1
|
Ha KP, Kumeko EK, Bouloc P. A small regulatory RNA controls antibiotic adaptation in Staphylococcus aureus by modulating efflux pump expression. Antimicrob Agents Chemother 2025; 69:e0117624. [PMID: 40178302 PMCID: PMC12057360 DOI: 10.1128/aac.01176-24] [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: 08/05/2024] [Accepted: 03/06/2025] [Indexed: 04/05/2025] Open
Abstract
Staphylococcus aureus is an opportunistic pathogen that poses a considerable burden to healthcare settings worldwide, aided by its ability to thrive in different environmental growth conditions and survive exposure to antibiotics. Small regulatory RNAs (sRNAs) are decisive in enhancing bacterial fitness by modulating gene expression in response to changing environmental conditions. We investigated the role of sRNAs in the adaptation of S. aureus to antibiotics. By assessing the fitness of a library of sRNA mutants, we identified that RsaA sRNA is required for optimal bacterial growth when exposed to low concentrations of fluoroquinolone, a class of antibiotics targeting DNA replication. We also found that in the absence of RsaA, S. aureus is less susceptible to β-lactam antibiotics, which act on the cell wall. RsaA has been reported to prevent the expression of MgrA, a master regulatory protein controlling the expression of efflux pumps. Here, we show that RsaA affects the sensitivity of S. aureus to fluoroquinolone and β-lactam antibiotics through MgrA. RsaA has two forms, a short one commonly referred to in RsaA studies, and a long form about twice the length, of which less is known. Interestingly, our phenotype was only restored when complemented with the long form of the gene or when it was supplied in two parts, the short form and the missing part to obtain the long form. This work demonstrates the role of regulatory RNAs in the adaptation of S. aureus to antibiotic resistance and highlights their value as potential therapeutic targets for manipulating individual sRNA responses to promote the efficacy of existing antibiotics.
Collapse
Affiliation(s)
- Kam Pou Ha
- Université Paris-Saclay, CEA, CNRS, Institut de Biologie Intégrative de la Cellule (I2BC), Gif-sur-Yvette, Île-de-France, France
| | - Etornam Kofi Kumeko
- Université Paris-Saclay, CEA, CNRS, Institut de Biologie Intégrative de la Cellule (I2BC), Gif-sur-Yvette, Île-de-France, France
| | - Philippe Bouloc
- Université Paris-Saclay, CEA, CNRS, Institut de Biologie Intégrative de la Cellule (I2BC), Gif-sur-Yvette, Île-de-France, France
| |
Collapse
|
2
|
Ojha S, Sinsinwar S, Chatterjee P, Biswal S, Pradhan P, Beuria TK. Efflux pump modulation by Montelukast and its roles in restoring antibiotic susceptibility in multidrug-resistant Staphylococcus aureus. EBioMedicine 2025; 114:105658. [PMID: 40157128 PMCID: PMC11994358 DOI: 10.1016/j.ebiom.2025.105658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 02/22/2025] [Accepted: 03/05/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND Staphylococcus aureus and its drug-resistant mutants are mentioned among the WHO's high-priority list of pathogens. Antibiotics like fluoroquinolones and cephalosporins are used to treat multidrug-resistant S. aureus infections. However, a higher expression of efflux pumps (NorA, NorB, and AbcA) induces multidrug resistance. The master regulator, MgrA, regulates the expression of most of these efflux pumps in S. aureus. The phosphorylation status of MgrA is determined by the cellular PknB/RsbU ratio, where PknB, a serine-threonine kinase, and RsbU, a serine-threonine phosphatase, are critical for MgrA functioning. METHODS An FDA-approved drug library was screened using an EtBr-accumulation assay to identify efflux pump inhibitors (EPIs). The synergy of EPIs with antibiotics was studied in vitro and in vivo in the murine skin infection model of female BALB/c mice. The effect of EPIs on mgrA, norB, pknB, and rsbU gene expression, interaction with MgrA, and effects on MgrA phosphorylation were studied. FINDINGS We identified Montelukast as an effective EPI, which showed synergy with moxifloxacin, a substrate of the NorB efflux pump, both in vitro and in the murine skin infection model. Further, Montelukast decreased norB expression and increased the pknB/rsbU expression ratio. Our in vitro results demonstrated that Montelukast strongly interacted with MgrA, facilitated MgrA phosphorylation, and enhanced its affinity for the norB promoter. INTERPRETATION Our study showed that Montelukast repressed MgrA expression and promoted MgrA phosphorylation to suppress norB expression and efflux pump activity, leading to the restoration of antibiotic susceptibility in multidrug-resistant S. aureus. FUNDING The study was supported by SERB-DST, India (CRG/2021/005069), and the BRIC-ILS core.
Collapse
Affiliation(s)
- Suvendu Ojha
- Department of Infectious Disease Biology, Institute of Life Sciences, NALCO Square, Chandrasekharpur, Bhubaneswar, Odisha, 751023, India; Regional Centre for Biotechnology, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad Rd, Faridabad, Haryana, 121001, India
| | - Simran Sinsinwar
- Department of Infectious Disease Biology, Institute of Life Sciences, NALCO Square, Chandrasekharpur, Bhubaneswar, Odisha, 751023, India
| | - Puja Chatterjee
- Department of Infectious Disease Biology, Institute of Life Sciences, NALCO Square, Chandrasekharpur, Bhubaneswar, Odisha, 751023, India; Regional Centre for Biotechnology, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad Rd, Faridabad, Haryana, 121001, India
| | - Sarmistha Biswal
- Department of Infectious Disease Biology, Institute of Life Sciences, NALCO Square, Chandrasekharpur, Bhubaneswar, Odisha, 751023, India
| | - Pinkilata Pradhan
- Department of Infectious Disease Biology, Institute of Life Sciences, NALCO Square, Chandrasekharpur, Bhubaneswar, Odisha, 751023, India; Regional Centre for Biotechnology, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad Rd, Faridabad, Haryana, 121001, India
| | - Tushar Kant Beuria
- Department of Infectious Disease Biology, Institute of Life Sciences, NALCO Square, Chandrasekharpur, Bhubaneswar, Odisha, 751023, India.
| |
Collapse
|
3
|
Sinha S, Aggarwal S, Singh DV. Efflux pumps: gatekeepers of antibiotic resistance in Staphylococcus aureus biofilms. MICROBIAL CELL (GRAZ, AUSTRIA) 2024; 11:368-377. [PMID: 39568862 PMCID: PMC11576857 DOI: 10.15698/mic2024.11.839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 10/01/2024] [Accepted: 10/08/2024] [Indexed: 11/22/2024]
Abstract
Staphylococcus aureus, a versatile human pathogen, poses a significant challenge in healthcare settings due to its ability to develop antibiotic resistance and form robust biofilms. Understanding the intricate mechanisms underlying the antibiotic resistance is crucial for effective infection treatment and control. This comprehensive review delves into the multifaceted roles of efflux pumps in S. aureus, with a focus on their contribution to antibiotic resistance and biofilm formation. Efflux pumps, integral components of the bacterial cell membrane, are responsible for expelling a wide range of toxic substances, including antibiotics, from bacterial cells. By actively extruding antibiotics, these pumps reduce intracellular drug concentrations, rendering antibiotics less effective. Moreover, efflux pumps have emerged as significant contributors to both antibiotic resistance and biofilm formation in S. aureus. Biofilms, structured communities of bacterial cells embedded in a protective matrix, enable S. aureus to adhere to surfaces, evade host immune responses, and resist antibiotic therapy. Efflux pumps play a pivotal role in the development and maintenance of S. aureus biofilms. However, the interplay between efflux pumps, antibiotic resistance and biofilm formation remains unexplored in S. aureus. This review aims to elucidate the complex relationship between efflux pumps, antibiotic resistance and biofilm formation in S. aureus with the aim to aid in the development of potential therapeutic targets for combating S. aureus infections, especially those associated with biofilms. The insights provided herein may contribute to the advancement of novel strategies to overcome antibiotic resistance and disrupt biofilm formation in this clinically significant pathogen.
Collapse
Affiliation(s)
- Shweta Sinha
- Department of Biotechnology, School of Earth, Biological and Environmental Sciences, Central University of South Bihar Gaya, 824236 India
| | - Shifu Aggarwal
- Department of Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, 751023 India
- Current Address: Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts USA
| | - Durg Vijai Singh
- Department of Biotechnology, School of Earth, Biological and Environmental Sciences, Central University of South Bihar Gaya, 824236 India
| |
Collapse
|
4
|
de Araújo ACJ, Freitas PR, Araújo IM, de Oliveira Borges JA, Gonçalves SA, Paulo CLR, Almeida RS, de Moraes Oliveira-Tintino CD, de Araújo-Neto JB, Dos Santos Nascimento IJ, de Araújo-Júnior JX, da Silva-Júnior EF, de Aquino TM, Junior FJBM, Marinho ES, Dos Santos HS, de Alencar Menezes IR, Tintino SR, Coutinho HDM, Braga MFBM. Assessment In vitro and In silico of the Activity of Thiadiazines as NorA Efflux Pump Inhibitors. Curr Microbiol 2024; 81:325. [PMID: 39182011 DOI: 10.1007/s00284-024-03836-0] [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: 05/16/2024] [Accepted: 08/08/2024] [Indexed: 08/27/2024]
Abstract
Antimicrobials fight microorganisms, preventing and treating infectious diseases. However, antimicrobial resistance (AMR) is a growing concern due to the inappropriate and excessive use of these drugs. Several mechanisms can lead to resistance, including efflux pumps such as the NorA pump in Staphylococcus aureus, which reduces the effectiveness of fluoroquinolones. Thiadiazines are heterocyclic compounds whose chemical structure resembles that of cephalosporins. Therefore, these compounds and their derivatives have been studied for their potential in combating increased bacterial resistance. To analyze this hypothesis, direct activity assays, antibiotic action-modifying activity, fluorescence assays to evaluate the retention of ethidium bromide inside bacteria, and molecular docking were carried out. These experiments involved serial dilutions in microplates against Staphylococcus aureus strain 1199B under the influence of six thiadiazine derivatives (IJ10, IJ11, IJ21, IJ22, IJ23, and IJ25). The tests revealed that, despite not showing effective direct activity, some thiadiazine derivatives (IJ11, IJ21, and IJ22) inhibited the function of the bromide pump both in microdilution tests and in fluorescence and docking assays. Particularly, the IJ11 compound stood out for its activity similar to efflux inhibitors, as well as its inhibition of the norfloxacin pump of this bacterium. Among the results of this study, it deserves to be highlighted for anchoring future experiments, as it represents the first investigation of this group of thiadiazine derivatives against the NorA pump.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - José Bezerra de Araújo-Neto
- Postgraduate Program in Biological Sciences, Biosciences Center, Federal University of Pernambuco, Recife, PE, 50740-570, Brazil
| | - Igor José Dos Santos Nascimento
- Laboratory of Medicinal Chemistry, Institute of Pharmaceutical Sciences, Federal University of Alagoas, Maceió, AL, 57072-900, Brazil
| | - João Xavier de Araújo-Júnior
- Laboratory of Medicinal Chemistry, Institute of Pharmaceutical Sciences, Federal University of Alagoas, Maceió, AL, 57072-900, Brazil
| | - Edeildo Ferreira da Silva-Júnior
- Biological and Molecular Chemistry Research Group, Institute of Chemistry and Biotechnology, Federal University of Alagoas, Maceió, AL, 57072-900, Brazil
| | - Thiago Mendonça de Aquino
- Laboratory of Synthesis and Research in Medicinal Chemistry, Research Group On Therapeutic Strategies-GPET, Institute of Chemistry and Biotechnology, Federal University of Alagoas, Maceió, AL, 57072-900, Brazil
| | | | - Emmanuel Silva Marinho
- Laboratory of Chemistry of Natural and Synthetic Product, State University of Ceará, UECE, Fortaleza, CE, Brazil
| | - Helcio Silva Dos Santos
- Laboratory of Chemistry of Natural and Synthetic Product, State University of Ceará, UECE, Fortaleza, CE, Brazil
| | | | | | | | | |
Collapse
|
5
|
Truong-Bolduc QC, Wang Y, Ferrer-Espada R, Reedy JL, Martens AT, Goulev Y, Paulsson J, Vyas JM, Hooper DC. Staphylococcus aureus AbcA transporter enhances persister formation under β-lactam exposure. Antimicrob Agents Chemother 2024; 68:e0134023. [PMID: 38364015 PMCID: PMC10916373 DOI: 10.1128/aac.01340-23] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 01/18/2024] [Indexed: 02/18/2024] Open
Abstract
We evaluated the role of Staphylococcus aureus AbcA transporter in bacterial persistence and survival following exposure to the bactericidal agents nafcillin and oxacillin at both the population and single-cell levels. We show that AbcA overexpression resulted in resistance to nafcillin but not oxacillin. Using distinct fluorescent reporters of cell viability and AbcA expression, we found that over 6-14 hours of persistence formation, the proportion of AbcA reporter-expressing cells assessed by confocal microscopy increased sixfold as cell viability reporters decreased. Similarly, single-cell analysis in a high-throughput microfluidic system found a strong correspondence between antibiotic exposure and AbcA reporter expression. Persister cells grown in the absence of antibiotics showed neither an increase in nafcillin MIC nor in abcA transcript levels, indicating that survival was not associated with stable mutational resistance or abcA overexpression. Furthermore, persister cell levels on exposure to 1×MIC and 25×MIC of nafcillin decreased in an abcA knockout mutant. Survivors of nafcillin and oxacillin treatment overexpressed transporter AbcA, contributing to an enrichment of the number of persisters during treatment with pump-substrate nafcillin but not with pump-non-substrate oxacillin, indicating that efflux pump expression can contribute selectively to the survival of a persister population.
Collapse
Affiliation(s)
- Q. C. Truong-Bolduc
- Infectious Diseases Division and Medical Services, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Y. Wang
- Infectious Diseases Division and Medical Services, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - R. Ferrer-Espada
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts, USA
| | - J. L. Reedy
- Infectious Diseases Division and Medical Services, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - A. T. Martens
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts, USA
| | - Y. Goulev
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts, USA
| | - J. Paulsson
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts, USA
| | - J. M. Vyas
- Infectious Diseases Division and Medical Services, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - D. C. Hooper
- Infectious Diseases Division and Medical Services, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
6
|
Ayoola MB, Das AR, Krishnan BS, Smith DR, Nanduri B, Ramkumar M. Predicting Salmonella MIC and Deciphering Genomic Determinants of Antibiotic Resistance and Susceptibility. Microorganisms 2024; 12:134. [PMID: 38257961 PMCID: PMC10819212 DOI: 10.3390/microorganisms12010134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 01/04/2024] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
Salmonella spp., a leading cause of foodborne illness, is a formidable global menace due to escalating antimicrobial resistance (AMR). The evaluation of minimum inhibitory concentration (MIC) for antimicrobials is critical for characterizing AMR. The current whole genome sequencing (WGS)-based approaches for predicting MIC are hindered by both computational and feature identification constraints. We propose an innovative methodology called the "Genome Feature Extractor Pipeline" that integrates traditional machine learning (random forest, RF) with deep learning models (multilayer perceptron (MLP) and DeepLift) for WGS-based MIC prediction. We used a dataset from the National Antimicrobial Resistance Monitoring System (NARMS), comprising 4500 assembled genomes of nontyphoidal Salmonella, each annotated with MIC metadata for 15 antibiotics. Our pipeline involves the batch downloading of annotated genomes, the determination of feature importance using RF, Gini-index-based selection of crucial 10-mers, and their expansion to 20-mers. This is followed by an MLP network, with four hidden layers of 1024 neurons each, to predict MIC values. Using DeepLift, key 20-mers and associated genes influencing MIC are identified. The 10 most significant 20-mers for each antibiotic are listed, showcasing our ability to discern genomic features affecting Salmonella MIC prediction with enhanced precision. The methodology replaces binary indicators with k-mer counts, offering a more nuanced analysis. The combination of RF and MLP addresses the limitations of the existing WGS approach, providing a robust and efficient method for predicting MIC values in Salmonella that could potentially be applied to other pathogens.
Collapse
Affiliation(s)
- Moses B. Ayoola
- Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Starkville, MS 39762, USA; (M.B.A.); (A.R.D.); (B.S.K.); (B.N.)
| | - Athish Ram Das
- Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Starkville, MS 39762, USA; (M.B.A.); (A.R.D.); (B.S.K.); (B.N.)
| | - B. Santhana Krishnan
- Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Starkville, MS 39762, USA; (M.B.A.); (A.R.D.); (B.S.K.); (B.N.)
| | - David R. Smith
- Department of Population Medicine, College of Veterinary Medicine, Mississippi State University, Starkville, MS 39762, USA;
| | - Bindu Nanduri
- Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Starkville, MS 39762, USA; (M.B.A.); (A.R.D.); (B.S.K.); (B.N.)
| | - Mahalingam Ramkumar
- Department of Computer Science and Engineering, Mississippi State University, Starkville, MS 39762, USA
| |
Collapse
|
7
|
Cai L, Zhang L, Yang J, Zhu X, Wei W, Ji M, Jiang H, Chen J. Encapsulating Antibiotic and Protein-Stabilized Nanosilver into Sandwich-Structured Electrospun Nanofibrous Scaffolds for MRSA-Infected Wound Treatment. ACS APPLIED MATERIALS & INTERFACES 2023; 15:48978-48995. [PMID: 37877381 DOI: 10.1021/acsami.3c10994] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2023]
Abstract
With the increasing prevalence of microbial infections, which results in prolonged inflammation and delayed wound healing, the development of effective and safe antimicrobial wound dressings of multiple properties remains challenging for public health. Despite their various formats, the available developed dressings with limited functions may not fulfill the diverse demands involved in the complex wound healing process. In this study, multifunctional sandwich-structured electrospinning nanofiber membranes (ENMs) were fabricated. According to the structural composition, the obtained ENMs included a hydrophilic inner layer loaded with curcumin and gentamicin sulfate, an antibacterial middle layer consisting of bovine serum albumin stabilized silver oxide nanoparticles, and a hydrophobic outer layer. The prepared sandwich-structured ENMs (SNM) exhibited good biocompatibility and killing efficacy on Escherichia coli, Staphylococcus aureus, and Methicillin-resistant S. aureus (MRSA). In particular, transcriptomic analysis revealed that SNM inactivated MRSA by inhibiting its carbohydrate and energy metabolism and reduced the bacterial resistance by downregulating mecA. In the animal experiment, SNM showed improved wound healing efficiency by reducing the bacterial load and inflammation. Moreover, 16S rDNA sequencing results indicated that SNM treatment may accelerate wound healing without observed influence on the normal skin flora. Therefore, the constructed sandwich-structured ENMs exhibited promising potential as dressings to deal with the infected wound management.
Collapse
Affiliation(s)
- Ling Cai
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Li Zhang
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Jing Yang
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Xinyi Zhu
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Wei Wei
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Minghui Ji
- School of Nursing, Nanjing Medical University, Nanjing 211166, China
| | - Huijun Jiang
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Jin Chen
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
- Jiangsu Province Engineering Research Center of Antibody Drug, Key Laboratory of Antibody Technique of National Health Commission, Nanjing Medical University, Nanjing 211166, China
| |
Collapse
|
8
|
Hsieh RC, Liu R, Burgin DJ, Otto M. Understanding mechanisms of virulence in MRSA: implications for antivirulence treatment strategies. Expert Rev Anti Infect Ther 2023; 21:911-928. [PMID: 37501364 DOI: 10.1080/14787210.2023.2242585] [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: 05/15/2023] [Accepted: 07/26/2023] [Indexed: 07/29/2023]
Abstract
INTRODUCTION Methicillin-resistant Staphylococcus aureus (MRSA) is a widespread pathogen, often causing recurrent and deadly infections in the hospital and community. Many S. aureus virulence factors have been suggested as potential targets for antivirulence therapy to decrease the threat of diminishing antibiotic availability. Antivirulence methods hold promise due to their adjunctive and prophylactic potential and decreased risk for selective pressure. AREAS COVERED This review describes the dominant virulence mechanisms exerted by MRSA and antivirulence therapeutics that are currently undergoing testing in clinical or preclinical stages. We also discuss the advantages and downsides of several investigational antivirulence approaches, including the targeting of bacterial transporters, host-directed therapy, and quorum-sensing inhibitors. For this review, a systematic search of literature on PubMed, Google Scholar, and Web of Science for relevant search terms was performed in April and May 2023. EXPERT OPINION Vaccine and antibody strategies have failed in clinical trials and could benefit from more basic science-informed approaches. Antivirulence-targeting approaches need to be set up better to meet the requirements of drug development, rather than only providing limited results to provide 'proof-of-principle' translational value of pathogenesis research. Nevertheless, there is great potential of such strategies and potential particular promise for novel probiotic approaches.
Collapse
Affiliation(s)
- Roger C Hsieh
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases (NIAID), U.S. National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Ryan Liu
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases (NIAID), U.S. National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Dylan J Burgin
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases (NIAID), U.S. National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Michael Otto
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases (NIAID), U.S. National Institutes of Health (NIH), Bethesda, Maryland, USA
| |
Collapse
|
9
|
Shu X, Shi Y, Huang Y, Yu D, Sun B. Transcription tuned by S-nitrosylation underlies a mechanism for Staphylococcus aureus to circumvent vancomycin killing. Nat Commun 2023; 14:2318. [PMID: 37085493 PMCID: PMC10120478 DOI: 10.1038/s41467-023-37949-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 04/06/2023] [Indexed: 04/23/2023] Open
Abstract
Treatment of Staphylococcus aureus infections is a constant challenge due to emerging resistance to vancomycin, a last-resort drug. S-nitrosylation, the covalent attachment of a nitric oxide (NO) group to a cysteine thiol, mediates redox-based signaling for eukaryotic cellular functions. However, its role in bacteria is largely unknown. Here, proteomic analysis revealed that S-nitrosylation is a prominent growth feature of vancomycin-intermediate S. aureus. Deletion of NO synthase (NOS) or removal of S-nitrosylation from the redox-sensitive regulator MgrA or WalR resulted in thinner cell walls and increased vancomycin susceptibility, which was due to attenuated promoter binding and released repression of genes involved in cell wall metabolism. These genes failed to respond to H2O2-induced oxidation, suggesting distinct transcriptional responses to alternative modifications of the cysteine residue. Furthermore, treatment with a NOS inhibitor significantly decreased vancomycin resistance in S. aureus. This study reveals that transcriptional regulation via S-nitrosylation underlies a mechanism for NO-mediated bacterial antibiotic resistance.
Collapse
Affiliation(s)
- Xueqin Shu
- Department of Oncology, The First Affiliated Hospital, University of Science and Technology of China, Hefei, China
- Department of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Yingying Shi
- Department of Oncology, The First Affiliated Hospital, University of Science and Technology of China, Hefei, China
- Department of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Yi Huang
- Department of Oncology, The First Affiliated Hospital, University of Science and Technology of China, Hefei, China
- Department of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Dan Yu
- Laboratory of Dermatology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Key Laboratory of Major Diseases in Children, Ministry of Education, National Center for Children's Health, Beijing, China.
| | - Baolin Sun
- Department of Oncology, The First Affiliated Hospital, University of Science and Technology of China, Hefei, China.
- Department of Life Science and Medicine, University of Science and Technology of China, Hefei, China.
- CAS Laboratory of Innate Immunity and Chronic Disease, University of Science and Technology of China, Hefei, China.
- Hefei National Laboratory for Physical Sciences at Microscale, Hefei, China.
| |
Collapse
|
10
|
Dickey SW, Burgin DJ, Huang S, Maguire D, Otto M. Two transporters cooperate to secrete amphipathic peptides from the cytoplasmic and membranous milieus. Proc Natl Acad Sci U S A 2023; 120:e2211689120. [PMID: 36787359 PMCID: PMC9974410 DOI: 10.1073/pnas.2211689120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 01/18/2023] [Indexed: 02/15/2023] Open
Abstract
Diverse organisms secrete amphipathic biomolecules for competitive gains. However, how cells cope with producing these membrane-permeabilizing molecules is unclear. We focused on the PSM family of secreted amphipathic peptides in the pathogen Staphylococcus aureus that uses two ABC transporters, PmtCD and AbcA, to export peptides across the bacterial cell membrane. We found that increased peptide hydrophobicity favors PSM secretion through PmtCD over AbcA and that only PmtCD protected cells against amphipathic peptides. We propose a two-system model in which PmtCD and AbcA independently export PSMs from either membrane or cytosolic environments, respectively. Our model provides a rationale for the encoding of multiple transport systems on diverse biosynthetic gene clusters used to produce distinct amphipathic molecules. In addition, our data serve as a guide for selectively blocking PSM secretion to achieve antimicrobial or antivirulence approaches and to disrupt established roles of PSM-mediated virulence.
Collapse
Affiliation(s)
- Seth W. Dickey
- National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892
- National Institute of General Medical Sciences, NIH, Bethesda, MD20892
- Department of Veterinary Medicine, University of Maryland, College Park, MD20740
- Virginia-Maryland College of Veterinary Medicine, College Park, MD20740
| | - Dylan J. Burgin
- National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892
| | - Steven Huang
- National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892
| | - David Maguire
- National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892
| | - Michael Otto
- National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892
| |
Collapse
|
11
|
Satishkumar N, Lai LY, Mukkayyan N, Vogel BE, Chatterjee SS. A Nonclassical Mechanism of β-Lactam Resistance in Methicillin-Resistant Staphylococcus aureus and Its Effect on Virulence. Microbiol Spectr 2022; 10:e0228422. [PMID: 36314912 PMCID: PMC9769611 DOI: 10.1128/spectrum.02284-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 10/07/2022] [Indexed: 11/07/2022] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is a group of pathogenic bacteria that are infamously resistant to β-lactam antibiotics, a property attributed to the mecA gene. Recent studies have reported that mutations associated with the promoter region of pbp4 demonstrated high levels of β-lactam resistance, suggesting the role of PBP4 as an important non-mecA mediator of β-lactam resistance. The pbp4-promoter-associated mutations have been detected in strains with or without mecA. Our previous studies that were carried out in strains devoid of mecA described that pbp4-promoter-associated mutations lead to PBP4 overexpression and β-lactam resistance. In this study, by introducing various pbp4-promoter-associated mutations in the genome of a MRSA strain, we demonstrate that PBP4 overexpression can supplement mecA-associated resistance in S. aureus and can lead to increased β-lactam resistance. The promoter and regulatory region of pbp4 is shared with a divergently transcribed gene, abcA, which encodes a multidrug exporter. We demonstrate that the promoter mutations caused an upregulation of pbp4 and downregulation of abcA, confirming that the resistant phenotype is associated with PBP4 overexpression. PBP4 has also been associated with staphylococcal pathogenesis, however, its exact role remains unclear. Using a Caenorhabditis elegans model, we demonstrate that strains having increased PBP4 expression are less virulent than wild-type strains, suggesting that β-lactam resistance mediated via PBP4 likely comes at the cost of virulence. IMPORTANCE Our study demonstrates the ability of PBP4 to be an important mediator of β-lactam resistance in not only methicillin-susceptible Staphylococcus aureus (MSSA) background strains as previously demonstrated but also in MRSA strains. When present together, PBP2a and PBP4 overexpression can produce increased levels of β-lactam resistance, causing complications in treatment. Thus, this study suggests the importance of monitoring PBP4-associated resistance in clinical settings, as well as understanding the mechanistic basis of associated resistance, so that treatments targeting PBP4 may be developed. This study also demonstrates that S. aureus strains with increased PBP4 expression are less pathogenic, providing important hints about the role of PBP4 in S. aureus resistance and pathogenesis.
Collapse
Affiliation(s)
- Nidhi Satishkumar
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, Baltimore, Maryland, USA
- Institute of Marine and Environmental Technology (IMET), Baltimore, Maryland, USA
| | - Li-Yin Lai
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, Baltimore, Maryland, USA
- Institute of Marine and Environmental Technology (IMET), Baltimore, Maryland, USA
| | - Nagaraja Mukkayyan
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, Baltimore, Maryland, USA
- Institute of Marine and Environmental Technology (IMET), Baltimore, Maryland, USA
| | - Bruce E. Vogel
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, University of Maryland, Baltimore, Maryland, USA
| | - Som S. Chatterjee
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, Baltimore, Maryland, USA
- Institute of Marine and Environmental Technology (IMET), Baltimore, Maryland, USA
| |
Collapse
|
12
|
Increased Expression of Efflux Pump norA Drives the Rapid Evolutionary Trajectory from Tolerance to Resistance against Ciprofloxacin in Staphylococcus aureus. Antimicrob Agents Chemother 2022; 66:e0059422. [PMID: 36445128 PMCID: PMC9765010 DOI: 10.1128/aac.00594-22] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
The intensively intermittent use of antibiotics promotes the rapid evolution of tolerance, which may lead to resistance acquisition in the following evolutionary trajectory. In addition to directly exporting antibiotics as an instant resistance strategy, efflux pumps are overexpressed in tolerant strains. To investigate how efflux pumps participate in resistance development from tolerance to resistance, we performed in vitro evolutional experiments against the antibiotic ciprofloxacin in norA efflux pump mutants of Staphylococcus aureus. These experiments demonstrated that overexpression of norA rapidly facilitated the development of ciprofloxacin resistance from tolerance to resistance through elevated spontaneous mutations. The generated resistance mutations were further fixed in the population by increasing survival ability. The observed Ser80Phe and Glu84Lys mutations in the topoisomerase IV ParC (GrlA in S. aureus) may be responsible for tolerant strains to develop resistance to ciprofloxacin since it has been reported that such mutations disrupt the water-metal ion bridge between quinolones and ParC. MepA and Sav1866 are related to the same antibiotic (ciprofloxacin) susceptibility as NorA, and they also contributed to resistance development against ciprofloxacin. MgrA positively regulated NorA expression and the development of ciprofloxacin resistance. Importantly, blocking the evolutionary pathway by coadministering ciprofloxacin with the efflux pump inhibitor reserpine effectively delayed the resistance acquisition in an in vitro experiment. This study illustrated the role of efflux pumps in the evolutionary trajectory from tolerance to resistance. The delayed resistance development caused by the efflux pump inhibitor illuminates a possible strategy for postponing the resistance acquisition from tolerance to resistance by disrupting efflux pumps.
Collapse
|
13
|
Sionov RV, Banerjee S, Bogomolov S, Smoum R, Mechoulam R, Steinberg D. Targeting the Achilles' Heel of Multidrug-Resistant Staphylococcus aureus by the Endocannabinoid Anandamide. Int J Mol Sci 2022; 23:7798. [PMID: 35887146 PMCID: PMC9319909 DOI: 10.3390/ijms23147798] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/10/2022] [Accepted: 07/12/2022] [Indexed: 02/06/2023] Open
Abstract
Antibiotic-resistant Staphylococcus aureus is a major health issue that requires new therapeutic approaches. Accumulating data suggest that it is possible to sensitize these bacteria to antibiotics by combining them with inhibitors targeting efflux pumps, the low-affinity penicillin-binding protein PBP2a, cell wall teichoic acid, or the cell division protein FtsZ. We have previously shown that the endocannabinoid Anandamide (N-arachidonoylethanolamine; AEA) could sensitize drug-resistant S. aureus to a variety of antibiotics, among others, through growth arrest and inhibition of drug efflux. Here, we looked at biochemical alterations caused by AEA. We observed that AEA increased the intracellular drug concentration of a fluorescent penicillin and augmented its binding to membrane proteins with concomitant altered membrane distribution of these proteins. AEA also prevented the secretion of exopolysaccharides (EPS) and reduced the cell wall teichoic acid content, both processes known to require transporter proteins. Notably, AEA was found to inhibit membrane ATPase activity that is necessary for transmembrane transport. AEA did not affect the membrane GTPase activity, and the GTPase cell division protein FtsZ formed the Z-ring of the divisome normally in the presence of AEA. Rather, AEA caused a reduction in murein hydrolase activities involved in daughter cell separation. Altogether, this study shows that AEA affects several biochemical processes that culminate in the sensitization of the drug-resistant bacteria to antibiotics.
Collapse
Affiliation(s)
- Ronit Vogt Sionov
- Biofilm Research Laboratory, Institute of Biomedical and Oral Sciences, Faculty of Dentistry, Hadassah Medical School, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel; (S.B.); (S.B.); (D.S.)
| | - Shreya Banerjee
- Biofilm Research Laboratory, Institute of Biomedical and Oral Sciences, Faculty of Dentistry, Hadassah Medical School, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel; (S.B.); (S.B.); (D.S.)
| | - Sergei Bogomolov
- Biofilm Research Laboratory, Institute of Biomedical and Oral Sciences, Faculty of Dentistry, Hadassah Medical School, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel; (S.B.); (S.B.); (D.S.)
| | - Reem Smoum
- Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel; (R.S.); (R.M.)
| | - Raphael Mechoulam
- Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel; (R.S.); (R.M.)
| | - Doron Steinberg
- Biofilm Research Laboratory, Institute of Biomedical and Oral Sciences, Faculty of Dentistry, Hadassah Medical School, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel; (S.B.); (S.B.); (D.S.)
| |
Collapse
|
14
|
Forecasting Staphylococcus aureus Infections Using Genome-Wide Association Studies, Machine Learning, and Transcriptomic Approaches. mSystems 2022; 7:e0037822. [PMID: 35862809 PMCID: PMC9426533 DOI: 10.1128/msystems.00378-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Staphylococcus aureus is a major human and animal pathogen, colonizing diverse ecological niches within its hosts. Predicting whether an isolate will infect a specific host and its subsequent clinical fate remains unknown. In this study, we investigated the S. aureus pangenome using a curated set of 356 strains, spanning a wide range of hosts, origins, and clinical display and antibiotic resistance profiles. We used genome-wide association study (GWAS) and random forest (RF) algorithms to discriminate strains based on their origins and clinical sources. Here, we show that the presence of sak and scn can discriminate strains based on their host specificity, while other genes such as mecA are often associated with virulent outcomes. Both GWAS and RF indicated the importance of intergenic regions (IGRs) and coding DNA sequence (CDS) but not sRNAs in forecasting an outcome. Additional transcriptomic analyses performed on the most prevalent clonal complex 8 (CC8) clonal types, in media mimicking nasal colonization or bacteremia, indicated three RNAs as potential RNA markers to forecast infection, followed by 30 others that could serve as infection severity predictors. Our report shows that genetic association and transcriptomics are complementary approaches that will be combined in a single analytical framework to improve our understanding of bacterial pathogenesis and ultimately identify potential predictive molecular markers. IMPORTANCE Predicting the outcome of bacterial colonization and infections, based on extensive genomic and transcriptomic data from a given pathogen, would be of substantial help for clinicians in treating and curing patients. In this report, genome-wide association studies and random forest algorithms have defined gene combinations that differentiate human from animal strains, colonization from diseases, and nonsevere from severe diseases, while it revealed the importance of IGRs and CDS, but not small RNAs (sRNAs), in anticipating an outcome. In addition, transcriptomic analyses performed on the most prevalent clonal types, in media mimicking either nasal colonization or bacteremia, revealed significant differences and therefore potent RNA markers. Overall, the use of both genomic and transcriptomic data in a single analytical framework can enhance our understanding of bacterial pathogenesis.
Collapse
|
15
|
Sionov RV, Steinberg D. Targeting the Holy Triangle of Quorum Sensing, Biofilm Formation, and Antibiotic Resistance in Pathogenic Bacteria. Microorganisms 2022; 10:1239. [PMID: 35744757 PMCID: PMC9228545 DOI: 10.3390/microorganisms10061239] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 06/12/2022] [Accepted: 06/14/2022] [Indexed: 12/12/2022] Open
Abstract
Chronic and recurrent bacterial infections are frequently associated with the formation of biofilms on biotic or abiotic materials that are composed of mono- or multi-species cultures of bacteria/fungi embedded in an extracellular matrix produced by the microorganisms. Biofilm formation is, among others, regulated by quorum sensing (QS) which is an interbacterial communication system usually composed of two-component systems (TCSs) of secreted autoinducer compounds that activate signal transduction pathways through interaction with their respective receptors. Embedded in the biofilms, the bacteria are protected from environmental stress stimuli, and they often show reduced responses to antibiotics, making it difficult to eradicate the bacterial infection. Besides reduced penetration of antibiotics through the intricate structure of the biofilms, the sessile biofilm-embedded bacteria show reduced metabolic activity making them intrinsically less sensitive to antibiotics. Moreover, they frequently express elevated levels of efflux pumps that extrude antibiotics, thereby reducing their intracellular levels. Some efflux pumps are involved in the secretion of QS compounds and biofilm-related materials, besides being important for removing toxic substances from the bacteria. Some efflux pump inhibitors (EPIs) have been shown to both prevent biofilm formation and sensitize the bacteria to antibiotics, suggesting a relationship between these processes. Additionally, QS inhibitors or quenchers may affect antibiotic susceptibility. Thus, targeting elements that regulate QS and biofilm formation might be a promising approach to combat antibiotic-resistant biofilm-related bacterial infections.
Collapse
Affiliation(s)
- Ronit Vogt Sionov
- The Biofilm Research Laboratory, The Institute of Biomedical and Oral Research, The Faculty of Dental Medicine, Hadassah Medical School, The Hebrew University, Jerusalem 9112102, Israel;
| | | |
Collapse
|
16
|
Marín-Miret J, González-Serrano F, Rosas T, Baixeras J, Latorre A, Pérez-Cobas AE, Moya A. Temporal variations shape the gut microbiome ecology of the moth Brithys crini. Environ Microbiol 2022; 24:3939-3953. [PMID: 35243736 DOI: 10.1111/1462-2920.15952] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 02/18/2022] [Accepted: 02/22/2022] [Indexed: 11/30/2022]
Affiliation(s)
- Jesús Marín-Miret
- Institute for Integrative Systems Biology (I2SysBio), University of Valencia and CSIC, Valencia, Spain
| | - Francisco González-Serrano
- Institute for Integrative Systems Biology (I2SysBio), University of Valencia and CSIC, Valencia, Spain.,Centro de Ciencias Genómicas, Universidad Nacional Autónoma de México, Cuernavaca, Mexico
| | - Tania Rosas
- Cavanilles Institute of Biodiversity and Evolutionary Biology, University of Valencia, Valencia, Spain
| | - Joaquín Baixeras
- Cavanilles Institute of Biodiversity and Evolutionary Biology, University of Valencia, Valencia, Spain
| | - Amparo Latorre
- Institute for Integrative Systems Biology (I2SysBio), University of Valencia and CSIC, Valencia, Spain.,Genomics and Health Area, Foundation for the Promotion of Sanitary and Biomedical Research (FISABIO), Valencia, Spain.,Biomedical Research Center Network of Epidemiology and Public Health (CIBEResp), Madrid, Spain
| | - Ana Elena Pérez-Cobas
- Department of Microbiology, Ramón y Cajal Institute for Health Research (IRYCIS), Ramón y Cajal University Hospital, Madrid, Spain
| | - Andrés Moya
- Institute for Integrative Systems Biology (I2SysBio), University of Valencia and CSIC, Valencia, Spain.,Genomics and Health Area, Foundation for the Promotion of Sanitary and Biomedical Research (FISABIO), Valencia, Spain.,Biomedical Research Center Network of Epidemiology and Public Health (CIBEResp), Madrid, Spain
| |
Collapse
|
17
|
Dey N, Kamatchi C, Vickram AS, Anbarasu K, Thanigaivel S, Palanivelu J, Pugazhendhi A, Ponnusamy VK. Role of nanomaterials in deactivating multiple drug resistance efflux pumps - A review. ENVIRONMENTAL RESEARCH 2022; 204:111968. [PMID: 34453898 DOI: 10.1016/j.envres.2021.111968] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 08/05/2021] [Accepted: 08/19/2021] [Indexed: 06/13/2023]
Abstract
The changes in lifestyle and living conditions have affected not only humans but also microorganisms. As man invents new drugs and therapies, pathogens alter themselves to survive and thrive. Multiple drug resistance (MDR) is the talk of the town for decades now. Many generations of medications have been termed useless as MDR rises among the infectious population. The surge in nanotechnology has brought a new hope in reducing this aspect of resistance in pathogens. It has been observed in several laboratory-based studies that the use of nanoparticles had a synergistic effect on the antibiotic being administered to the pathogen; several resistant strains scummed to the stress created by the nanoparticles and became susceptible to the drug. The major cause of resistance to date is the efflux system, which makes the latest generation of antibiotics ineffective without reaching the target site. If species-specific nanomaterials are used to control the activity of efflux pumps, it could revolutionize the field of medicine and make the previous generation resistant medications active once again. Therefore, the current study was devised to assess and review nanoparticles' role on efflux systems and discuss how specialized particles can be designed towards an infectious host's particular drug ejection systems.
Collapse
Affiliation(s)
- Nibedita Dey
- Department of Biotechnology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - C Kamatchi
- Department of Biotechnology, The Oxford College of Science, Bengaluru, India
| | - A S Vickram
- Department of Biotechnology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - K Anbarasu
- Department of Bioinformatics, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - S Thanigaivel
- Department of Biomedical Engineering, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - Jeyanthi Palanivelu
- Department of Biotechnology, Vel Tech Rangarajan Dr. Sagunthala R&D Institute of Science and Technology, Chennai, India
| | | | - Vinoth Kumar Ponnusamy
- Department of Medicinal and Applied Chemistry & Research Center for Environmental Medicine, Kaohsiung Medical University (KMU), Kaohsiung City, 807, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital (KMUH), Kaohsiung City, 807, Taiwan; Program of Aquatic Science and Technology, College of Hydrosphere Science, National Kaohsiung University of Science and Technology (NKUST), Kaohsiung City, Taiwan.
| |
Collapse
|
18
|
Shang Y, Lv P, Su D, Li Y, Liang Y, Ma C, Yang C. Evolutionary conservative analysis revealed novel functional sites in the efflux pump NorA of Staphylococcus aureus. J Antimicrob Chemother 2021; 77:675-681. [PMID: 34910133 DOI: 10.1093/jac/dkab453] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 11/10/2021] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVES The NorA antiporter of Staphylococcus aureus belongs to the major facilitator superfamily (MFS) and extrudes various kinds of drugs. With no structure available for this drug efflux pump, the aim of this study was to explore its important structural elements that contribute to substrate binding and drug transport. METHODS Evolutionary conservative analyses were conducted on different compilations of NorA homologues to identify conservative motifs and residues. Site-directed mutations were constructed to verify the functional changes in NorA efflux capacities and the conformational changes were further measured by fluorescence resonance energy transfer (FRET) and microscale thermophoresis (MST) analysis. RESULTS Besides Motif-A, Motif-B and Motif-C that were reported previously in MFS proteins, two other motifs, Motif-1 and Motif-2, were identified in NorA. Site-directed mutations of Motif-1 and Motif-2 as well as 11 predicted binding sites all caused remarkable reductions in drug resistance and efflux activity. Among these, mutant F16A/E222A/F303A/D307A showed an altered binding affinity for tetraphenylphosphonium chloride when measured by MST and Motif-1 mutant G114D/A117E/D118G/V119I and Motif-2 mutant Q325E/G326E/A328E/G330E displayed obvious conformational alterations when compared with the wild-type NorA in the FRET signal spectra. CONCLUSIONS The NorA structure agrees well with the typical structures of MFS proteins, with two newly identified motifs (Motif-1 and Motif-2) that are critical to the structural stability of NorA, and sites F16, E222, F303 and D307 are involved in substrate binding.
Collapse
Affiliation(s)
- Yan Shang
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao 266237, P. R. China
| | - Peiwen Lv
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao 266237, P. R. China
| | - Dandan Su
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao 266237, P. R. China
| | - Yaru Li
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao 266237, P. R. China
| | - Yu Liang
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao 266237, P. R. China
| | - Cuiqing Ma
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao 266237, P. R. China
| | - Chunyu Yang
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao 266237, P. R. China
| |
Collapse
|
19
|
Abeywickrama TD, Perera IC. In Silico Characterization and Virtual Screening of GntR/HutC Family Transcriptional Regulator MoyR: A Potential Monooxygenase Regulator in Mycobacterium tuberculosis. BIOLOGY 2021; 10:biology10121241. [PMID: 34943156 PMCID: PMC8698889 DOI: 10.3390/biology10121241] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 07/01/2021] [Accepted: 07/02/2021] [Indexed: 12/31/2022]
Abstract
Simple Summary In an era where the world faces new diseases and pathogens, another emerging challenge is neglected pathogens becoming more notorious. Transcriptional regulators play a vital role in the pathogenesis and survival of these pathogens. Hence, characterizing transcriptional regulators, either in vitro or in silico, is of great importance. Here, we present the first structural characterization of a GntR/HutC regulator in Mycobacterium tuberculosis via in silico methods. We have suggested its possible role and potential as a drug target as well as identified possible drug leads that can be used for further improvements. Abstract Mycobacterium tuberculosis is a well-known pathogen due to the emergence of drug resistance associated with it, where transcriptional regulators play a key role in infection, colonization and persistence. The genome of M. tuberculosis encodes many transcriptional regulators, and here we report an in-depth in silico characterization of a GntR regulator: MoyR, a possible monooxygenase regulator. Homology modelling provided a reliable structure for MoyR, showing homology with a HutC regulator DasR from Streptomyces coelicolor. In silico physicochemical analysis revealed that MoyR is a cytoplasmic protein with higher thermal stability and higher pI. Four highly probable binding pockets were determined in MoyR and the druggability was higher in the orthosteric binding site consisting of three conserved critical residues: TYR179, ARG223 and GLU234. Two highly conserved leucine residues were identified in the effector-binding region of MoyR and other HutC homologues, suggesting that these two residues can be crucial for structure stability and oligomerization. Virtual screening of drug leads resulted in four drug-like compounds with greater affinity to MoyR with potential inhibitory effects for MoyR. Our findings support that this regulator protein can be valuable as a therapeutic target that can be used for developing drug leads.
Collapse
|
20
|
Rodrigues Dos Santos Barbosa C, Feitosa Muniz D, Silvino Pereira P, Maria de Arruda Lima S, Datiane de Morais Oliveira Tintino C, Cintia Alexandrino de Souza V, Mariana Assis da Silva J, Henrique Sousa da Costa R, Cosmo Andrade Pinheiro J, Maria Lobo Soares de Matos Y, Rose Alencar Menezes I, Gonçalves da Silva T, Manoella de Souza Lima G, Cristina Leal Balbino T, Pinto Siqueira-Júnior J, Assis Bezerra da Cunha F, Douglas Melo Coutinho H, Relison Tintino S. Evaluation of Elaiophylin extracted from Streptomyces hygroscopicus as a potential inhibitor of the NorA efflux protein in Staphylococcus aureus: An in vitro and in silico approach. Bioorg Med Chem Lett 2021; 50:128334. [PMID: 34425202 DOI: 10.1016/j.bmcl.2021.128334] [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: 04/30/2021] [Revised: 08/13/2021] [Accepted: 08/16/2021] [Indexed: 10/20/2022]
Abstract
Compounds capable of inhibiting the efflux pump mechanism are a promising alternative against bacterial resistance because, when combined with antibiotics, they can increase the effectiveness of these drugs by inhibiting active efflux. Elaiophylin, derived from Streptomyces hygroscopicus, is a natural antibiotic that exhibits a variety of biological activities, including antibacterial activity. However, its potential as an inhibitor of the bacterial efflux mechanism has not been investigated. This study evaluated the ability of Elaiophylin to inhibit the NorA efflux pump in Staphylococcus aureus strains. Therefore, tests were performed to obtain the Minimum Inhibitory Concentration (MIC) and to verify the ability of Elaiophylin to potentiate the MIC of the antibiotic Norfloxacin and Ethidium Bromide (EtBr), known substrates of NorA efflux. Real-time PCR and molecular docking assays were also performed to assess the potential of Elaiophylin against NorA. The strains SA-1199 (wild type) and SA-1199B (NorA over-expressed) of S. aureus were used for this study. The results showed that Elaiophylin significantly decreased the MIC of Norfloxacin and EtBr, increasing the activity of these substrates against S. aureus, which carries the efflux protein NorA. However, Elaiophylin provided a non-significant reduction in norA gene expression, however, molecular docking demonstrated a high binding affinity between Elaiophylin and NorA efflux protein, indicating that Elaiophylin can act as a potential NorA in S. aureus.
Collapse
Affiliation(s)
| | - Débora Feitosa Muniz
- Laboratory of Microbiology and Molecular Biology (LMBM), Department of Biological Chemistry/CCBS/URCA, Brazil
| | | | | | | | | | | | | | | | | | - Irwin Rose Alencar Menezes
- Laboratory of Microbiology and Molecular Biology (LMBM), Department of Biological Chemistry/CCBS/URCA, Brazil
| | | | | | | | | | | | | | - Saulo Relison Tintino
- Laboratory of Microbiology and Molecular Biology (LMBM), Department of Biological Chemistry/CCBS/URCA, Brazil
| |
Collapse
|
21
|
Martini CL, Coronado AZ, Melo MCN, Gobbi CN, Lopez ÚS, de Mattos MC, Amorim TT, Botelho AMN, Vasconcelos ATR, Almeida LGP, Planet PJ, Zingali RB, Figueiredo AMS, Ferreira-Carvalho BT. Cellular Growth Arrest and Efflux Pumps Are Associated With Antibiotic Persisters in Streptococcus pyogenes Induced in Biofilm-Like Environments. Front Microbiol 2021; 12:716628. [PMID: 34621249 PMCID: PMC8490960 DOI: 10.3389/fmicb.2021.716628] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 08/23/2021] [Indexed: 11/13/2022] Open
Abstract
Streptococcus pyogenes (group A Streptococcus-GAS) is an important pathogen for humans. GAS has been associated with severe and invasive diseases. Despite the fact that these bacteria remain universally susceptible to penicillin, therapeutic failures have been reported in some GAS infections. Many hypotheses have been proposed to explain these antibiotic-unresponsive infections; however, none of them have fully elucidated this phenomenon. In this study, we show that GAS strains have the ability to form antimicrobial persisters when inoculated on abiotic surfaces to form a film of bacterial agglomerates (biofilm-like environment). Our data suggest that efflux pumps were possibly involved in this phenomenon. In fact, gene expression assays by real-time qRT-PCR showed upregulation of some genes associated with efflux pumps in persisters arising in the presence of penicillin. Phenotypic reversion assay and whole-genome sequencing indicated that this event was due to non-inherited resistance mechanisms. The persister cells showed downregulation of genes associated with protein biosynthesis and cell growth, as demonstrated by gene expression assays. Moreover, the proteomic analysis revealed that susceptible cells express higher levels of ribosome proteins. It is remarkable that previous studies have reported the recovery of S. pyogenes viable cells from tissue biopsies of patients presented with GAS invasive infections and submitted to therapy with antibiotics. The persistence phenomenon described herein brings new insights into the origin of therapeutic failures in S. pyogenes infections. Multifactorial mechanisms involving protein synthesis inhibition, cell growth impairment and efflux pumps seem to play roles in the formation of antimicrobial persisters in S. pyogenes.
Collapse
Affiliation(s)
- Caroline Lopes Martini
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Amada Zambrana Coronado
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Maria Celeste Nunes Melo
- Departamento de Microbiologia e Parasitologia, Universidade Federal do Rio Grande do Norte, Natal, Brazil
| | - Clarice Neffa Gobbi
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Úrsula Santos Lopez
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcos Correa de Mattos
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Thais Tavares Amorim
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana Maria Nunes Botelho
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | - Paul J Planet
- Department of Pediatrics, Perelman College of Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Sackler Institute for Comparative Genomics, American Museum of Natural History, New York, NY, United States.,Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Russolina Benedeta Zingali
- Unidade de Espectrometria de Massas e Proteomica - UEMP, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Agnes Marie Sá Figueiredo
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | |
Collapse
|
22
|
Almeida BC, Kaczmarek JA, Figueiredo PR, Prather KLJ, Carvalho ATP. Transcription factor allosteric regulation through substrate coordination to zinc. NAR Genom Bioinform 2021; 3:lqab033. [PMID: 33987533 PMCID: PMC8092373 DOI: 10.1093/nargab/lqab033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 03/30/2021] [Accepted: 04/08/2021] [Indexed: 11/14/2022] Open
Abstract
The development of new synthetic biology circuits for biotechnology and medicine requires deeper mechanistic insight into allosteric transcription factors (aTFs). Here we studied the aTF UxuR, a homodimer of two domains connected by a highly flexible linker region. To explore how ligand binding to UxuR affects protein dynamics we performed molecular dynamics simulations in the free protein, the aTF bound to the inducer D-fructuronate or the structural isomer D-glucuronate. We then validated our results by constructing a sensor plasmid for D-fructuronate in Escherichia coli and performed site-directed mutagenesis. Our results show that zinc coordination is necessary for UxuR function since mutation to alanines prevents expression de-repression by D-fructuronate. Analyzing the different complexes, we found that the disordered linker regions allow the N-terminal domains to display fast and large movements. When the inducer is bound, UxuR can sample an open conformation with a more pronounced negative charge at the surface of the N-terminal DNA binding domains. In opposition, in the free and D-glucuronate bond forms the protein samples closed conformations, with a more positive character at the surface of the DNA binding regions. These molecular insights provide a new basis to harness these systems for biological systems engineering.
Collapse
Affiliation(s)
- Beatriz C Almeida
- CNC-Center for Neuroscience and Cell Biology, Institute for Interdisciplinary Research (IIIUC), University of Coimbra, 3004-504 Coimbra, Portugal
| | - Jennifer A Kaczmarek
- MIT-Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Pedro R Figueiredo
- CNC-Center for Neuroscience and Cell Biology, Institute for Interdisciplinary Research (IIIUC), University of Coimbra, 3004-504 Coimbra, Portugal
| | - Kristala L J Prather
- MIT-Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Alexandra T P Carvalho
- CNC-Center for Neuroscience and Cell Biology, Institute for Interdisciplinary Research (IIIUC), University of Coimbra, 3004-504 Coimbra, Portugal
| |
Collapse
|
23
|
Tong C, Hu H, Chen G, Li Z, Li A, Zhang J. Disinfectant resistance in bacteria: Mechanisms, spread, and resolution strategies. ENVIRONMENTAL RESEARCH 2021; 195:110897. [PMID: 33617866 DOI: 10.1016/j.envres.2021.110897] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 02/02/2021] [Accepted: 02/15/2021] [Indexed: 05/19/2023]
Abstract
Disinfectants are widely acknowledged for removing microorganisms from the surface of the objects and transmission media. However, the emergence of disinfectant resistance has become a severe threat to the safety of life and health and the rational allocation of resources due to the reduced disinfectant effectiveness. The horizontal gene transfer (HGT) of disinfectant resistance genes has also expanded the resistant flora, making the situation worse. This review focused on the resistance mechanisms of disinfectant resistant bacteria on biofilms, cell membrane permeability, efflux pumps, degradable enzymes, and disinfectant targets. Efflux can be the fastest and most effective resistance mechanism for bacteria to respond to stress. The qac genes, located on some plasmids which can transmit resistance through conjugative transfer, are the most commonly reported in the study of disinfectant resistance genes. Whether the qac genes can be transferred through transformation or transduction is still unclear. Studying the factors affecting the resistance of bacteria to disinfectants can find breakthrough methods to more adequately deal with the problem of reduced disinfectant effectiveness. It has been confirmed that the interaction of probiotics and bacteria or the addition of 4-oxazolidinone can inhibit the formation of biofilms. Chemicals such as eugenol and indole derivatives can increase bacterial sensitivity by reducing the expression of efflux pumps. The role of these findings in anti-disinfectant resistance has proved invaluable.
Collapse
Affiliation(s)
- Chaoyu Tong
- Collage of Environmental Science and Engineering, Ocean University of China, Qingdao, 266100, China.
| | - Hong Hu
- Collage of Environmental Science and Engineering, Ocean University of China, Qingdao, 266100, China.
| | - Gang Chen
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China.
| | - Zhengyan Li
- Collage of Environmental Science and Engineering, Ocean University of China, Qingdao, 266100, China.
| | - Aifeng Li
- Collage of Environmental Science and Engineering, Ocean University of China, Qingdao, 266100, China.
| | - Jianye Zhang
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China.
| |
Collapse
|
24
|
Pain M, Wolden R, Jaén-Luchoro D, Salvà-Serra F, Iglesias BP, Karlsson R, Klingenberg C, Cavanagh JP. Staphylococcus borealis sp. nov., isolated from human skin and blood. Int J Syst Evol Microbiol 2021; 70:6067-6078. [PMID: 33048039 DOI: 10.1099/ijsem.0.004499] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
When analysing a large cohort of Staphylococcus haemolyticus, using whole-genome sequencing, five human isolates (four from the skin and one from a blood culture) with aberrant phenotypic and genotypic traits were identified. They were phenotypically similar with yellow colonies, nearly identical 16S rRNA gene sequences and initially speciated as S. haemolyticus based on 16S rRNA gene sequence and MALDI-TOF MS. However, compared to S. haemolyticus, these five strains demonstrate: (i) considerable phylogenetic distance with an average nucleotide identity <95 % and inferred DNA-DNA hybridization <70 %; (ii) a pigmented phenotype; (iii) urease production; and (iv) different fatty acid composition. Based on the phenotypic and genotypic results, we conclude that these strains represent a novel species, for which the name Staphylococcus borealis sp. nov. is proposed. The novel species belong to the genus Staphylococcus and is coagulase- and oxidase-negative and catalase-positive. The type strain, 51-48T, is deposited in the Culture Collection University of Gothenburg (CCUG 73747T) and in the Spanish Type Culture Collection (CECT 30011T).
Collapse
Affiliation(s)
- Maria Pain
- Pediatric Infection Group, Department of Clinical Medicine, UiT The Arctic University of Norway, Tromsø, Norway
| | - Runa Wolden
- Pediatric Infection Group, Department of Clinical Medicine, UiT The Arctic University of Norway, Tromsø, Norway
| | - Daniel Jaén-Luchoro
- Centre for Antibiotic Resistance Research (CARe), University of Gothenburg, Gothenburg, Sweden.,Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Francisco Salvà-Serra
- Department of Clinical Microbiology, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden.,Culture Collection University of Gothenburg (CCUG), Sahlgrenska Academy, University of Gothenburg, Sweden.,Microbiology, Department of Biology, University of the Balearic Islands, Palma de Mallorca, Spain.,Centre for Antibiotic Resistance Research (CARe), University of Gothenburg, Gothenburg, Sweden
| | - Beatriz Piñeiro Iglesias
- Department of Clinical Microbiology, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden.,Centre for Antibiotic Resistance Research (CARe), University of Gothenburg, Gothenburg, Sweden
| | - Roger Karlsson
- Department of Clinical Microbiology, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden.,Centre for Antibiotic Resistance Research (CARe), University of Gothenburg, Gothenburg, Sweden
| | - Claus Klingenberg
- Department of Paediatrics, University Hospital of North Norway, Tromsø, Norway.,Pediatric Infection Group, Department of Clinical Medicine, UiT The Arctic University of Norway, Tromsø, Norway
| | - Jorunn Pauline Cavanagh
- Department of Paediatrics, University Hospital of North Norway, Tromsø, Norway.,Pediatric Infection Group, Department of Clinical Medicine, UiT The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
25
|
Antimicrobial resistance genes in raw milk for human consumption. Sci Rep 2020; 10:7464. [PMID: 32366826 PMCID: PMC7198526 DOI: 10.1038/s41598-020-63675-4] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 04/03/2020] [Indexed: 12/19/2022] Open
Abstract
The increasing prevalence of antimicrobial resistance (AMR) is a significant threat to global health. More and more multi-drug-resistant bacterial strains cause life-threatening infections and the death of thousands of people each year. Beyond disease control animals are often given antibiotics for growth promotion or increased feed efficiency, which further increase the chance of the development of multi-resistant strains. After the consumption of unprocessed animal products, these strains may meet the human bacteriota. Among the foodborne and the human populations, antimicrobial resistance genes (ARGs) may be shared by horizontal gene transfer. This study aims to test the presence of antimicrobial resistance genes in milk metagenome, investigate their genetic position and their linkage to mobile genetic elements. We have analyzed raw milk samples from public markets sold for human consumption. The milk samples contained genetic material from various bacterial species and the in-depth analysis uncovered the presence of several antimicrobial resistance genes. The samples contained complete ARGs influencing the effectiveness of acridine dye, cephalosporin, cephamycin, fluoroquinolone, penam, peptide antibiotics and tetracycline. One of the ARGs, PC1 beta-lactamase may also be a mobile element that facilitates the transfer of resistance genes to other bacteria, e.g. to the ones living in the human gut.
Collapse
|
26
|
Epidemiology of efflux pumps genes mediating resistance among Staphylococcus aureus; A systematic review. Microb Pathog 2019; 139:103850. [PMID: 31706002 DOI: 10.1016/j.micpath.2019.103850] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 10/22/2019] [Accepted: 11/05/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND Efflux of antibiotics is an effective resistance mechanism among antibiotic-resistant Staphylococcus aureus. This systematic review aims to evaluate the frequency and expression of efflux pump genes in S.aureus around the world. METHOD A comprehensive literature search of several databases (Medline Pub Med, ISI, Scopus, Google Scholar, ISC, Science direct and Persian Journals Online, and citation lists) was performed. We considered published studies from 2001 to 2018. Articles reporting the prevalence and expression of efflux pump genes were selected. RESULT Among 183 articles, 36 studies were selected. Of the 36, 23 articles were conducted in Asia.6 in Europe, 5 in America and 2 in African countries. In most of these studies norA, norB, qacA/B genes were commonly evaluated by molecular methods. The presence of efflux pump genes such as norA, norB, norC, mepA, mdeA, qacA/B was detected by PCR in 21 studies and over-expression of genes were reported in 13 studies. The most frequently reported genes in Asia were norA (75%), norB (60%), mepA (35%), mdeA (33%) and qacA/B (20.8%). In European studies, the prevalence of norB was mostly reported among S.aureus isolates and norA and qacA/B were commonly found in similar studies in America. The investigation of gene expression patterns showed that norA was most frequent single-pattern in Asia and America, norB or mdeA in Europe. CONCLUSION According to this study MDR efflux pumps not only cause high-level resistance but also it considerably associated with over-expression of these genes. Due to the selective pressure on MRSA isolate, the enormous diversity of plasmid-encoded genes had been recorded in different regions, owing to the various numbers and types of isolates in each study or types of disinfectants for general use.
Collapse
|
27
|
Multidrug ABC transporters in bacteria. Res Microbiol 2019; 170:381-391. [DOI: 10.1016/j.resmic.2019.06.001] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Revised: 06/12/2019] [Accepted: 06/17/2019] [Indexed: 12/23/2022]
|
28
|
Bai J, Zhu X, Zhao K, Yan Y, Xu T, Wang J, Zheng J, Huang W, Shi L, Shang Y, Lv Z, Wang X, Wu Y, Qu D. The role of ArlRS in regulating oxacillin susceptibility in methicillin-resistant Staphylococcus aureus indicates it is a potential target for antimicrobial resistance breakers. Emerg Microbes Infect 2019; 8:503-515. [PMID: 30924407 PMCID: PMC6455253 DOI: 10.1080/22221751.2019.1595984] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA), also known as oxacillin-resistant S. aureus, is a leading cause of community and hospital associated infections globally. In this work, we found that deletion of the arlRS two-component system genes in the USA300 and USA500 strains resulted in increased susceptibilities to oxacillin (8–16-fold decrease in minimal inhibitory concentrations). In USA300ΔarlRS, transcriptional levels of mecA or blaZ showed no obvious change, while mRNA levels of spx showed a 4-fold decrease at 4 h and a 6.3-fold decrease at 10 h. Overexpression of spx in ΔarlRS restored oxacillin resistance to a similar level in USA300. In addition, gel shift assay showed that the recombinant ArlR bound to spx promoter region. Furthermore, silencing of spx led to a significant increase of oxacillin susceptibility in multiple MRSA isolates. Our results indicate that ArlRS plays a strong role in regulating oxacillin resistance in MRSA strains, which involves direct modulation of spx expression. Moreover, oritavancin showed inhibition to ATPase activity of the recombinant histidine kinase ArlS (IC50 = 5.47 μM). Oritavancin had synergy effect on oxacillin activity against the MRSA strains in both planktonic and biofilm state. Our data suggest that ArlRS is an attractive target for breaking antimicrobial resistance of MRSA.
Collapse
Affiliation(s)
- Jinna Bai
- a Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences , Shanghai Medical College of Fudan University , Shanghai , People's Republic of China
| | - Xiaoyi Zhu
- a Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences , Shanghai Medical College of Fudan University , Shanghai , People's Republic of China
| | - Keqing Zhao
- b Department of Otorhinolaryngology-Head and Neck Surgery, Eye & ENT Hospital, Shanghai Key Clinical Disciplines of otorhinolaryngology , Fudan University , Shanghai , People's Republic of China
| | - Yingjie Yan
- a Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences , Shanghai Medical College of Fudan University , Shanghai , People's Republic of China
| | - Tao Xu
- c Key Laboratory of Medical Molecular Virology , Huashan Hospital, Shanghai Medical College of Fudan University , Shanghai , People's Republic of China
| | - Jiaxue Wang
- d Department of Laboratory Medicine , Hangzhou Medical College , Hangzhou , Zhejiang , People's Republic of China
| | - Jinxing Zheng
- a Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences , Shanghai Medical College of Fudan University , Shanghai , People's Republic of China.,e Department of Infectious Diseases and the Key Laboratory of Endogenous Infection , Shenzhen Nanshan People's Hospital of Shenzhen University , Shenzhen , People's Republic of China
| | - Wei Huang
- f Medical Clinic, Hangzhou Haiqin Sanatorium , Hangzhou , Zhejiang , People's Republic of China
| | - Le Shi
- b Department of Otorhinolaryngology-Head and Neck Surgery, Eye & ENT Hospital, Shanghai Key Clinical Disciplines of otorhinolaryngology , Fudan University , Shanghai , People's Republic of China
| | - Yongpeng Shang
- a Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences , Shanghai Medical College of Fudan University , Shanghai , People's Republic of China
| | - Zhihui Lv
- a Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences , Shanghai Medical College of Fudan University , Shanghai , People's Republic of China
| | - Xiaofei Wang
- a Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences , Shanghai Medical College of Fudan University , Shanghai , People's Republic of China
| | - Yang Wu
- a Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences , Shanghai Medical College of Fudan University , Shanghai , People's Republic of China
| | - Di Qu
- a Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences , Shanghai Medical College of Fudan University , Shanghai , People's Republic of China
| |
Collapse
|
29
|
Phillips-Jones MK, Harding SE. Antimicrobial resistance (AMR) nanomachines-mechanisms for fluoroquinolone and glycopeptide recognition, efflux and/or deactivation. Biophys Rev 2018; 10:347-362. [PMID: 29525835 PMCID: PMC5899746 DOI: 10.1007/s12551-018-0404-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 02/05/2018] [Indexed: 12/11/2022] Open
Abstract
In this review, we discuss mechanisms of resistance identified in bacterial agents Staphylococcus aureus and the enterococci towards two priority classes of antibiotics-the fluoroquinolones and the glycopeptides. Members of both classes interact with a number of components in the cells of these bacteria, so the cellular targets are also considered. Fluoroquinolone resistance mechanisms include efflux pumps (MepA, NorA, NorB, NorC, MdeA, LmrS or SdrM in S. aureus and EfmA or EfrAB in the enterococci) for removal of fluoroquinolone from the intracellular environment of bacterial cells and/or protection of the gyrase and topoisomerase IV target sites in Enterococcus faecalis by Qnr-like proteins. Expression of efflux systems is regulated by GntR-like (S. aureus NorG), MarR-like (MgrA, MepR) regulators or a two-component signal transduction system (TCS) (S. aureus ArlSR). Resistance to the glycopeptide antibiotic teicoplanin occurs via efflux regulated by the TcaR regulator in S. aureus. Resistance to vancomycin occurs through modification of the D-Ala-D-Ala target in the cell wall peptidoglycan and removal of high affinity precursors, or by target protection via cell wall thickening. Of the six Van resistance types (VanA-E, VanG), the VanA resistance type is considered in this review, including its regulation by the VanSR TCS. We describe the recent application of biophysical approaches such as the hydrodynamic technique of analytical ultracentrifugation and circular dichroism spectroscopy to identify the possible molecular effector of the VanS receptor that activates expression of the Van resistance genes; both approaches demonstrated that vancomycin interacts with VanS, suggesting that vancomycin itself (or vancomycin with an accessory factor) may be an effector of vancomycin resistance. With 16 and 19 proteins or protein complexes involved in fluoroquinolone and glycopeptide resistances, respectively, and the complexities of bacterial sensing mechanisms that trigger and regulate a wide variety of possible resistance mechanisms, we propose that these antimicrobial resistance mechanisms might be considered complex 'nanomachines' that drive survival of bacterial cells in antibiotic environments.
Collapse
Affiliation(s)
- Mary K Phillips-Jones
- National Centre for Macromolecular Hydrodynamics, School of Biosciences, University of Nottingham, Sutton Bonington, LE12 5RD, Loughborough, Leicestershire, UK.
| | - Stephen E Harding
- National Centre for Macromolecular Hydrodynamics, School of Biosciences, University of Nottingham, Sutton Bonington, LE12 5RD, Loughborough, Leicestershire, UK
| |
Collapse
|
30
|
Genome-wide screen for genes involved in eDNA release during biofilm formation by Staphylococcus aureus. Proc Natl Acad Sci U S A 2017; 114:E5969-E5978. [PMID: 28674000 DOI: 10.1073/pnas.1704544114] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Staphylococcus aureus is a leading cause of both nosocomial and community-acquired infection. Biofilm formation at the site of infection reduces antimicrobial susceptibility and can lead to chronic infection. During biofilm formation, a subset of cells liberate cytoplasmic proteins and DNA, which are repurposed to form the extracellular matrix that binds the remaining cells together in large clusters. Using a strain that forms robust biofilms in vitro during growth under glucose supplementation, we carried out a genome-wide screen for genes involved in the release of extracellular DNA (eDNA). A high-density transposon insertion library was grown under biofilm-inducing conditions, and the relative frequency of insertions was compared between genomic DNA (gDNA) collected from cells in the biofilm and eDNA from the matrix. Transposon insertions into genes encoding functions necessary for eDNA release were identified by reduced representation in the eDNA. On direct testing, mutants of some of these genes exhibited markedly reduced levels of eDNA and a concomitant reduction in cell clustering. Among the genes with robust mutant phenotypes were gdpP, which encodes a phosphodiesterase that degrades the second messenger cyclic-di-AMP, and xdrA, the gene for a transcription factor that, as revealed by RNA-sequencing analysis, influences the expression of multiple genes, including many involved in cell wall homeostasis. Finally, we report that growth in biofilm-inducing medium lowers cyclic-di-AMP levels and does so in a manner that depends on the gdpP phosphodiesterase gene.
Collapse
|
31
|
Yılmaz Ç, Özcengiz G. Antibiotics: Pharmacokinetics, toxicity, resistance and multidrug efflux pumps. Biochem Pharmacol 2017; 133:43-62. [DOI: 10.1016/j.bcp.2016.10.005] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 10/14/2016] [Indexed: 02/03/2023]
|
32
|
Effects of Luteolin and Quercetin in Combination with Some Conventional Antibiotics against Methicillin-Resistant Staphylococcus aureus. Int J Mol Sci 2016; 17:ijms17111947. [PMID: 27879665 PMCID: PMC5133941 DOI: 10.3390/ijms17111947] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 11/11/2016] [Accepted: 11/14/2016] [Indexed: 12/04/2022] Open
Abstract
The present study was designed to evaluate the effects of flavonoids luteolin (L) and quercetin + luteolin (Q + L) in combination with commonly used antibacterial agents against methicillin-resistant Staphylococcus aureus (MRSA) clinical isolates and S. aureus (ATCC 43300). Minimum inhibitory concentrations (MICs) of L and Q + L, as well as the MICs of flavonoids in combination with antibiotics were determined and results showed an increased activity of flavonoids with antibiotics. The synergistic, additive, or antagonistic relationships between flavonoids (L and Q + L) and antibiotics were also evaluated, and additive and synergistic effects were observed for some antibiotic + flavonoid combinations. In addition, some combinations were also found to damage the bacterial cytoplasmic membrane, as assessed through potassium leakage assay. The effects of flavonoids and flavonoids + antibiotics on mecA gene mutations were also tested, and no functional variation was detected in the coding region.
Collapse
|
33
|
Hooper DC, Jacoby GA. Topoisomerase Inhibitors: Fluoroquinolone Mechanisms of Action and Resistance. Cold Spring Harb Perspect Med 2016; 6:cshperspect.a025320. [PMID: 27449972 DOI: 10.1101/cshperspect.a025320] [Citation(s) in RCA: 303] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Quinolone antimicrobials are widely used in clinical medicine and are the only current class of agents that directly inhibit bacterial DNA synthesis. Quinolones dually target DNA gyrase and topoisomerase IV binding to specific domains and conformations so as to block DNA strand passage catalysis and stabilize DNA-enzyme complexes that block the DNA replication apparatus and generate double breaks in DNA that underlie their bactericidal activity. Resistance has emerged with clinical use of these agents and is common in some bacterial pathogens. Mechanisms of resistance include mutational alterations in drug target affinity and efflux pump expression and acquisition of resistance-conferring genes. Resistance mutations in one or both of the two drug target enzymes are commonly in a localized domain of the GyrA and ParC subunits of gyrase and topoisomerase IV, respectively, and reduce drug binding to the enzyme-DNA complex. Other resistance mutations occur in regulatory genes that control the expression of native efflux pumps localized in the bacterial membrane(s). These pumps have broad substrate profiles that include other antimicrobials as well as quinolones. Mutations of both types can accumulate with selection pressure and produce highly resistant strains. Resistance genes acquired on plasmids confer low-level resistance that promotes the selection of mutational high-level resistance. Plasmid-encoded resistance is because of Qnr proteins that protect the target enzymes from quinolone action, a mutant aminoglycoside-modifying enzyme that also modifies certain quinolones, and mobile efflux pumps. Plasmids with these mechanisms often encode additional antimicrobial resistances and can transfer multidrug resistance that includes quinolones.
Collapse
Affiliation(s)
- David C Hooper
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, Massachusetts 02114
| | - George A Jacoby
- Lahey Hospital and Medical Center, Burlington, Massachusetts 01805
| |
Collapse
|
34
|
Schindler BD, Kaatz GW. Multidrug efflux pumps of Gram-positive bacteria. Drug Resist Updat 2016; 27:1-13. [DOI: 10.1016/j.drup.2016.04.003] [Citation(s) in RCA: 126] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 03/28/2016] [Accepted: 04/22/2016] [Indexed: 11/16/2022]
|
35
|
Blancato VS, Pagliai FA, Magni C, Gonzalez CF, Lorca GL. Functional Analysis of the Citrate Activator CitO from Enterococcus faecalis Implicates a Divalent Metal in Ligand Binding. Front Microbiol 2016; 7:101. [PMID: 26903980 PMCID: PMC4746285 DOI: 10.3389/fmicb.2016.00101] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 01/19/2016] [Indexed: 02/04/2023] Open
Abstract
The regulator of citrate metabolism, CitO, from Enterococcus faecalis belongs to the FCD family within the GntR superfamily. In the presence of citrate, CitO binds to cis-acting sequences located upstream of the cit promoters inducing the expression of genes involved in citrate utilization. The quantification of the molecular binding affinities, performed by isothermal titration calorimetry (ITC), indicated that CitO has a high affinity for citrate (KD = 1.2 ± 0.2 μM), while it did not recognize other metabolic intermediates. Based on a structural model of CitO where a putative small molecule and a metal binding site were identified, it was hypothesized that the metal ion is required for citrate binding. In agreement with this model, citrate binding to CitO sharply decreased when the protein was incubated with EDTA. This effect was reverted by the addition of Ni2+, and Zn2+ to a lesser extent. Structure-based site-directed mutagenesis was conducted and it was found that changes to alanine in residues Arg97 and His191 resulted in decreased binding affinities for citrate, as determined by EMSA and ITC. Further assays using lacZ fusions confirmed that these residues in CitO are involved in sensing citrate in vivo. These results indicate that the molecular modifications induced by a ligand and a metal binding in the C-terminal domain of CitO are required for optimal DNA binding activity, and consequently, transcriptional activation.
Collapse
Affiliation(s)
- Víctor S Blancato
- Laboratorio de Fisiología y Genética de Bacterias Lácticas, Instituto de Biología Molecular de Rosario, Consejo Nacional de Investigaciones Científicas y TécnicasRosario, Argentina; Department of Microbiology and Cell Science, Genetics Institute, Institute of Food and Agricultural Science, University of FloridaGainesville, FL, USA
| | - Fernando A Pagliai
- Department of Microbiology and Cell Science, Genetics Institute, Institute of Food and Agricultural Science, University of Florida Gainesville, FL, USA
| | - Christian Magni
- Laboratorio de Fisiología y Genética de Bacterias Lácticas, Instituto de Biología Molecular de Rosario, Consejo Nacional de Investigaciones Científicas y Técnicas Rosario, Argentina
| | - Claudio F Gonzalez
- Department of Microbiology and Cell Science, Genetics Institute, Institute of Food and Agricultural Science, University of Florida Gainesville, FL, USA
| | - Graciela L Lorca
- Department of Microbiology and Cell Science, Genetics Institute, Institute of Food and Agricultural Science, University of Florida Gainesville, FL, USA
| |
Collapse
|
36
|
Yu L, Gao W, Li S, Pan Y, Liu G. GntR family regulator SCO6256 is involved in antibiotic production and conditionally regulates the transcription of myo-inositol catabolic genes in Streptomyces coelicolor A3(2). MICROBIOLOGY-SGM 2016; 162:537-551. [PMID: 26744083 DOI: 10.1099/mic.0.000235] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
SCO6256 belongs to the GntR family and shows 74% identity with SCO6974, which is the repressor of myo-inositol catabolism in Streptomyces coelicolor A3(2). Disruption of SCO6256 significantly enhanced the transcription of myo-inositol catabolic genes in R2YE medium. The purified recombinant SCO6256 directly bound to the upstream regions of SCO2727, SCO6978 and SCO6985, as well as its encoding gene. Footprinting assays demonstrated that SCO6256 bound to the same sites in the myo-inositol catabolic gene cluster as SCO6974. The expression of SCO6256 was repressed by SCO6974 in minimal medium with myo-inositol as the carbon source, but not in R2YE medium. Glutathione-S-transferase pull-down assays demonstrated that SCO6974 and SCO6256 interacted with each other; and both of the proteins controlled the transcription of myo-inositol catabolic genes in R2YE medium. These results indicated SCO6256 regulates the transcription of myo-inositol catabolic genes in coordination with SCO6974 in R2YE medium. In addition, SCO6256 negatively regulated the production of actinorhodin and calcium-dependent antibiotic via control of the transcription of actII-ORF4 and cdaR. SCO6256 bound to the upstream region of cdaR and the binding sequence was proved to be TTTCGGCACGCAGACAT, which was further confirmed through base substitution. Four putative targets (SCO2652, SCO4034, SCO4237 and SCO6377) of SCO6256 were found by screening the genome sequence of Strep. coelicolor A3(2) based on the conserved binding motif, and confirmed by transcriptional analysis and electrophoretic mobility shift assays. These results revealed that SCO6256 is involved in the regulation of myo-inositol catabolic gene transcription and antibiotic production in Strep. coelicolor A3(2).
Collapse
Affiliation(s)
- Lingjun Yu
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, PR China.,University of the Chinese Academy of Sciences, Beijing 100101, PR China
| | - Wenyan Gao
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, PR China
| | - Shuxian Li
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, PR China
| | - Yuanyuan Pan
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, PR China
| | - Gang Liu
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, PR China
| |
Collapse
|
37
|
Jang S. Multidrug efflux pumps in Staphylococcus aureus and their clinical implications. J Microbiol 2016; 54:1-8. [DOI: 10.1007/s12275-016-5159-z] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Revised: 10/29/2015] [Accepted: 10/30/2015] [Indexed: 12/13/2022]
|
38
|
Hu J, Zhao L, Yang M. A GntR family transcription factor positively regulates mycobacterial isoniazid resistance by controlling the expression of a putative permease. BMC Microbiol 2015; 15:214. [PMID: 26474554 PMCID: PMC4609117 DOI: 10.1186/s12866-015-0556-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 10/08/2015] [Indexed: 01/13/2023] Open
Abstract
Background Bacteria use transcriptional regulation to respond to environmental stresses. Specifically, exposure to antibacterial drugs is deemed to be an atypical stress, and altering transcriptional regulation in response to such stress can increase bacterial drug resistance. However, only a few transcription factors that regulate drug resistance have been reported. Results In the present study, a GntR family transcription factor, encoded by the MSMEG_0535 (Ms0535) gene, was shown to be an isoniazid (INH) resistance regulator in Mycobacterium smegmatis. When the Ms0535 gene was overexpressed, cells showed a significant increase in INH resistance. First, the interaction between Ms0535 and its own promoter was determined, and a conserved 26-bp palindromic DNA binding motif was identified using electrophoretic mobility shift and DNaseI footprinting assays. Second, quantitative reverse transcription-PCR assays showed that Ms0535 acted as a transcriptional activator, and positively regulated its own expression, as well as that of a permease encoded by the MSMEG_0534 (Ms0534) gene. Similar to the case for the Ms0535 gene, a recombinant Ms0534-overexpressing strain also exhibited increased INH resistance compared with the wild-type strain. Furthermore, we showed that Ms0535 and Ms0534 deletion strains were more sensitive to INH than the wild-type strain. Interestingly, overexpressing Ms0534 in the Ms0535 deletion strain enhanced its INH resistance. In contrast, the Ms0534 deletion strain was still sensitive to INH even when Ms0535 was overexpressed. These findings suggest that Ms0534 is an effector protein that affects INH resistance in M. smegmatis. Conclusions In summary, the GntR transcriptional regulator Ms0535 positively regulates INH resistance by transcriptionally regulating the expression of the Ms0534 permease in M. smegmatis. These results improve our understanding of the role of transcriptional regulation in INH drug resistance in mycobacteria. Electronic supplementary material The online version of this article (doi:10.1186/s12866-015-0556-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jialing Hu
- National Key Laboratory of Agricultural Microbiology, Center for Proteomics Research, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Lei Zhao
- National Key Laboratory of Agricultural Microbiology, Center for Proteomics Research, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Min Yang
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China.
| |
Collapse
|
39
|
Role of the Tet38 Efflux Pump in Staphylococcus aureus Internalization and Survival in Epithelial Cells. Infect Immun 2015; 83:4362-72. [PMID: 26324534 DOI: 10.1128/iai.00723-15] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 08/24/2015] [Indexed: 12/22/2022] Open
Abstract
We previously identified the protein Tet38 as a chromosomally encoded efflux pump of Staphylococcus aureus that confers resistance to tetracycline and certain unsaturated fatty acids. Tet38 also contributes to mouse skin colonization. In this study, we discovered a novel regulator of tet38, named tetracycline regulator 21 (TetR21), that bound specifically to the tet38 promoter and repressed pump expression. A ΔtetR21 mutant showed a 5-fold increase in tet38 transcripts and an 8-fold increase in resistance to tetracycline and fatty acids. The global regulator MgrA bound to the tetR21 promoter and indirectly repressed the expression of tet38. To further assess the full role of Tet38 in S. aureus adaptability, we tested its effect on host cell invasion using A549 (lung) and HMEC-1 (heart) cell lines. We used S. aureus RN6390, its Δtet38, ΔtetR21, and ΔmgrA mutants, and a Δtet38 ΔtetR21 double mutant. After 2 h of contact, the Δtet38 mutant was internalized in 6-fold-lower numbers than RN6390 in A549 and HMEC-1 cells, and the ΔtetR21 mutant was internalized in 2-fold-higher numbers than RN6390. A slight increase of 1.5-fold in internalization was found for the ΔmgrA mutant. The growth patterns of RN6390 and the ΔmgrA and ΔtetR21 mutants within A549 cells were similar, while no growth was observed for the Δtet38 mutant. These data indicate that the Tet38 efflux pump is regulated by TetR21 and contributes to the ability of S. aureus to internalize and replicate within epithelial cells.
Collapse
|
40
|
Moche M, Schlüter R, Bernhardt J, Plate K, Riedel K, Hecker M, Becher D. Time-Resolved Analysis of Cytosolic and Surface-Associated Proteins of Staphylococcus aureus HG001 under Planktonic and Biofilm Conditions. J Proteome Res 2015; 14:3804-22. [DOI: 10.1021/acs.jproteome.5b00148] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Martin Moche
- Institute for Microbiology, Ernst-Moritz-Arndt-University, Friedrich-Ludwig-Jahn-Straße
15, 17487 Greifswald, Germany
| | - Rabea Schlüter
- Institute for Microbiology, Ernst-Moritz-Arndt-University, Friedrich-Ludwig-Jahn-Straße
15, 17487 Greifswald, Germany
| | - Jörg Bernhardt
- Institute for Microbiology, Ernst-Moritz-Arndt-University, Friedrich-Ludwig-Jahn-Straße
15, 17487 Greifswald, Germany
| | - Kristina Plate
- Institute for Microbiology, Ernst-Moritz-Arndt-University, Friedrich-Ludwig-Jahn-Straße
15, 17487 Greifswald, Germany
| | - Katharina Riedel
- Institute for Microbiology, Ernst-Moritz-Arndt-University, Friedrich-Ludwig-Jahn-Straße
15, 17487 Greifswald, Germany
| | - Michael Hecker
- Institute for Microbiology, Ernst-Moritz-Arndt-University, Friedrich-Ludwig-Jahn-Straße
15, 17487 Greifswald, Germany
| | - Dörte Becher
- Institute for Microbiology, Ernst-Moritz-Arndt-University, Friedrich-Ludwig-Jahn-Straße
15, 17487 Greifswald, Germany
| |
Collapse
|
41
|
Abstract
Quinolone antimicrobials are synthetic and widely used in clinical medicine. Resistance emerged with clinical use and became common in some bacterial pathogens. Mechanisms of resistance include two categories of mutation and acquisition of resistance-conferring genes. Resistance mutations in one or both of the two drug target enzymes, DNA gyrase and DNA topoisomerase IV, are commonly in a localized domain of the GyrA and ParE subunits of the respective enzymes and reduce drug binding to the enzyme-DNA complex. Other resistance mutations occur in regulatory genes that control the expression of native efflux pumps localized in the bacterial membrane(s). These pumps have broad substrate profiles that include quinolones as well as other antimicrobials, disinfectants, and dyes. Mutations of both types can accumulate with selection pressure and produce highly resistant strains. Resistance genes acquired on plasmids can confer low-level resistance that promotes the selection of mutational high-level resistance. Plasmid-encoded resistance is due to Qnr proteins that protect the target enzymes from quinolone action, one mutant aminoglycoside-modifying enzyme that also modifies certain quinolones, and mobile efflux pumps. Plasmids with these mechanisms often encode additional antimicrobial resistances and can transfer multidrug resistance that includes quinolones. Thus, the bacterial quinolone resistance armamentarium is large.
Collapse
Affiliation(s)
- David C Hooper
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - George A Jacoby
- Lahey Hospital and Medical Center, Burlington, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
42
|
Yoshikai H, Kizaki H, Saito Y, Omae Y, Sekimizu K, Kaito C. Multidrug-Resistance Transporter AbcA Secretes Staphylococcus aureus Cytolytic Toxins. J Infect Dis 2015; 213:295-304. [PMID: 26160745 DOI: 10.1093/infdis/jiv376] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 07/03/2015] [Indexed: 12/21/2022] Open
Abstract
Phenol-soluble modulins (PSMs) are Staphylococcus aureus cytolytic toxins that lyse erythrocytes and neutrophils and have important functions in the S. aureus infectious process. The molecular mechanisms of PSM secretion, however, are not well understood. Here we report that knockout of the multidrug-resistance ABC transporter AbcA, which contributes to S. aureus resistance against antibiotics and chemicals, diminished the secreted amount of PSM, leading to the accumulation of PSM in the intracellular fraction. The amount of PSM in the culture supernatants of the abcA knockout mutants was restored by introduction of the wild-type abcA gene, whereas it was not completely restored by introduction of mutant abcA genes encoding AbcA mutant proteins carrying amino acid substitutions in the adenosine triphosphate binding motifs. The abcA knockout mutant exhibited attenuated virulence in a mouse systemic infection model. These findings suggest that the multidrug resistance transporter AbcA secretes PSMs and contributes to S. aureus virulence.
Collapse
Affiliation(s)
- Hirono Yoshikai
- Laboratory of Microbiology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Japan
| | - Hayato Kizaki
- Laboratory of Microbiology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Japan
| | - Yuki Saito
- Laboratory of Microbiology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Japan
| | - Yosuke Omae
- Laboratory of Microbiology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Japan
| | - Kazuhisa Sekimizu
- Laboratory of Microbiology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Japan
| | - Chikara Kaito
- Laboratory of Microbiology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Japan
| |
Collapse
|
43
|
Andersen JL, He GX, Kakarla P, K C R, Kumar S, Lakra WS, Mukherjee MM, Ranaweera I, Shrestha U, Tran T, Varela MF. Multidrug efflux pumps from Enterobacteriaceae, Vibrio cholerae and Staphylococcus aureus bacterial food pathogens. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2015; 12:1487-547. [PMID: 25635914 PMCID: PMC4344678 DOI: 10.3390/ijerph120201487] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 01/15/2015] [Indexed: 02/07/2023]
Abstract
Foodborne illnesses caused by bacterial microorganisms are common worldwide and constitute a serious public health concern. In particular, microorganisms belonging to the Enterobacteriaceae and Vibrionaceae families of Gram-negative bacteria, and to the Staphylococcus genus of Gram-positive bacteria are important causative agents of food poisoning and infection in the gastrointestinal tract of humans. Recently, variants of these bacteria have developed resistance to medically important chemotherapeutic agents. Multidrug resistant Escherichia coli, Salmonella enterica, Vibrio cholerae, Enterobacter spp., and Staphylococcus aureus are becoming increasingly recalcitrant to clinical treatment in human patients. Of the various bacterial resistance mechanisms against antimicrobial agents, multidrug efflux pumps comprise a major cause of multiple drug resistance. These multidrug efflux pump systems reside in the biological membrane of the bacteria and actively extrude antimicrobial agents from bacterial cells. This review article summarizes the evolution of these bacterial drug efflux pump systems from a molecular biological standpoint and provides a framework for future work aimed at reducing the conditions that foster dissemination of these multidrug resistant causative agents through human populations.
Collapse
Affiliation(s)
- Jody L Andersen
- Department of Biology, Eastern New Mexico University, Portales, NM 88130, USA.
| | - Gui-Xin He
- Department of Clinical Laboratory and Nutritional Sciences, University of Massachusetts Lowell, Lowell, MA 01854, USA.
| | - Prathusha Kakarla
- Department of Biology, Eastern New Mexico University, Portales, NM 88130, USA.
| | - Ranjana K C
- Department of Biology, Eastern New Mexico University, Portales, NM 88130, USA.
| | - Sanath Kumar
- QC Laboratory, Harvest and Post-Harvest Technology Division, Central Institute of Fisheries Education (CIFE), Seven Bungalows, Versova, Andheri (W), Mumbai 400061, India.
| | - Wazir Singh Lakra
- QC Laboratory, Harvest and Post-Harvest Technology Division, Central Institute of Fisheries Education (CIFE), Seven Bungalows, Versova, Andheri (W), Mumbai 400061, India.
| | - Mun Mun Mukherjee
- Department of Biology, Eastern New Mexico University, Portales, NM 88130, USA.
| | - Indrika Ranaweera
- Department of Biology, Eastern New Mexico University, Portales, NM 88130, USA.
| | - Ugina Shrestha
- Department of Biology, Eastern New Mexico University, Portales, NM 88130, USA.
| | - Thuy Tran
- Department of Clinical Laboratory and Nutritional Sciences, University of Massachusetts Lowell, Lowell, MA 01854, USA.
| | - Manuel F Varela
- Department of Biology, Eastern New Mexico University, Portales, NM 88130, USA.
| |
Collapse
|
44
|
Regulation of expression of abcA and its response to environmental conditions. J Bacteriol 2014; 196:1532-9. [PMID: 24509312 DOI: 10.1128/jb.01406-13] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The ATP-dependent transporter gene abcA in Staphylococcus aureus confers resistance to hydrophobic β-lactams. In strain ISP794, abcA is regulated by the transcriptional regulators MgrA and NorG and shares a 420-nucleotide intercistronic region with the divergently transcribed pbp4 gene, which encodes the transpeptidase Pbp4. Exposure of exponentially growing cells to iron-limited media, oxidative stress, and acidic pH (5.5) for 0.5 to 2 h had no effect on abcA expression. In contrast, nutrient limitation produced a significant increase in abcA transcripts. We identified three additional regulators (SarA, SarZ, and Rot) that bind to the overlapping promoter region of abcA and pbp4 in strain MW2 and investigated their role in the regulation of abcA expression. Expression of abcA is decreased by 10.0-fold in vivo in a subcutaneous abscess model. In vitro, abcA expression depends on rot and sarZ regulators. Moenomycin A exposure of strain MW2 produced an increase in abcA transcripts. Relative to MW2, the MIC of moenomycin was decreased 8-fold for MW2ΔabcA and increased 10-fold for the MW2 abcA overexpresser, suggesting that moenomycin is a substrate of AbcA.
Collapse
|
45
|
Modulation of Bacterial Multidrug Resistance Efflux Pumps of the Major Facilitator Superfamily. INTERNATIONAL JOURNAL OF BACTERIOLOGY 2013; 2013. [PMID: 25750934 PMCID: PMC4347946 DOI: 10.1155/2013/204141] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Bacterial infections pose a serious public health concern, especially when an infectious disease has a multidrug resistant causative agent. Such multidrug resistant bacteria can compromise the clinical utility of major chemotherapeutic antimicrobial agents. Drug and multidrug resistant bacteria harbor several distinct molecular mechanisms for resistance. Bacterial antimicrobial agent efflux pumps represent a major mechanism of clinical resistance. The major facilitator superfamily (MFS) is one of the largest groups of solute transporters to date and includes a significant number of bacterial drug and multidrug efflux pumps. We review recent work on the modulation of multidrug efflux pumps, paying special attention to those transporters belonging primarily to the MFS.
Collapse
|
46
|
Costa SS, Viveiros M, Amaral L, Couto I. Multidrug Efflux Pumps in Staphylococcus aureus: an Update. Open Microbiol J 2013; 7:59-71. [PMID: 23569469 PMCID: PMC3617543 DOI: 10.2174/1874285801307010059] [Citation(s) in RCA: 248] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2012] [Revised: 01/24/2013] [Accepted: 01/25/2013] [Indexed: 11/22/2022] Open
Abstract
The emergence of infections caused by multi- or pan-resistant bacteria in the hospital or in the community settings is an increasing health concern. Albeit there is no single resistance mechanism behind multiresistance, multidrug efflux pumps, proteins that cells use to detoxify from noxious compounds, seem to play a key role in the emergence of these multidrug resistant (MDR) bacteria. During the last decades, experimental data has established their contribution to low level resistance to antimicrobials in bacteria and their potential role in the appearance of MDR phenotypes, by the extrusion of multiple, unrelated compounds. Recent studies suggest that efflux pumps may be used by the cell as a first-line defense mechanism, avoiding the drug to reach lethal concentrations, until a stable, more efficient alteration occurs, that allows survival in the presence of that agent. In this paper we review the current knowledge on MDR efflux pumps and their intricate regulatory network in Staphylococcus aureus, a major pathogen, responsible from mild to life-threatening infections. Particular emphasis will be given to the potential role that S. aureus MDR efflux pumps, either chromosomal or plasmid-encoded, have on resistance towards different antimicrobial agents and on the selection of drug - resistant strains. We will also discuss the many questions that still remain on the role of each specific efflux pump and the need to establish appropriate methodological approaches to address all these questions.
Collapse
Affiliation(s)
- Sofia Santos Costa
- 1Grupo de Micobactérias, Unidade de Microbiologia Médica, Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa (IHMT, UNL), Portugal
- 2Centro de Recursos Microbiológicos (CREM), UNL, Portugal
| | - Miguel Viveiros
- 1Grupo de Micobactérias, Unidade de Microbiologia Médica, Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa (IHMT, UNL), Portugal
- 3COST ACTION BM0701 (ATENS), Brussels, Belgium
| | - Leonard Amaral
- 1Grupo de Micobactérias, Unidade de Microbiologia Médica, Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa (IHMT, UNL), Portugal
- 3COST ACTION BM0701 (ATENS), Brussels, Belgium
| | - Isabel Couto
- 1Grupo de Micobactérias, Unidade de Microbiologia Médica, Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa (IHMT, UNL), Portugal
- 2Centro de Recursos Microbiológicos (CREM), UNL, Portugal
| |
Collapse
|
47
|
Adaptive and mutational resistance: role of porins and efflux pumps in drug resistance. Clin Microbiol Rev 2013; 25:661-81. [PMID: 23034325 DOI: 10.1128/cmr.00043-12] [Citation(s) in RCA: 576] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The substantial use of antibiotics in the clinic, combined with a dearth of new antibiotic classes, has led to a gradual increase in the resistance of bacterial pathogens to these compounds. Among the various mechanisms by which bacteria endure the action of antibiotics, those affecting influx and efflux are of particular importance, as they limit the interaction of the drug with its intracellular targets and, consequently, its deleterious effects on the cell. This review evaluates the impact of porins and efflux pumps on two major types of resistance, namely, mutational and adaptive types of resistance, both of which are regarded as key phenomena in the global rise of antibiotic resistance among pathogenic microorganisms. In particular, we explain how adaptive and mutational events can dramatically influence the outcome of antibiotic therapy by altering the mechanisms of influx and efflux of antibiotics. The identification of porins and pumps as major resistance markers has opened new possibilities for the development of novel therapeutic strategies directed specifically against these mechanisms.
Collapse
|
48
|
Qin R, Xiao K, Li B, Jiang W, Peng W, Zheng J, Zhou H. The combination of catechin and epicatechin callate from Fructus Crataegi potentiates beta-lactam antibiotics against methicillin-resistant staphylococcus aureus (MRSA) in vitro and in vivo. Int J Mol Sci 2013; 14:1802-21. [PMID: 23325048 PMCID: PMC3565349 DOI: 10.3390/ijms14011802] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Revised: 11/29/2012] [Accepted: 01/08/2013] [Indexed: 11/17/2022] Open
Abstract
Fructus crataegi (hawthorn) is the common name of all plant species in the genus Crataegus of the Rosaceae family. In the present study, three monomers of (+)-catechin (C), (-)-epicatechin gallate (ECg) and (-)-epigallocatechin (EGC) were isolated from the hawthorn under the guide of antibacterial sensitization activity. The bioactivity of the composite fraction in enhancing the antibacterial effect of oxacillin against methicillin-resistant Staphylococcus aureus (MRSA) was greater than that of the individual monomer of the hawthorn extract in vitro. Two-fold dilution and checkerboard methods were used to analyze antibacterial activity and screen for the combination and proportion of monomers with the best bioactivity. The result showed that C (128 mg/L) combined with ECg (16 mg/L) had the greatest effect and the combination also reduced the bacterial load in blood of septic mice challenged with a sublethal dose of MRSA, increased daunomycin accumulation within MRSA and down-regulated the mRNA expression of norA, norC and abcA, three important efflux pumps of MRSA. In summary, C and ECg enhanced the antibacterial effect of β-lactam antibiotics against MRSA in vitro and in vivo, which might be related to the increased accumulation of antibiotics within MRSA via suppression of important efflux pumps' gene expression.
Collapse
Affiliation(s)
- Rongxin Qin
- Department of Pharmacology, College of Pharmacy, The Third Military Medical University, Chongqing 400038, China; E-Mails: (R.Q.); (K.X.); (B.L.); (W.J.); (W.P.)
| | - Kangkang Xiao
- Department of Pharmacology, College of Pharmacy, The Third Military Medical University, Chongqing 400038, China; E-Mails: (R.Q.); (K.X.); (B.L.); (W.J.); (W.P.)
| | - Bin Li
- Department of Pharmacology, College of Pharmacy, The Third Military Medical University, Chongqing 400038, China; E-Mails: (R.Q.); (K.X.); (B.L.); (W.J.); (W.P.)
| | - Weiwei Jiang
- Department of Pharmacology, College of Pharmacy, The Third Military Medical University, Chongqing 400038, China; E-Mails: (R.Q.); (K.X.); (B.L.); (W.J.); (W.P.)
| | - Wei Peng
- Department of Pharmacology, College of Pharmacy, The Third Military Medical University, Chongqing 400038, China; E-Mails: (R.Q.); (K.X.); (B.L.); (W.J.); (W.P.)
| | - Jiang Zheng
- Medical Research Center, Southwestern Hospital, The Third Military Medical University, Chongqing 400038, China
| | - Hong Zhou
- Department of Pharmacology, College of Pharmacy, The Third Military Medical University, Chongqing 400038, China; E-Mails: (R.Q.); (K.X.); (B.L.); (W.J.); (W.P.)
| |
Collapse
|
49
|
Staphylococcus aureus NorD, a putative efflux pump coregulated with the Opp1 oligopeptide permease, contributes selectively to fitness in vivo. J Bacteriol 2012; 194:6586-93. [PMID: 23042988 DOI: 10.1128/jb.01414-12] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Staphylococcus aureus readily infects humans, causing infections from mild superficial skin infections to lethal bacteremia and endocarditis. Transporters produced by S. aureus allow the pathogen to adapt to a variety of settings, including survival at sites of infection and in the presence of antibiotics. The native functions of many transporters are unknown, but their potential dual contribution to fitness and antimicrobial resistance highlights their importance in staphylococcal infections. Here, we show that S. aureus NorD, a newly recognized efflux pump of the major facilitator superfamily, contributes to fitness in a murine subcutaneous abscess model. In community-associated methicillin-resistant S. aureus (CA-MRSA) strain MW2, norD was selectively upregulated 36-fold at the infection site relative to growth in vitro, and the norD mutant demonstrated significant fitness impairment in abscesses, with fitness 20- to 40-fold lower than that of the parent MW2 strain. Plasmid-encoded NorD could complement the fitness defect of the MW2 norD mutant. Chromosomal norD expression is polycistronic with the upstream oligopeptide permease genes (opp1ABCDF), which encode an ABC oligopeptide transporter. Both norD and opp1 were upregulated in abscesses and iron-restricted culture medium and negatively regulated by Fur, but only NorD contributed to fitness in the murine abscess model.
Collapse
|
50
|
Lebreton F, van Schaik W, Sanguinetti M, Posteraro B, Torelli R, Le Bras F, Verneuil N, Zhang X, Giard JC, Dhalluin A, Willems RJL, Leclercq R, Cattoir V. AsrR is an oxidative stress sensing regulator modulating Enterococcus faecium opportunistic traits, antimicrobial resistance, and pathogenicity. PLoS Pathog 2012; 8:e1002834. [PMID: 22876178 PMCID: PMC3410868 DOI: 10.1371/journal.ppat.1002834] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Accepted: 06/18/2012] [Indexed: 12/20/2022] Open
Abstract
Oxidative stress serves as an important host/environmental signal that triggers a wide range of responses in microorganisms. Here, we identified an oxidative stress sensor and response regulator in the important multidrug-resistant nosocomial pathogen Enterococcus faecium belonging to the MarR family and called AsrR (antibiotic and stress response regulator). The AsrR regulator used cysteine oxidation to sense the hydrogen peroxide which results in its dissociation to promoter DNA. Transcriptome analysis showed that the AsrR regulon was composed of 181 genes, including representing functionally diverse groups involved in pathogenesis, antibiotic and antimicrobial peptide resistance, oxidative stress, and adaptive responses. Consistent with the upregulated expression of the pbp5 gene, encoding a low-affinity penicillin-binding protein, the asrR null mutant was found to be more resistant to β-lactam antibiotics. Deletion of asrR markedly decreased the bactericidal activity of ampicillin and vancomycin, which are both commonly used to treat infections due to enterococci, and also led to over-expression of two major adhesins, acm and ecbA, which resulted in enhanced in vitro adhesion to human intestinal cells. Additional pathogenic traits were also reinforced in the asrR null mutant including greater capacity than the parental strain to form biofilm in vitro and greater persistance in Galleria mellonella colonization and mouse systemic infection models. Despite overexpression of oxidative stress-response genes, deletion of asrR was associated with a decreased oxidative stress resistance in vitro, which correlated with a reduced resistance to phagocytic killing by murine macrophages. Interestingly, both strains showed similar amounts of intracellular reactive oxygen species. Finally, we observed a mutator phenotype and enhanced DNA transfer frequencies in the asrR deleted strain. These data indicate that AsrR plays a major role in antimicrobial resistance and adaptation for survival within the host, thereby contributes importantly to the opportunistic traits of E. faecium. Multiple antibiotic-resistant isolates of the opportunistic pathogen Enterococcus faecium have emerged and spread worldwide. However, studies aimed at identifying mechanisms that underlie the transformation of E. faecium from its commensal nature into a nosocomial pathogen are scarce. We report pleiotropic roles for a novel oxidative-sensing regulator, called AsrR (antibiotic and stress response regulator), in E. faecium. Based on transcriptomic analysis, phenotypic studies, and animal models, we demonstrate that asrR deletion is responsible for i) diminished susceptibility to penicillins, vancomycin, and cationic antimicrobial peptides, ii) increased adhesion to human cells and biofilm formation, iii) a mutator phenotype and enhanced DNA transfer frequencies, iv) decreased resistance to oxidative stress both in vitro and in murine macrophages, and v) increased host-persistence in both insect and mouse models. AsrR is a stress-sensor and is promptly inactivated in the presence of hydrogen peroxide. Therefore, oxidative stress, which is a main challenge during infection, may be a significant signal used by E. faecium to promote opportunistic traits. This provides a significant resource combining, for the first time in E. faecium, a global transcriptomic approach and a thorough phenotypic study, which places AsrR as a key regulator modulating pathogenicity, antimicrobial resistance, and environmental adaptation.
Collapse
Affiliation(s)
- François Lebreton
- University of Caen Basse-Normandie, EA4655 (team “Antibioresistance”), Medical School, Caen, France
| | - Willem van Schaik
- University Medical Center Utrecht, Department of Medical Microbiology, Utrecht, The Netherlands
| | | | | | - Riccardo Torelli
- Catholic University of Sacred Heart, Institute of Microbiology, Rome, Italy
| | - Florian Le Bras
- University of Caen Basse-Normandie, EA4655 (team “Antibioresistance”), Medical School, Caen, France
| | - Nicolas Verneuil
- University of Caen Basse-Normandie, EA4655 (team “Stress and Virulence”), Caen, France
| | - Xinglin Zhang
- University Medical Center Utrecht, Department of Medical Microbiology, Utrecht, The Netherlands
| | - Jean-Christophe Giard
- University of Caen Basse-Normandie, EA4655 (team “Antibioresistance”), Medical School, Caen, France
| | - Anne Dhalluin
- University of Caen Basse-Normandie, EA4655 (team “Antibioresistance”), Medical School, Caen, France
| | - Rob J. L. Willems
- University Medical Center Utrecht, Department of Medical Microbiology, Utrecht, The Netherlands
| | - Roland Leclercq
- University of Caen Basse-Normandie, EA4655 (team “Antibioresistance”), Medical School, Caen, France
- University Hospital of Caen, Department of Microbiology, Caen, France
| | - Vincent Cattoir
- University of Caen Basse-Normandie, EA4655 (team “Antibioresistance”), Medical School, Caen, France
- University Hospital of Caen, Department of Microbiology, Caen, France
- * E-mail:
| |
Collapse
|