithaca NY 14853, USA
| | - Anas Saleh
- Department of Medicine, Division of Infectious Diseases, Weill Cornell Medicine, New York, NY 10021
| | - Kyu Rhee
- Cornell Institute of Host-Microbe Interactions and Disease, Cornell University, Ithaca NY 14853, USA
- Department of Medicine, Division of Infectious Diseases, Weill Cornell Medicine, New York, NY 10021
| | - Tobias Doerr
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA
- Department of Microbiology, Cornell University, Ithaca NY 14853, USA
- Cornell Institute of Host-Microbe Interactions and Disease, Cornell University, Ithaca NY 14853, USA
| |
Collapse
|
9
|
Horak RD, Ciemniecki JA, Newman DK. Bioenergetic suppression by redox-active metabolites promotes antibiotic tolerance in Pseudomonas aeruginosa. Proc Natl Acad Sci U S A 2024; 121:e2406555121. [PMID: 39503891 PMCID: PMC11573671 DOI: 10.1073/pnas.2406555121] [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: 04/01/2024] [Accepted: 10/01/2024] [Indexed: 11/21/2024] Open
Abstract
The proton-motive force (PMF), consisting of a pH gradient and a membrane potential (ΔΨ) underpins many processes essential to bacterial growth and/or survival. Yet bacteria often enter a bioenergetically diminished state characterized by a low PMF. Consequently, they have increased tolerance for diverse stressors, including clinical antibiotics. Despite the ubiquity of low metabolic rates in the environment, the extent to which bacteria have agency over entry into such a low-bioenergetic state has received relatively little attention. Here, we tested the hypothesis that production of redox-active metabolites (RAMs) could drive such a physiological transition. Pseudomonas aeruginosa is an opportunistic pathogen that produces phenazines, model RAMs that are highly toxic in the presence of molecular oxygen (O2). Under oxic conditions, the phenazines pyocyanin and phenazine-1-carboximide, as well as toxoflavin-a RAM produced by Burkholderia species-suppress the ΔΨ in distinct ways across distributions of single cells, reduce the efficiency of proton pumping, and lower cellular adenosine-triphosphate (ATP) levels. In planktonic culture, the degree and rate by which each RAM lowers the ΔΨ correlates with the protection it confers against antibiotics that strongly impact cellular energy flux. This bioenergetic suppression requires the RAM's presence and corresponds to its cellular reduction rate and abiotic oxidation rate by O2; it can be reversed by increasing the ΔΨ with nigericin. RAMs similarly impact the bioenergetic state of cells in (hyp)oxic biofilm aggregates. Collectively, these findings demonstrate that bacteria can suppress their bioenergetic state by the production of endogenous toxins in a manner that bolsters stress resilience.
Collapse
Affiliation(s)
- Richard D. Horak
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA91125
| | - John A. Ciemniecki
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA91125
| | - Dianne K. Newman
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA91125
- Division of Geological and Planetary Sciences, California Institute of Technology, Pasadena, CA91125
| |
Collapse
|
10
|
Gupta R, Bhando T, Pathania R. Overexpression of l,d-Transpeptidase A Induces Dispensability of Rod Complex in Escherichia coli. ACS Infect Dis 2024; 10:3928-3938. [PMID: 39412350 DOI: 10.1021/acsinfecdis.4c00597] [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: 11/09/2024]
Abstract
Antimicrobial resistance (AMR) is a significant global threat, and the presence of resistance-determinant genes is one of the major driving forces behind it. The bacterial rod complex is an essential set of proteins that is crucial for cell survival due to its role in cell wall biogenesis and shape maintenance. Therefore, these proteins offer excellent potential as drug targets; however, compensatory mutations in nontarget genes render this complex nonessential. The MreB protein of this complex is an actin homologue that rotates along the longitudinal axis of the cell to provide rod shape to the bacteria. In this study, using chemical-chemical interaction profiling and FtsZ suppression assay, we identified the MreB targeting activity of IITR07865, a previously discovered small molecule in our lab. Escherichia coli suppressors against IITR07865 revealed mutations in two cell division-associated genes, min C and pal, that have not been previously implicated in rod complex essentiality. IITR07865 resistant mutants were found to inactivate and render the rod complex nonessential, making the rod complex inhibitors ineffective. Further, through transcriptome analysis, we reveal the primary cause of resistance in suppressor strains to be the overexpression of an l, d-transpeptidase A enzyme, which is involved in peptidoglycan and Braun's lipoprotein cross-linking. Our results demonstrate a novel mechanism of resistance development in rod-shaped Gram-negative bacterial pathogen E. coli involved in UTIs where mecillinam, a clinically used antibiotic that targets rod complex, is a drug of choice.
Collapse
Affiliation(s)
- Rinki Gupta
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247 667, India
| | - Timsy Bhando
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247 667, India
| | - Ranjana Pathania
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247 667, India
| |
Collapse
|
11
|
Zhang M, Yang S, Liu Y, Zou Z, Zhang Y, Tian Y, Zhang R, Liu D, Wu C, Shen J, Song H, Wang Y. Anticancer agent 5-fluorouracil reverses meropenem resistance in carbapenem-resistant Gram-negative pathogens. Int J Antimicrob Agents 2024; 64:107337. [PMID: 39293771 DOI: 10.1016/j.ijantimicag.2024.107337] [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: 04/02/2024] [Revised: 08/24/2024] [Accepted: 09/09/2024] [Indexed: 09/20/2024]
Abstract
The global increasing incidence of clinical infections caused by carbapenem-resistant Gram-negative pathogens requires urgent and effective treatment strategies. Antibiotic adjuvants represent a promising approach to enhance the efficacy of meropenem against carbapenem-resistant bacteria. This study shows that the anticancer agent 5-fluorouracil (5-FU, 50 µM) significantly reduced the minimum inhibitory concentration of meropenem against blaNDM-5 positive Escherichia coli by 32-fold through cell-based high-throughput screening. Further pharmacological studies indicated that 5-FU exhibited potentiation effects on carbapenem antibiotics against 42 Gram-negative bacteria producing either metallo-β-lactamases (MBLs), such as NDM and IMP, or serine β-lactamases (Ser-BLs), like KPC and OXA. These bacteria included E. coli, Klebsiella pneumoniae, Pseudomonas aeruginosa and Acinetobacter spp., 32 of which were obtained from human clinical samples. Mechanistic investigations revealed that 5-FU inhibited the transcription and expression of the blaNDM-5 gene. In addition, 5-FU combined with meropenem enhanced bacterial metabolism, and stimulated the production of reactive oxygen species (ROS), thereby rendering bacteria more susceptible to meropenem. In a mouse systemic infection model, 5-FU combined with meropenem reduced bacterial loads and effectively elevated the survival rate of 83.3%, compared with 16.7% with meropenem monotherapy. Collectively, these findings indicate the potential of 5-FU as a novel meropenem adjuvant to improve treatment outcomes against infections caused by carbapenem-resistant bacteria.
Collapse
Affiliation(s)
- Muchen Zhang
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China agricultural University, Beijing, China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - Siyuan Yang
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China agricultural University, Beijing, China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - Yongqing Liu
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China agricultural University, Beijing, China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - Zhiyu Zou
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China agricultural University, Beijing, China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - Yan Zhang
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China agricultural University, Beijing, China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - Yunrui Tian
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China agricultural University, Beijing, China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - Rong Zhang
- The Second Affiliated Hospital of Zhejiang University, Zhejiang University, Hangzhou, China
| | - Dejun Liu
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China agricultural University, Beijing, China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - Congming Wu
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China agricultural University, Beijing, China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - Jianzhong Shen
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China agricultural University, Beijing, China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - Huangwei Song
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China.
| | - Yang Wang
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China agricultural University, Beijing, China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China.
| |
Collapse
|
12
|
Peterson E, Söderström B, Prins N, Le GHB, Hartley-Tassell LE, Evenhuis C, Grønnemose RB, Andersen TE, Møller-Jensen J, Iosifidis G, Duggin IG, Saunders B, Harry EJ, Bottomley AL. The role of bacterial size, shape and surface in macrophage engulfment of uropathogenic E. coli cells. PLoS Pathog 2024; 20:e1012458. [PMID: 39241059 PMCID: PMC11410268 DOI: 10.1371/journal.ppat.1012458] [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: 06/29/2023] [Revised: 09/18/2024] [Accepted: 07/26/2024] [Indexed: 09/08/2024] Open
Abstract
Uropathogenic Escherichia coli (UPEC) can undergo extensive filamentation in the host during acute urinary tract infections (UTIs). It has been hypothesised that this morphological plasticity allows bacteria to avoid host immune responses such as macrophage engulfment. However, it is still unclear what properties of filaments are important in macrophage-bacteria interactions. The aim of this work was to investigate the contribution of bacterial biophysical parameters, such as cell size and shape, and physiological parameters, such as cell surface and the environment, to macrophage engulfment efficiency. Viable, reversible filaments of known lengths and volumes were produced in the UPEC strain UTI89 using a variety of methods, including exposure to cell-wall targeting antibiotics, genetic manipulation and isolation from an in vitro human bladder cell model. Quantification of the engulfment ability of macrophages using gentamicin-protection assays and fluorescence microscopy demonstrated that the ability of filaments to avoid macrophage engulfment is dependent on a combination of size (length and volume), shape, cell surface and external environmental factors. UTI89 filamentation and macrophage engulfment efficiency were also found to occur independently of the SOS-inducible filamentation genes, sulA and ymfM in both in vivo and in vitro models of infection. Compared to filaments formed via antibiotic inhibition of division, the infection-derived filaments were preferentially targeted by macrophages. With several strains of UPEC now resistant to current antibiotics, our work identifies the importance of bacterial physiological and morphological states during infection.
Collapse
Affiliation(s)
- Elizabeth Peterson
- Australian Institute for Microbiology & Infection, University of Technology Sydney, Australia
| | - Bill Söderström
- Australian Institute for Microbiology & Infection, University of Technology Sydney, Australia
| | - Nienke Prins
- Australian Institute for Microbiology & Infection, University of Technology Sydney, Australia
| | - Giang H B Le
- School of Life Sciences, University of Technology Sydney, Sydney, Australia
| | | | - Chris Evenhuis
- Australian Institute for Microbiology & Infection, University of Technology Sydney, Australia
| | - Rasmus Birkholm Grønnemose
- Research Unit of Clinical Microbiology, University of Southern Denmark and Odense University Hospital, Odense, Denmark
| | - Thomas Emil Andersen
- Research Unit of Clinical Microbiology, University of Southern Denmark and Odense University Hospital, Odense, Denmark
| | - Jakob Møller-Jensen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Gregory Iosifidis
- Australian Institute for Microbiology & Infection, University of Technology Sydney, Australia
| | - Iain G Duggin
- Australian Institute for Microbiology & Infection, University of Technology Sydney, Australia
| | | | - Elizabeth J Harry
- Australian Institute for Microbiology & Infection, University of Technology Sydney, Australia
| | - Amy L Bottomley
- Australian Institute for Microbiology & Infection, University of Technology Sydney, Australia
| |
Collapse
|
13
|
Nilson R, Penumutchu S, Pagano FS, Belenky P. Metabolic changes associated with polysaccharide utilization reduce susceptibility to some β-lactams in Bacteroides thetaiotaomicron. mSphere 2024; 9:e0010324. [PMID: 39109911 PMCID: PMC11351048 DOI: 10.1128/msphere.00103-24] [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: 04/29/2024] [Accepted: 07/07/2024] [Indexed: 08/29/2024] Open
Abstract
Antibiotic therapy alters bacterial abundance and metabolism in the gut microbiome, leading to dysbiosis and opportunistic infections. Bacteroides thetaiotaomicron (Bth) is both a commensal in the gut and an opportunistic pathogen in other body sites. Past work has shown that Bth responds to β-lactam treatment differently depending on the metabolic environment both in vitro and in vivo. Studies of other bacteria show that an increase in respiratory metabolism independent of growth rate promotes susceptibility to bactericidal antibiotics. We propose that Bth enters a protected state linked to an increase in polysaccharide utilization and a decrease in the use of simple sugars. Here, we apply antibiotic susceptibility testing, transcriptomic analysis, and genetic manipulation to characterize this polysaccharide-mediated tolerance (PM tolerance) phenotype. We found that a variety of mono- and disaccharides increased the susceptibility of Bth to several different β-lactams compared to polysaccharides. Transcriptomics indicated a metabolic shift from reductive to oxidative branches of the tricarboxylic acid cycle on polysaccharides. Accordingly, supplementation with intermediates of central carbon metabolism had varying effects on PM tolerance. Transcriptional analysis also showed a decrease in the expression of the electron transport chain (ETC) protein NQR and an increase in the ETC protein NUO, when given fiber versus glucose. Deletion of NQR increased Bth susceptibility while deletion of NUO and a third ETC protein NDH2 had no effect. This work confirms that carbon source utilization modulates antibiotic susceptibility in Bth and that anaerobic respiratory metabolism and the ETC play an essential role.IMPORTANCEAntibiotics are indispensable medications that revolutionized modern medicine. However, their effectiveness is challenged by a large array of resistance and tolerance mechanisms. Treatment with antibiotics also disrupts the gut microbiome which can adversely affect health. Bacteroides are prevalent in the gut microbiome and yet are frequently involved in anaerobic infections. Thus, understanding how antibiotics affect these bacteria is necessary to implement proper treatment. Recent work has investigated the role of metabolism in antibiotic susceptibility in distantly related bacteria such as Escherichia coli. Using antibiotic susceptibility testing, transcriptomics, and genetic manipulation, we demonstrate that polysaccharides reduce β-lactam susceptibility when compared to monosaccharides. This finding underscores the profound impact of metabolic adaptation on the therapeutic efficacy of antibiotics. In the long term, this work indicates that modulation of metabolism could make Bacteroides more susceptible during infections or protect them in the context of the microbiome.
Collapse
Affiliation(s)
- Rachael Nilson
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| | - Swathi Penumutchu
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| | - Francesco S. Pagano
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| | - Peter Belenky
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| |
Collapse
|
14
|
Dubey AK, Sardana D, Verma T, Alam P, Chattopadhyay A, Nandini SS, Khamari B, Bulagonda EP, Sen S, Nandi D. Quantifying Membrane Alterations with Tailored Fluorescent Dyes: A Rapid Antibiotic Resistance Profiling Methodology. ACS Infect Dis 2024; 10:2836-2859. [PMID: 39024306 DOI: 10.1021/acsinfecdis.4c00249] [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: 07/20/2024]
Abstract
Accurate detection of bacterial antibiotic sensitivity is crucial for theranostics and the containment of antibiotic-resistant infections. However, the intricate task of detecting and quantifying the antibiotic-induced changes in the bacterial cytoplasmic membrane, and their correlation with other metabolic pathways leading to antibiotic resistance, poses significant challenges. Using a novel class of 4-aminophthalimide (4AP)-based fluorescent dyes with precisely tailored alkyl chains, namely 4AP-C9 and 4AP-C13, we quantify stress-mediated alterations in E. coli membranes. Leveraging the unique depth-dependent positioning and environment-sensitive fluorescence properties of these dyes, we detect antibiotic-induced membrane damage through single-cell imaging and monitoring the fluorescence peak maxima difference ratio (PMDR) of the dyes within the bacterial membrane, complemented by other methods. The correlation between the ROS-induced cytoplasmic membrane damage and the PMDR of dyes quantifies sensitivity against bactericidal antibiotics, which correlates to antibiotic-induced lipid peroxidation. Significantly, our findings largely extend to clinical isolates of E. coli and other ESKAPE pathogens like K. pneumoniae and Enterobacter subspecies. Our data reveal that 4AP-Cn probes can potentially act as precise scales to detect antibiotic-induced membrane damage ("thinning") occurring at a subnanometer scale through the quantification of dyes' PMDR, making them promising membrane dyes for rapid detection of bacterial antibiotic resistance, distinguishing sensitive and resistant infections with high specificity in a clinical setup.
Collapse
Affiliation(s)
- Ashim Kumar Dubey
- Undergraduate Programme, Indian Institute of Science, Bangalore 560012, Karnataka, India
| | - Deepika Sardana
- School of Physical Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Taru Verma
- Centre for BioSystems, Science and Engineering, Indian Institute of Science, Bangalore 560012, Karnataka, India
| | - Parvez Alam
- School of Physical Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Avik Chattopadhyay
- Department of Biochemistry, Indian Institute of Science, Bangalore 560012, Karnataka, India
| | - Santhi Sanil Nandini
- Department of Biochemistry, Indian Institute of Science, Bangalore 560012, Karnataka, India
| | - Balaram Khamari
- Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Puttaparthi 515134, Andhra Pradesh, India
| | - Eswarappa Pradeep Bulagonda
- Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Puttaparthi 515134, Andhra Pradesh, India
| | - Sobhan Sen
- School of Physical Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Dipankar Nandi
- Department of Biochemistry, Indian Institute of Science, Bangalore 560012, Karnataka, India
| |
Collapse
|
15
|
Li B, Srivastava S, Shaikh M, Mereddy G, Garcia MR, Shah A, Ofori-Anyinam N, Chu T, Cheney N, Yang JH. Bioenergetic stress potentiates antimicrobial resistance and persistence. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.12.603336. [PMID: 39026737 PMCID: PMC11257553 DOI: 10.1101/2024.07.12.603336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Antimicrobial resistance (AMR) is a global health crisis and there is an urgent need to better understand AMR mechanisms. Antibiotic treatment alters several aspects of bacterial physiology, including increased ATP utilization, carbon metabolism, and reactive oxygen species (ROS) formation. However, how the "bioenergetic stress" induced by increased ATP utilization affects treatment outcomes is unknown. Here we utilized a synthetic biology approach to study the direct effects of bioenergetic stress on antibiotic efficacy. We engineered a genetic system that constitutively hydrolyzes ATP or NADH in Escherichia coli. We found that bioenergetic stress potentiates AMR evolution via enhanced ROS production, mutagenic break repair, and transcription-coupled repair. We also find that bioenergetic stress potentiates antimicrobial persistence via potentiated stringent response activation. We propose a unifying model that antibiotic-induced antimicrobial resistance and persistence is caused by antibiotic-induced. This has important implications for preventing or curbing the spread of AMR infections.
Collapse
|
16
|
Yu T, Huang J, Huang X, Hao J, Zhang P, Guo T, Bao G, Li G. Sub-MIC antibiotics increased the fitness cost of CRISPR-Cas in Acinetobacter baumannii. Front Microbiol 2024; 15:1381749. [PMID: 39011146 PMCID: PMC11246858 DOI: 10.3389/fmicb.2024.1381749] [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: 02/05/2024] [Accepted: 06/04/2024] [Indexed: 07/17/2024] Open
Abstract
Introduction The escalating prevalence of bacterial resistance, particularly multidrug-resistant bacteria like Acinetobacter baumannii, has become a significant global public health concern. The CRISPR-Cas system, a crucial defense mechanism in bacteria against foreign genetic elements, provides a competitive advantage. Type I-Fb and Type I-Fa are two subtypes of CRISPR-Cas systems that were found in A. baumannii, and the I-Fb CRISPR-Cas system regulates antibiotic resistance in A. baumannii. However, it is noteworthy that a majority of clinical isolates of A. baumannii lack or have incomplete CRISPR-Cas systems and most of them are multidrug-resistant. In light of this, our study aimed to examine the impact of antibiotic pressure on the fitness cost of the I-Fb CRISPR-Cas system in A. baumannii. Methods and Results In the study, we conducted in vitro competition experiments to investigate the influence of sub-minimum inhibitory concentration (sub-MIC) on the CRISPR-Cas systems' fitness cost in A. baumannii. We found that the fitness cost of the CRISPR-Cas system was increased under sub-MIC conditions. The expression of CRISPR-Cas-related genes was decreased, while the conjugation frequency was increased in AB43 under sub-MIC conditions. Through metabolomic analysis, we identified that sub-MIC conditions primarily affected energy metabolism pathways. In particular, we observed increased carbon metabolism, nitrogen metabolism, and intracellular ATP. Notably, the CRISPR-Cas system demonstrated resistance to the efflux pump-mediated resistance. Furthermore, the expression of efflux pump-related genes was increased under sub-MIC conditions. Conclusion Our findings suggest that the I-Fb CRISPR-Cas system confers a significant competitive advantage in A. baumanni. However, under sub-MIC conditions, its function and the ability to inhibit the energy required for efflux pumps are reduced, resulting in an increased fitness cost and loss of competitive advantage.
Collapse
Affiliation(s)
- Ting Yu
- Department of Microbiology, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Department of Laboratory Medicine, Affiliated Hospital, Yangzhou University, Yangzhou, China
| | - Jiayuan Huang
- Department of Microbiology, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Xinyue Huang
- Department of Microbiology, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Jingchen Hao
- Department of Microbiology, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Pengyu Zhang
- Department of Microbiology, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Tingting Guo
- Department of Microbiology, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Department of Laboratory Medicine, Affiliated Hospital, Yangzhou University, Yangzhou, China
| | - Guangyu Bao
- Department of Laboratory Medicine, Affiliated Hospital, Yangzhou University, Yangzhou, China
| | - Guocai Li
- Department of Microbiology, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Department of Laboratory Medicine, Affiliated Hospital, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College/Guangling College, Yangzhou University, Yangzhou, China
| |
Collapse
|
17
|
Liu M, Wang M, Huang M, Gao Q, Zhu D, Wang M, Jia R, Chen S, Zhao X, Yang Q, Wu Y, Zhang S, Huang J, Ou X, Mao S, Tian B, Sun D, Cheng A. Iron efflux by IetA enhances β-lactam aztreonam resistance and is linked to oxidative stress through cellular respiration in Riemerella anatipestifer. J Antimicrob Chemother 2024; 79:1385-1396. [PMID: 38629469 DOI: 10.1093/jac/dkae114] [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: 07/07/2023] [Accepted: 03/20/2024] [Indexed: 06/04/2024] Open
Abstract
BACKGROUND Riemerella anatipestifer encodes an iron acquisition system, but whether it encodes the iron efflux pump and its role in antibiotic resistance are largely unknown. OBJECTIVES To screen and identify an iron efflux gene in R. anatipestifer and determine whether and how the iron efflux gene is involved in antibiotic resistance. METHODS In this study, gene knockout, streptonigrin susceptibility assay and inductively coupled plasma mass spectrometry were used to screen for the iron efflux gene ietA. The MIC measurements, scanning electron microscopy and reactive oxygen species (ROS) detection were used to verify the role of IetA in aztreonam resistance and its mechanism. Mortality and colonization assay were used to investigate the role of IetA in virulence. RESULTS The deletion mutant ΔietA showed heightened susceptibility to streptonigrin, and prominent intracellular iron accumulation was observed in ΔfurΔietA under excess iron conditions. Additionally, ΔietA exhibited increased sensitivity to H2O2-produced oxidative stress. Under aerobic conditions with abundant iron, ΔietA displayed increased susceptibility to the β-lactam antibiotic aztreonam due to heightened ROS production. However, the killing efficacy of aztreonam was diminished in both WT and ΔietA under anaerobic or iron restriction conditions. Further experiments demonstrated that the efficiency of aztreonam against ΔietA was dependent on respiratory complexes Ⅰ and Ⅱ. Finally, in a duckling model, ΔietA had reduced virulence compared with the WT. CONCLUSION Iron efflux is critical to alleviate oxidative stress damage and β-lactam aztreonam killing in R. anatipestifer, which is linked by cellular respiration.
Collapse
Affiliation(s)
- Mafeng Liu
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu 611130, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu 611130, China
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Mengying Wang
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu 611130, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu 611130, China
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Mi Huang
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu 611130, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu 611130, China
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Qun Gao
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu 611130, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu 611130, China
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Dekang Zhu
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu 611130, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu 611130, China
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Mingshu Wang
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu 611130, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu 611130, China
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Renyong Jia
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu 611130, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu 611130, China
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Shun Chen
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu 611130, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu 611130, China
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Xinxin Zhao
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu 611130, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu 611130, China
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Qiao Yang
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu 611130, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu 611130, China
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Ying Wu
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu 611130, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu 611130, China
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Shaqiu Zhang
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu 611130, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu 611130, China
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Juan Huang
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu 611130, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu 611130, China
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Xumin Ou
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu 611130, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu 611130, China
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Sai Mao
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu 611130, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu 611130, China
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Bin Tian
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu 611130, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu 611130, China
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Di Sun
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu 611130, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu 611130, China
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Anchun Cheng
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu 611130, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu 611130, China
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| |
Collapse
|
18
|
Yang L, Yu P, Wang J, Zhao T, Zhao Y, Pan Y, Chen L. Genomic and Transcriptomic Analyses Reveal Multiple Strategies for Vibrio parahaemolyticus to Tolerate Sub-Lethal Concentrations of Three Antibiotics. Foods 2024; 13:1674. [PMID: 38890902 PMCID: PMC11171697 DOI: 10.3390/foods13111674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 05/16/2024] [Accepted: 05/22/2024] [Indexed: 06/20/2024] Open
Abstract
Vibrio parahaemolyticus can cause acute gastroenteritis, wound infections, and septicemia in humans. The overuse of antibiotics in aquaculture may lead to a high incidence of the multidrug-resistant (MDR) pathogen. Nevertheless, the genome evolution of V. parahaemolyticus in aquatic animals and the mechanism of its antibiotic tolerance remain to be further deciphered. Here, we investigated the molecular basis of the antibiotic tolerance of V. parahaemolyticus isolates (n = 3) originated from shellfish and crustaceans using comparative genomic and transcriptomic analyses. The genome sequences of the V. parahaemolyticus isolates were determined (5.0-5.3 Mb), and they contained 4709-5610 predicted protein-encoding genes, of which 823-1099 genes were of unknown functions. Comparative genomic analyses revealed a number of mobile genetic elements (MGEs, n = 69), antibiotic resistance-related genes (n = 7-9), and heavy metal tolerance-related genes (n = 2-4). The V. parahaemolyticus isolates were resistant to sub-lethal concentrations (sub-LCs) of ampicillin (AMP, 512 μg/mL), kanamycin (KAN, 64 μg/mL), and streptomycin (STR, 16 μg/mL) (p < 0.05). Comparative transcriptomic analyses revealed that there were significantly altered metabolic pathways elicited by the sub-LCs of the antibiotics (p < 0.05), suggesting the existence of multiple strategies for antibiotic tolerance in V. parahaemolyticus. The results of this study enriched the V. parahaemolyticus genome database and should be useful for controlling the MDR pathogen worldwide.
Collapse
Affiliation(s)
- Lianzhi Yang
- Key Laboratory of Quality and Safety Risk Assessment for Aquatic Products on Storage and Preservation (Shanghai), Ministry of Agriculture and Rural Affairs of China, Shanghai 201306, China; (L.Y.); (P.Y.)
- College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Pan Yu
- Key Laboratory of Quality and Safety Risk Assessment for Aquatic Products on Storage and Preservation (Shanghai), Ministry of Agriculture and Rural Affairs of China, Shanghai 201306, China; (L.Y.); (P.Y.)
- College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Juanjuan Wang
- Key Laboratory of Quality and Safety Risk Assessment for Aquatic Products on Storage and Preservation (Shanghai), Ministry of Agriculture and Rural Affairs of China, Shanghai 201306, China; (L.Y.); (P.Y.)
- College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Taixia Zhao
- Key Laboratory of Quality and Safety Risk Assessment for Aquatic Products on Storage and Preservation (Shanghai), Ministry of Agriculture and Rural Affairs of China, Shanghai 201306, China; (L.Y.); (P.Y.)
- College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
- College of Tea and Food Science, Wuyi University, Wuyishan 354300, China
| | - Yong Zhao
- Key Laboratory of Quality and Safety Risk Assessment for Aquatic Products on Storage and Preservation (Shanghai), Ministry of Agriculture and Rural Affairs of China, Shanghai 201306, China; (L.Y.); (P.Y.)
- College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Yingjie Pan
- Key Laboratory of Quality and Safety Risk Assessment for Aquatic Products on Storage and Preservation (Shanghai), Ministry of Agriculture and Rural Affairs of China, Shanghai 201306, China; (L.Y.); (P.Y.)
- College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Lanming Chen
- Key Laboratory of Quality and Safety Risk Assessment for Aquatic Products on Storage and Preservation (Shanghai), Ministry of Agriculture and Rural Affairs of China, Shanghai 201306, China; (L.Y.); (P.Y.)
- College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| |
Collapse
|
19
|
Voedts H, Anoyatis-Pelé C, Langella O, Rusconi F, Hugonnet JE, Arthur M. (p)ppGpp modifies RNAP function to confer β-lactam resistance in a peptidoglycan-independent manner. Nat Microbiol 2024; 9:647-656. [PMID: 38443580 DOI: 10.1038/s41564-024-01609-w] [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: 02/21/2023] [Accepted: 01/16/2024] [Indexed: 03/07/2024]
Abstract
(p)ppGpp is a nucleotide alarmone that controls bacterial response to nutrient deprivation. Since elevated (p)ppGpp levels confer mecillinam resistance and are essential for broad-spectrum β-lactam resistance as mediated by the β-lactam-insensitive transpeptidase YcbB (LdtD), we hypothesized that (p)ppGpp might affect cell wall peptidoglycan metabolism. Here we report that (p)ppGpp-dependent β-lactam resistance does not rely on any modification of peptidoglycan metabolism, as established by analysis of Escherichia coli peptidoglycan structure using high-resolution mass spectrometry. Amino acid substitutions in the β or β' RNA polymerase (RNAP) subunits, alone or in combination with the CRISPR interference-mediated downregulation of three of seven ribosomal RNA operons, were sufficient for resistance, although β-lactams have no known impact on the RNAP or ribosomes. This implies that modifications of RNAP and ribosome functions are critical to prevent downstream effects of the inactivation of peptidoglycan transpeptidases by β-lactams.
Collapse
Affiliation(s)
- Henri Voedts
- Centre de Recherche des Cordeliers, Sorbonne Université, INSERM, Université Paris Cité, Paris, France
| | - Constantin Anoyatis-Pelé
- Centre de Recherche des Cordeliers, Sorbonne Université, INSERM, Université Paris Cité, Paris, France
| | - Olivier Langella
- GQE-Le Moulon/PAPPSO, Université Paris-Saclay, INRAE, CNRS, AgroParisTech, IDEEV, Gif-sur-Yvette, France
| | - Filippo Rusconi
- Centre de Recherche des Cordeliers, Sorbonne Université, INSERM, Université Paris Cité, Paris, France
- GQE-Le Moulon/PAPPSO, Université Paris-Saclay, INRAE, CNRS, AgroParisTech, IDEEV, Gif-sur-Yvette, France
| | - Jean-Emmanuel Hugonnet
- Centre de Recherche des Cordeliers, Sorbonne Université, INSERM, Université Paris Cité, Paris, France.
| | - Michel Arthur
- Centre de Recherche des Cordeliers, Sorbonne Université, INSERM, Université Paris Cité, Paris, France.
| |
Collapse
|
20
|
Gupta R, Singh M, Pathania R. Chemical genetic approaches for the discovery of bacterial cell wall inhibitors. RSC Med Chem 2023; 14:2125-2154. [PMID: 37974958 PMCID: PMC10650376 DOI: 10.1039/d3md00143a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Accepted: 08/10/2023] [Indexed: 11/19/2023] Open
Abstract
Antimicrobial resistance (AMR) in bacterial pathogens is a worldwide health issue. The innovation gap in discovering new antibiotics has remained a significant hurdle in combating the AMR problem. Currently, antibiotics target various vital components of the bacterial cell envelope, nucleic acid and protein biosynthesis machinery and metabolic pathways essential for bacterial survival. The critical role of the bacterial cell envelope in cell morphogenesis and integrity makes it an attractive drug target. While a significant number of in-clinic antibiotics target peptidoglycan biosynthesis, several components of the bacterial cell envelope have been overlooked. This review focuses on various antibacterial targets in the bacterial cell wall and the strategies employed to find their novel inhibitors. This review will further elaborate on combining forward and reverse chemical genetic approaches to discover antibacterials that target the bacterial cell envelope.
Collapse
Affiliation(s)
- Rinki Gupta
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee Roorkee - 247 667 Uttarakhand India
| | - Mangal Singh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee Roorkee - 247 667 Uttarakhand India
| | - Ranjana Pathania
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee Roorkee - 247 667 Uttarakhand India
| |
Collapse
|
21
|
Yaeger LN, French S, Brown ED, Côté JP, Burrows LL. Central metabolism is a key player in E. coli biofilm stimulation by sub-MIC antibiotics. PLoS Genet 2023; 19:e1011013. [PMID: 37917668 PMCID: PMC10645362 DOI: 10.1371/journal.pgen.1011013] [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: 07/22/2023] [Revised: 11/14/2023] [Accepted: 10/10/2023] [Indexed: 11/04/2023] Open
Abstract
Exposure of Escherichia coli to sub-inhibitory antibiotics stimulates biofilm formation through poorly characterized mechanisms. Using a high-throughput Congo Red binding assay to report on biofilm matrix production, we screened ~4000 E. coli K12 deletion mutants for deficiencies in this biofilm stimulation response. We screened using three different antibiotics to identify core components of the biofilm stimulation response. Mutants lacking acnA, nuoE, or lpdA failed to respond to sub-MIC cefixime and novobiocin, implicating central metabolism and aerobic respiration in biofilm stimulation. These genes are members of the ArcA/B regulon-controlled by a respiration-sensitive two-component system. Mutants of arcA and arcB had a 'pre-activated' phenotype, where biofilm formation was already high relative to wild type in vehicle control conditions, and failed to increase further with the addition of sub-MIC cefixime. Using a tetrazolium dye and an in vivo NADH sensor, we showed spatial co-localization of increased metabolic activity with sub-lethal concentrations of the bactericidal antibiotics cefixime and novobiocin. Supporting a role for respiratory stress, the biofilm stimulation response to cefixime and novobiocin was inhibited when nitrate was provided as an alternative electron acceptor. Deletion of a gene encoding part of the machinery for respiring nitrate abolished its ameliorating effects, and nitrate respiration increased during growth with sub-MIC cefixime. Finally, in probing the generalizability of biofilm stimulation, we found that the stimulation response to translation inhibitors, unlike other antibiotic classes, was minimally affected by nitrate supplementation, suggesting that targeting the ribosome stimulates biofilm formation in distinct ways. By characterizing the biofilm stimulation response to sub-MIC antibiotics at a systems level, we identified multiple avenues for design of therapeutics that impair bacterial stress management.
Collapse
Affiliation(s)
- Luke N. Yaeger
- Department of Biochemistry and Biomedical Sciences, and the Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Shawn French
- Department of Biochemistry and Biomedical Sciences, and the Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Eric D. Brown
- Department of Biochemistry and Biomedical Sciences, and the Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Jean Philippe Côté
- Department of Biochemistry and Biomedical Sciences, and the Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
- Département de Biologie, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Lori L. Burrows
- Department of Biochemistry and Biomedical Sciences, and the Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
22
|
Peng B, Li H, Peng XX. Call for next-generation drugs that remove the uptake barrier to combat antibiotic resistance. Drug Discov Today 2023; 28:103753. [PMID: 37640151 DOI: 10.1016/j.drudis.2023.103753] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/15/2023] [Accepted: 08/23/2023] [Indexed: 08/31/2023]
Abstract
Existing antibacterial agents can be categorized into two generations, but bacterial insensitivity towards both of these generations poses a serious public health challenge worldwide. Thus, novel approaches and/or novel antibacterials are urgently needed to maintain a concentration of antibacterials that is lethal to bacteria that are resistant to existing antibiotic treatments. Metabolite(s)-based adjuvants that promote antibiotic uptake and enhance antibiotic efficacy are an effective strategy that is unlikely to develop resistance. Thus, we propose a metabolite(s)-based approach, in which metabolites and antibacterials are combined, as a promising strategy for the development of next-generation agents to combat a variety of antibiotic-resistant pathogens.
Collapse
Affiliation(s)
- Bo Peng
- State Key Laboratory of Bio-Control, School of Life Sciences, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, Sun Yat-sen University, University City, Guangzhou 510006, People's Republic of China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266071, People's Republic of China
| | - Hui Li
- State Key Laboratory of Bio-Control, School of Life Sciences, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, Sun Yat-sen University, University City, Guangzhou 510006, People's Republic of China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266071, People's Republic of China
| | - Xuan-Xian Peng
- State Key Laboratory of Bio-Control, School of Life Sciences, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, Sun Yat-sen University, University City, Guangzhou 510006, People's Republic of China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266071, People's Republic of China.
| |
Collapse
|
23
|
Kawai Y, Kawai M, Mackenzie ES, Dashti Y, Kepplinger B, Waldron KJ, Errington J. On the mechanisms of lysis triggered by perturbations of bacterial cell wall biosynthesis. Nat Commun 2023; 14:4123. [PMID: 37433811 DOI: 10.1038/s41467-023-39723-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 06/20/2023] [Indexed: 07/13/2023] Open
Abstract
Inhibition of bacterial cell wall synthesis by antibiotics such as β-lactams is thought to cause explosive lysis through loss of cell wall integrity. However, recent studies on a wide range of bacteria have suggested that these antibiotics also perturb central carbon metabolism, contributing to death via oxidative damage. Here, we genetically dissect this connection in Bacillus subtilis perturbed for cell wall synthesis, and identify key enzymatic steps in upstream and downstream pathways that stimulate the generation of reactive oxygen species through cellular respiration. Our results also reveal the critical role of iron homeostasis for the oxidative damage-mediated lethal effects. We show that protection of cells from oxygen radicals via a recently discovered siderophore-like compound uncouples changes in cell morphology normally associated with cell death, from lysis as usually judged by a phase pale microscopic appearance. Phase paling appears to be closely associated with lipid peroxidation.
Collapse
Affiliation(s)
- Yoshikazu Kawai
- Centre for Bacterial Cell Biology, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Richardson Road, Newcastle upon Tyne, NE2 4AX, UK.
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, 2006, Australia.
| | - Maki Kawai
- Centre for Bacterial Cell Biology, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Richardson Road, Newcastle upon Tyne, NE2 4AX, UK
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, 2006, Australia
| | - Eilidh Sohini Mackenzie
- Bioscience Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Yousef Dashti
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, 2006, Australia
| | - Bernhard Kepplinger
- Department of Molecular Microbiology, Faculty of Biotechnology, University of Wrocław, 50-383, Wrocław, Poland
| | - Kevin John Waldron
- Bioscience Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, 02-106, Poland
| | - Jeff Errington
- Centre for Bacterial Cell Biology, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Richardson Road, Newcastle upon Tyne, NE2 4AX, UK.
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, 2006, Australia.
| |
Collapse
|
24
|
Keller MR, Dörr T. Bacterial metabolism and susceptibility to cell wall-active antibiotics. Adv Microb Physiol 2023; 83:181-219. [PMID: 37507159 PMCID: PMC11024984 DOI: 10.1016/bs.ampbs.2023.04.002] [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: 07/30/2023]
Abstract
Bacterial infections are increasingly resistant to antimicrobial therapy. Intense research focus has thus been placed on identifying the mechanisms that bacteria use to resist killing or growth inhibition by antibiotics and the ways in which bacteria share these traits with one another. This work has led to the advancement of new drugs, combination therapy regimens, and a deeper appreciation for the adaptability seen in microorganisms. However, while the primary mechanisms of action of most antibiotics are well understood, the more subtle contributions of bacterial metabolic state to repairing or preventing damage caused by antimicrobials (thereby promoting survival) are still understudied. Here, we review a modern viewpoint on a classical system: examining bacterial metabolism's connection to antibiotic susceptibility. We dive into the relationship between metabolism and antibiotic efficacy through the lens of growth rate, energy state, resource allocation, and the infection environment, focusing on cell wall-active antibiotics.
Collapse
Affiliation(s)
- Megan Renee Keller
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, United States
| | - Tobias Dörr
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, United States; Department of Microbiology, Cornell University, Ithaca, NY, United States; Cornell Institute of Host-Microbe Interactions and Disease, Cornell University, Ithaca, NY, United States.
| |
Collapse
|
25
|
Patel Y, Soni V, Rhee KY, Helmann JD. Mutations in rpoB That Confer Rifampicin Resistance Can Alter Levels of Peptidoglycan Precursors and Affect β-Lactam Susceptibility. mBio 2023; 14:e0316822. [PMID: 36779708 PMCID: PMC10128067 DOI: 10.1128/mbio.03168-22] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 01/23/2023] [Indexed: 02/14/2023] Open
Abstract
Bacteria can adapt to stressful conditions through mutations affecting the RNA polymerase core subunits that lead to beneficial changes in transcription. In response to selection with rifampicin (RIF), mutations arise in the RIF resistance-determining region (RRDR) of rpoB that reduce antibiotic binding. These changes can also alter transcription and thereby have pleiotropic effects on bacterial fitness. Here, we studied the evolution of resistance in Bacillus subtilis to the synergistic combination of RIF and the β-lactam cefuroxime (CEF). Two independent evolution experiments led to the recovery of a single rpoB allele (S487L) that was able to confer resistance to RIF and CEF through a single mutation. Two other common RRDR mutations made the cells 32 times more sensitive to CEF (H482Y) or led to only modest CEF resistance (Q469R). The diverse effects of these three mutations on CEF resistance are correlated with differences in the expression of peptidoglycan (PG) synthesis genes and in the levels of two metabolites crucial in regulating PG synthesis, glucosamine-6-phosphate (GlcN-6-P) and UDP-N-acetylglucosamine (UDP-GlcNAc). We conclude that RRDR mutations can have widely varying effects on pathways important for cell wall biosynthesis, and this may restrict the spectrum of mutations that arise during combination therapy. IMPORTANCE Rifampicin (RIF) is one of the most valued drugs in the treatment of tuberculosis. TB treatment relies on a combination therapy and for multidrug-resistant strains may include β-lactams. Mutations in rpoB present a common route for emergence of resistance to RIF. In this study, using B. subtilis as a model, we evaluate the emergence of resistance for the synergistic combination of RIF and the β-lactam cefuroxime (CEF). One clinically relevant rpoB mutation conferred resistance to both RIF and CEF, whereas one other increased CEF sensitivity. We were able to link these CEF sensitivity phenotypes to accumulation of UDP-N-acetylglucosamine (UDP-GlcNAc), which feedback regulates GlmS activity and thereby peptidoglycan synthesis. Further, we found that higher CEF concentrations precluded the emergence of high RIF resistance. Collectively, these results suggest that multidrug treatment regimens may limit the available pathways for the evolution of antibiotic resistance.
Collapse
Affiliation(s)
- Yesha Patel
- Department of Microbiology, Cornell University, Ithaca, New York, USA
| | - Vijay Soni
- Department of Medicine, Division of Infectious Diseases, Weill Cornell Medicine, New York, New York, USA
| | - Kyu Y. Rhee
- Department of Medicine, Division of Infectious Diseases, Weill Cornell Medicine, New York, New York, USA
| | - John D. Helmann
- Department of Microbiology, Cornell University, Ithaca, New York, USA
| |
Collapse
|
26
|
Han Y, Jiang N, Xu H, Yuan Z, Xiu J, Mao S, Liu X, Huang J. Extracellular Matrix Rigidities Regulate the Tricarboxylic Acid Cycle and Antibiotic Resistance of Three-Dimensionally Confined Bacterial Microcolonies. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206153. [PMID: 36658695 PMCID: PMC10037996 DOI: 10.1002/advs.202206153] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/22/2022] [Indexed: 06/06/2023]
Abstract
As a major cause of clinical chronic infection, microbial biofilms/microcolonies in host tissues essentially live in 3D-constrained microenvironments, which potentially modulate their spatial self-organization and morphodynamics. However, it still remains unclear whether and how mechanical cues of 3D confined microenvironments, for example, extracellular matrix (ECM) stiffness, exert an impact on antibiotic resistance of bacterial biofilms/microcolonies. With a high-throughput antibiotic sensitivity testing (AST) platform, it is revealed that 3D ECM rigidities greatly modulate their resistance to diverse antibiotics. The microcolonies in 3D ECM with human tissue-specific rigidities varying from 0.5 to 20 kPa show a ≈2-10 000-fold increase in minimum inhibitory concentration, depending on the types of antibiotics. The authors subsequently identified that the increase in 3D ECM rigidities leads to the downregulation of the tricarboxylic acid (TCA) cycle, which is responsible for enhanced antibiotic resistance. Further, it is shown that fumarate, as a potentiator of TCA cycle activity, can alleviate the elevated antibiotic resistance and thus remarkably improve the efficacy of antibiotics against bacterial microcolonies in 3D confined ECM, as confirmed in the chronic infection mice model. These findings suggest fumarate can be employed as an antibiotic adjuvant to effectively treat infections induced by bacterial biofilms/microcolonies in a 3D-confined environment.
Collapse
Affiliation(s)
- Yiming Han
- Department of Mechanics and Engineering Science, and Beijing Innovation Center for Engineering Science and Advanced TechnologyCollege of EngineeringPeking University100871BeijingChina
| | - Nan Jiang
- Department of Mechanics and Engineering Science, and Beijing Innovation Center for Engineering Science and Advanced TechnologyCollege of EngineeringPeking University100871BeijingChina
| | - Hongwei Xu
- Department of Mechanics and Engineering Science, and Beijing Innovation Center for Engineering Science and Advanced TechnologyCollege of EngineeringPeking University100871BeijingChina
| | - Zuoying Yuan
- Department of Mechanics and Engineering Science, and Beijing Innovation Center for Engineering Science and Advanced TechnologyCollege of EngineeringPeking University100871BeijingChina
| | - Jidong Xiu
- Department of Mechanics and Engineering Science, and Beijing Innovation Center for Engineering Science and Advanced TechnologyCollege of EngineeringPeking University100871BeijingChina
| | - Sheng Mao
- Department of Mechanics and Engineering Science, and Beijing Innovation Center for Engineering Science and Advanced TechnologyCollege of EngineeringPeking University100871BeijingChina
| | - Xiaozhi Liu
- Tianjin Key Laboratory of Epigenetics for Organ Development of Premature InfantsFifth Central Hospital of TianjinTianjin300450China
| | - Jianyong Huang
- Department of Mechanics and Engineering Science, and Beijing Innovation Center for Engineering Science and Advanced TechnologyCollege of EngineeringPeking University100871BeijingChina
| |
Collapse
|
27
|
Voedts H, Kennedy SP, Sezonov G, Arthur M, Hugonnet JE. Genome-wide identification of genes required for alternative peptidoglycan cross-linking in Escherichia coli revealed unexpected impacts of β-lactams. Nat Commun 2022; 13:7962. [PMID: 36575173 PMCID: PMC9794725 DOI: 10.1038/s41467-022-35528-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 12/06/2022] [Indexed: 12/28/2022] Open
Abstract
The D,D-transpeptidase activity of penicillin-binding proteins (PBPs) is the well-known primary target of β-lactam antibiotics that block peptidoglycan polymerization. β-lactam-induced bacterial killing involves complex downstream responses whose causes and consequences are difficult to resolve. Here, we use the functional replacement of PBPs by a β-lactam-insensitive L,D-transpeptidase to identify genes essential to mitigate the effects of PBP inactivation by β-lactams in actively dividing bacteria. The functions of the 179 conditionally essential genes identified by this approach extend far beyond L,D-transpeptidase partners for peptidoglycan polymerization to include proteins involved in stress response and in the assembly of outer membrane polymers. The unsuspected effects of β-lactams include loss of the lipoprotein-mediated covalent bond that links the outer membrane to the peptidoglycan, destabilization of the cell envelope in spite of effective peptidoglycan cross-linking, and increased permeability of the outer membrane. The latter effect indicates that the mode of action of β-lactams involves self-promoted penetration through the outer membrane.
Collapse
Affiliation(s)
- Henri Voedts
- grid.417925.cCentre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, F-75006 Paris, France
| | - Sean P. Kennedy
- Institut Pasteur, Université Paris Cité, Département Biologie Computationnelle, F-75015 Paris, France
| | - Guennadi Sezonov
- grid.417925.cCentre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, F-75006 Paris, France
| | - Michel Arthur
- grid.417925.cCentre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, F-75006 Paris, France
| | - Jean-Emmanuel Hugonnet
- grid.417925.cCentre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, F-75006 Paris, France
| |
Collapse
|
28
|
Thomson NM, Turner AK, Yasir M, Bastkowski S, Lott M, Webber MA, Charles IG. A whole-genome assay identifies four principal gene functions that confer tolerance of meropenem stress upon Escherichia coli. FRONTIERS IN ANTIBIOTICS 2022; 1:957942. [PMID: 39816415 PMCID: PMC11731830 DOI: 10.3389/frabi.2022.957942] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 08/23/2022] [Indexed: 01/18/2025]
Abstract
We report here the identification of four gene functions of principal importance for the tolerance of meropenem stress in Escherichia coli: cell division, cell envelope synthesis and maintenance, ATP metabolism, and transcription regulation. The primary mechanism of β-lactam antibiotics such as meropenem is inhibition of penicillin binding proteins, thus interfering with peptidoglycan crosslinking, weakening the cell envelope, and promoting cell lysis. However, recent systems biology approaches have revealed numerous downstream effects that are triggered by cell envelope damage and involve diverse cell processes. Subpopulations of persister cells can also arise, which can survive elevated concentrations of meropenem despite the absence of a specific resistance factor. We used Transposon-Directed Insertion Sequencing with inducible gene expression to simultaneously assay the effects of upregulation, downregulation, and disruption of every gene in a model E. coli strain on survival of exposure to four concentrations of meropenem. Automated Gene Functional Classification and manual categorization highlighted the importance at all meropenem concentrations of genes involved in peptidoglycan remodeling during cell division, suggesting that cell division is the primary function affected by meropenem. Genes involved in cell envelope synthesis and maintenance, ATP metabolism, and transcriptional regulation were generally important at higher meropenem concentrations, suggesting that these three functions are therefore secondary or downstream targets. Our analysis revealed the importance of multiple two-component signal transduction mechanisms, suggesting an as-yet unexplored coordinated transcriptional response to meropenem stress. The inclusion of an inducible, transposon-encoded promoter allowed sensitive detection of genes involved in proton transport, ATP production and tRNA synthesis, for which modulation of expression affects survival in the presence of meropenem: a finding that would not be possible with other technologies. We were also able to suggest new targets for future antibiotic development or for synergistic effects between gene or protein inhibitors and existing antibiotics. Overall, in a single massively parallel assay we were able to recapitulate many of the findings from decades of research into β-lactam antibiotics, add to the list of genes known to be important for meropenem tolerance, and categorize the four principal gene functions involved.
Collapse
Affiliation(s)
- Nicholas M. Thomson
- Microbes in the Food Chain, Quadram Institute Bioscience, Norwich, United Kingdom
| | - A. Keith Turner
- Microbes in the Food Chain, Quadram Institute Bioscience, Norwich, United Kingdom
| | - Muhammad Yasir
- Microbes in the Food Chain, Quadram Institute Bioscience, Norwich, United Kingdom
| | - Sarah Bastkowski
- Microbes in the Food Chain, Quadram Institute Bioscience, Norwich, United Kingdom
| | - Martin Lott
- Microbes in the Food Chain, Quadram Institute Bioscience, Norwich, United Kingdom
| | - Mark A. Webber
- Microbes in the Food Chain, Quadram Institute Bioscience, Norwich, United Kingdom
- Norwich Medical School, University of East Anglia, Norwich, United Kingdom
| | - Ian G. Charles
- Microbes in the Food Chain, Quadram Institute Bioscience, Norwich, United Kingdom
- Norwich Medical School, University of East Anglia, Norwich, United Kingdom
| |
Collapse
|
29
|
Wong F, Stokes JM, Bening SC, Vidoudez C, Trauger SA, Collins JJ. Reactive metabolic byproducts contribute to antibiotic lethality under anaerobic conditions. Mol Cell 2022; 82:3499-3512.e10. [PMID: 35973427 PMCID: PMC10149100 DOI: 10.1016/j.molcel.2022.07.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 05/19/2022] [Accepted: 07/17/2022] [Indexed: 01/21/2023]
Abstract
Understanding how bactericidal antibiotics kill bacteria remains an open question. Previous work has proposed that primary drug-target corruption leads to increased energetic demands, resulting in the generation of reactive metabolic byproducts (RMBs), particularly reactive oxygen species, that contribute to antibiotic-induced cell death. Studies have challenged this hypothesis by pointing to antibiotic lethality under anaerobic conditions. Here, we show that treatment of Escherichia coli with bactericidal antibiotics under anaerobic conditions leads to changes in the intracellular concentrations of central carbon metabolites, as well as the production of RMBs, particularly reactive electrophilic species (RES). We show that antibiotic treatment results in DNA double-strand breaks and membrane damage and demonstrate that antibiotic lethality under anaerobic conditions can be decreased by RMB scavengers, which reduce RES accumulation and mitigate associated macromolecular damage. This work indicates that RMBs, generated in response to antibiotic-induced energetic demands, contribute in part to antibiotic lethality under anaerobic conditions.
Collapse
Affiliation(s)
- Felix Wong
- Institute for Medical Engineering & Science and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Jonathan M Stokes
- Institute for Medical Engineering & Science and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Sarah C Bening
- Institute for Medical Engineering & Science and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Charles Vidoudez
- Harvard Center for Mass Spectrometry, Harvard University, Cambridge, MA 02138, USA
| | - Sunia A Trauger
- Harvard Center for Mass Spectrometry, Harvard University, Cambridge, MA 02138, USA
| | - James J Collins
- Institute for Medical Engineering & Science and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA.
| |
Collapse
|
30
|
Strain-specific predation of Bdellovibrio bacteriovorus on Pseudomonas aeruginosa with a higher range for cystic fibrosis than for bacteremia isolates. Sci Rep 2022; 12:10523. [PMID: 35732651 PMCID: PMC9217795 DOI: 10.1038/s41598-022-14378-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 06/06/2022] [Indexed: 12/23/2022] Open
Abstract
This work aimed to evaluate the predatory activity of Bdellovibrio bacteriovorus 109J on clinical isolates of Pseudomonas aeruginosa selected from well-characterized collections of cystic fibrosis (CF) lung colonization (n = 30) and bloodstream infections (BSI) (n = 48) including strains selected by genetic lineage (frequent and rare sequence types), antibiotic resistance phenotype (susceptible and multidrug-resistant isolates), and colony phenotype (mucoid and non-mucoid isolates). The intraspecies predation range (I-PR) was defined as the proportion of susceptible strains within the entire collection. In contrast, the predation efficiency (PE) is the ratio of viable prey cells remaining after predation compared to the initial inoculum. I-PR was significantly higher for CF (67%) than for BSI P. aeruginosa isolates (35%) probably related to an environmental origin of CF strains whereas invasive strains are more adapted to humans. I-PR correlation with bacterial features such as mucoid morphotype, genetic background, or antibiotic susceptibility profile was not detected. To test the possibility of increasing I-PR of BSI isolates, a polyhydroxyalkanoate depolymerase deficient B. bacteriovorus bd2637 mutant was used. Global median I-PR and PE values remained constant for both predators, but 31.2% of 109J-resistant isolates were susceptible to the mutant, and 22.9% of 109J-susceptible isolates showed resistance to predation by the mutant, pointing to a predator–prey specificity process. The potential use of predators in the clinical setting should be based on the determination of the I-PR for each species, and the PE of each particular target strain.
Collapse
|
31
|
Kim SM, Zou G, Kim H, Kang M, Ahn S, Heo HY, Kim JS, Lim KM, Ausubel FM, Mylonakis E, Gao H, Kim W. Antimicrobial activity of the membrane-active compound nTZDpa is enhanced at low pH. Biomed Pharmacother 2022; 150:112977. [PMID: 35447554 DOI: 10.1016/j.biopha.2022.112977] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/03/2022] [Accepted: 04/12/2022] [Indexed: 11/30/2022] Open
Abstract
The opportunistic human pathogen Staphylococcus aureus can evade antibiotics by acquiring antibiotic resistance genes or by entering into a non-growing dormant state. Moreover, the particular circumstances of a specific infection site, such as acidity or anaerobicity, often weaken antibiotic potency. Decreased bacterial susceptibility combined with diminished antibiotic potency is responsible for high failure rates when treating S. aureus infections. Here, we report that the membrane-active antimicrobial agent nTZDpa does not only exhibit enhanced antibiotic activity against multidrug-resistant Gram-positive pathogens in acidic pH, but also retains antimicrobial potency under anaerobic conditions. This agent completely eradicated highly antibiotic-tolerant cells and biofilms formed by methicillin-resistant S. aureus at pH 5.5 at concentrations at which it was not potent at pH 7.4. Furthermore, nTZDpa was more potent at synergistically potentiating gentamicin killing against antibiotic-tolerant MRSA cells at low pH than at high pH. All-atom molecular dynamics simulations combined with membrane-permeabilization assays revealed that the neutral form of nTZDpa, which contains carboxylic acid, is more effective than the deprotonated form at penetrating the bacterial membrane and plays an essential role in membrane activity. An acidic pH increases the proportion of the neutrally charged nTZDpa, which results in antimicrobial enhancement. Our results provide key insights into rational design of pH-sensitive membrane-active antimicrobials and antibiotic adjuvants that are effective in an infection environment. These findings demonstrate that nTZDpa is a promising lead compound for developing new therapeutics against hard-to-cure infections caused by drug-resistant and -tolerant S. aureus.
Collapse
Affiliation(s)
- Soo Min Kim
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Guijin Zou
- Institute of High Performance Computing, A⁎STAR, Singapore 138632, Singapore
| | - Hyerim Kim
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Minjeong Kang
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Soyeon Ahn
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Hee Young Heo
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Jae-Seok Kim
- Department of Laboratory Medicine, Kangdong Sacred Heart Hospital, Hallym University College of Medicine, Seoul 05355, Republic of Korea
| | - Kyung-Min Lim
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Frederick M Ausubel
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Eleftherios Mylonakis
- Division of Infectious Diseases, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Huajian Gao
- Institute of High Performance Computing, A⁎STAR, Singapore 138632, Singapore; School of Mechanical and Aerospace Engineering, College of Engineering, Nanyang Technological University, Singapore 639789, Singapore
| | - Wooseong Kim
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea.
| |
Collapse
|
32
|
Naclerio GA, Onyedibe KI, Karanja CW, Aryal UK, Sintim HO. Comparative Studies to Uncover Mechanisms of Action of N-(1,3,4-Oxadiazol-2-yl)benzamide Containing Antibacterial Agents. ACS Infect Dis 2022; 8:865-877. [PMID: 35297603 PMCID: PMC9188027 DOI: 10.1021/acsinfecdis.1c00613] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Drug-resistant bacterial pathogens still cause high levels of mortality annually despite the availability of many antibiotics. Methicillin-resistant Staphylococcus aureus (MRSA) is especially problematic, and the rise in resistance to front-line treatments like vancomycin and linezolid calls for new chemical modalities to treat chronic and relapsing MRSA infections. Halogenated N-(1,3,4-oxadiazol-2-yl)benzamides are an interesting class of antimicrobial agents, which have been described by multiple groups to be effective against different bacterial pathogens. The modes of action of a few N-(1,3,4-oxadiazol-2-yl)benzamides have been elucidated. For example, oxadiazoles KKL-35 and MBX-4132 have been described as inhibitors of trans-translation (a ribosome rescue pathway), while HSGN-94 was shown to inhibit lipoteichoic acid (LTA). However, other similarly halogenated N-(1,3,4-oxadiazol-2-yl)benzamides neither inhibit trans-translation nor LTA biosynthesis but are potent antimicrobial agents. For example, HSGN-220, -218, and -144 are N-(1,3,4-oxadiazol-2-yl)benzamides that are modified with OCF3, SCF3, or SF5 and have remarkable minimum inhibitory concentrations ranging from 1 to 0.06 μg/mL against MRSA clinical isolates and show a low propensity to develop resistance to MRSA over 30 days. The mechanism of action of these highly potent oxadiazoles is however unknown. To provide insights into how these halogenated N-(1,3,4-oxadiazol-2-yl)benzamides inhibit bacterial growth, we performed global proteomics and RNA expression analysis of some essential genes of S. aureus treated with HSGN-220, -218, and -144. These studies revealed that the oxadiazoles HSGN-220, -218, and -144 are multitargeting antibiotics that regulate menaquinone biosynthesis and other essential proteins like DnaX, Pol IIIC, BirA, LexA, and DnaC. In addition, these halogenated N-(1,3,4-oxadiazol-2-yl)benzamides were able to depolarize bacterial membranes and regulate siderophore biosynthesis and heme regulation. Iron starvation appears to be part of the mechanism of action that led to bacterial killing. This study demonstrates that N-(1,3,4-oxadiazol-2-yl)benzamides are indeed privileged scaffolds for the development of antibacterial agents and that subtle modifications lead to changes to the mechanism of action.
Collapse
Affiliation(s)
- George A. Naclerio
- Chemistry Department, Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Kenneth I. Onyedibe
- Chemistry Department, Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
- Purdue Institute for Inflammation, Immunology, and Infectious Diseases, West Lafayette, Indiana 47907, United States
| | - Caroline W. Karanja
- Chemistry Department, Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Uma K. Aryal
- Department of Comparative Pathobiology, Purdue University, West Lafayette, Indiana 47907, United States
- Purdue Proteomics Facility, Bindley Bioscience Center, Purdue University, West Lafayette, Indiana 47907, United States
| | - Herman O. Sintim
- Chemistry Department, Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
- Purdue Institute for Inflammation, Immunology, and Infectious Diseases, West Lafayette, Indiana 47907, United States
| |
Collapse
|