1
|
Qu T, Wang P, Zhao X, Liang L, Ji Q, Ge Y, Chen Y. Metagenomic profiles of the antimicrobial resistance in traditional Chinese fermented meat products: Core resistome and co-occurrence patterns. Int J Food Microbiol 2024; 418:110740. [PMID: 38754174 DOI: 10.1016/j.ijfoodmicro.2024.110740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 05/04/2024] [Accepted: 05/07/2024] [Indexed: 05/18/2024]
Abstract
Antimicrobial resistance (AMR) poses a significant challenge to global health, and the presence of antibiotic resistance genes (ARGs) in food poses a potential threat to public health. Traditional Chinese fermented meat products (FMPs) are highly favored because of their unique flavors and cultural value. However, microbial safety and the potential distribution and composition of AMR in these products remain unclear. In this study, a comprehensive analysis of bacterial composition and antibiotic-resistant populations in 216 samples of traditional fermented meat products from different regions of China was conducted using a metagenomic approach. Staphylococcus was the most abundant genus in the samples, accounting for an average abundance of 29.9 %, followed by Tetragenococcus (17.1 %), and Latilactobacillus (3.6 %). A core resistome of FMP samples was constructed for the first time using co-occurrence network analysis, which revealed the distribution and interrelationships of ARGs and bio/metal-resistant genes (BMRGs). Random forest analysis identified the lincosamide nucleotidyltransferase lnuA and the multidrug and toxic compound extrusion (MATE) transporter abeM as potential indicators for assessing the overall abundance of the core resistome. Additionally, Staphylococcus, Acinetobacter, and Pseudomonas were identified as hosts constituting the core resistome. Despite their low abundance, the latter two still serve as major reservoirs of antibiotic resistance genes. Notably, Lactococcus cremoris was identified as the key host for tetracycline resistance genes in the samples, highlighting the need for enhanced resistance monitoring in lactic acid bacteria. Based on our findings, in the microbial safety assessment of fermented meat products, beyond common foodborne pathogens, attention should be focused on detecting and controlling coagulase-negative Staphylococcus, Acinetobacter, and Pseudomonas, and addressing bacterial resistance. The quantitative detection of lnuA and abeM could provide a convenient and rapid method for assessing the overall abundance of the core resistome. Our findings have important implications for the control of bacterial resistance and prevention of pathogenic bacteria in fermented meat products.
Collapse
Affiliation(s)
- Tianming Qu
- Chinese Academy of Inspection and Quarantine, Beijing 100176, China; College of Food Science and Engineering, Jilin Agricultural University, Changchun, 130118, Jilin, China
| | - Ping Wang
- Chinese Academy of Inspection and Quarantine, Beijing 100176, China
| | - Xiaomei Zhao
- Chinese Academy of Inspection and Quarantine, Beijing 100176, China
| | - Lijiao Liang
- Chinese Academy of Inspection and Quarantine, Beijing 100176, China; College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, China
| | - Qinglong Ji
- Chinese Academy of Inspection and Quarantine, Beijing 100176, China
| | - Yiqiang Ge
- China Rural Technology Development Center, Beijing 100045, China; College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Ying Chen
- Chinese Academy of Inspection and Quarantine, Beijing 100176, China.
| |
Collapse
|
2
|
Kerek Á, Török B, Laczkó L, Somogyi Z, Kardos G, Bányai K, Kaszab E, Bali K, Jerzsele Á. In Vitro Microevolution and Co-Selection Assessment of Amoxicillin and Cefotaxime Impact on Escherichia coli Resistance Development. Antibiotics (Basel) 2024; 13:247. [PMID: 38534682 DOI: 10.3390/antibiotics13030247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 03/01/2024] [Accepted: 03/05/2024] [Indexed: 03/28/2024] Open
Abstract
The global spread of antimicrobial resistance has become a prominent issue in both veterinary and public health in the 21st century. The extensive use of amoxicillin, a beta-lactam antibiotic, and consequent resistance development are particularly alarming in food-producing animals, with a focus on the swine and poultry sectors. Another beta-lactam, cefotaxime, is widely utilized in human medicine, where the escalating resistance to third- and fourth-generation cephalosporins is a major concern. The aim of this study was to simulate the development of phenotypic and genotypic resistance to beta-lactam antibiotics, focusing on amoxicillin and cefotaxime. The investigation of the minimal inhibitory concentrations (MIC) of antibiotics was performed at 1×, 10×, 100×, and 1000× concentrations using the modified microbial evolution and growth arena (MEGA-plate) method. Our results indicate that amoxicillin significantly increased the MIC values of several tested antibiotics, except for oxytetracycline and florfenicol. In the case of cefotaxime, this increase was observed in all classes. A total of 44 antimicrobial resistance genes were identified in all samples. Chromosomal point mutations, particularly concerning cefotaxime, revealed numerous complex mutations, deletions, insertions, and single nucleotide polymorphisms (SNPs) that were not experienced in the case of amoxicillin. The findings suggest that, regarding amoxicillin, the point mutation of the acrB gene could explain the observed MIC value increases due to the heightened activity of the acrAB-tolC efflux pump system. However, under the influence of cefotaxime, more intricate processes occurred, including complex amino acid substitutions in the ampC gene promoter region, increased enzyme production induced by amino acid substitutions and SNPs, as well as mutations in the acrR and robA repressor genes that heightened the activity of the acrAB-tolC efflux pump system. These changes may contribute to the significant MIC increases observed for all tested antibiotics. The results underscore the importance of understanding cross-resistance development between individual drugs when choosing clinical alternative drugs. The point mutations in the mdtB and emrR genes may also contribute to the increased activity of the mdtABC-tolC and emrAB-tolC pump systems against all tested antibiotics. The exceptionally high mutation rate induced by cephalosporins justifies further investigations to clarify the exact mechanism behind.
Collapse
Affiliation(s)
- Ádám Kerek
- Department of Pharmacology and Toxicology, University of Veterinary Medicine Budapest, H-1078 Budapest, Hungary
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, H-1078 Budapest, Hungary
| | - Bence Török
- Department of Pharmacology and Toxicology, University of Veterinary Medicine Budapest, H-1078 Budapest, Hungary
| | - Levente Laczkó
- One Health Institute, University of Debrecen, Nagyerdei krt. 98, H-4032 Debrecen, Hungary
- HUN-REN-UD Conservation Biology Research Group, Egyetem tér 1, H-4032 Debrecen, Hungary
| | - Zoltán Somogyi
- Department of Pharmacology and Toxicology, University of Veterinary Medicine Budapest, H-1078 Budapest, Hungary
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, H-1078 Budapest, Hungary
| | - Gábor Kardos
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, H-1078 Budapest, Hungary
- One Health Institute, University of Debrecen, Nagyerdei krt. 98, H-4032 Debrecen, Hungary
- National Public Health Center, Albert Flórián út 2-6, H-1097 Budapest, Hungary
- Department of Gerontology, Faculty of Health Sciences, University of Debrecen, Sóstói út 2-4, H-4400 Nyíregyháza, Hungary
| | - Krisztián Bányai
- Department of Pharmacology and Toxicology, University of Veterinary Medicine Budapest, H-1078 Budapest, Hungary
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, H-1078 Budapest, Hungary
- Veterinary Medical Research Institute, H-1143 Budapest, Hungary
| | - Eszter Kaszab
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, H-1078 Budapest, Hungary
- One Health Institute, University of Debrecen, Nagyerdei krt. 98, H-4032 Debrecen, Hungary
- Department of Microbiology and Infectious Diseases, University of Veterinary Medicine, István u 2, H-1078 Budapest, Hungary
| | - Krisztina Bali
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, H-1078 Budapest, Hungary
- Department of Microbiology and Infectious Diseases, University of Veterinary Medicine, István u 2, H-1078 Budapest, Hungary
| | - Ákos Jerzsele
- Department of Pharmacology and Toxicology, University of Veterinary Medicine Budapest, H-1078 Budapest, Hungary
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, H-1078 Budapest, Hungary
| |
Collapse
|
3
|
Turbant F, Esnouf E, Rosaz F, Wien F, Węgrzyn G, Chauvet H, Arluison V. Role of the Bacterial Amyloid-like Hfq in Fluoroquinolone Fluxes. Microorganisms 2023; 12:53. [PMID: 38257880 PMCID: PMC10819720 DOI: 10.3390/microorganisms12010053] [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: 12/22/2023] [Accepted: 12/26/2023] [Indexed: 01/24/2024] Open
Abstract
Due to their two-cell membranes, Gram-negative bacteria are particularly resistant to antibiotics. Recent investigations aimed at exploring new target proteins involved in Gram-negative bacteria adaptation helped to identify environmental changes encountered during infection. One of the most promising approaches in finding novel targets for antibacterial drugs consists of blocking noncoding RNA-based regulation using the protein cofactor, Hfq. Although Hfq is important in many bacterial pathogens, its involvement in antibiotics response is still unclear. Indeed, Hfq may mediate drug resistance by regulating the major efflux system in Escherichia coli, but it could also play a role in the influx of antibiotics. Here, using an imaging approach, we addressed this problem quantitatively at the single-cell level. More precisely, we analyzed how Hfq affects the dynamic influx and efflux of ciprofloxacin, an antibiotic from the group of fluoroquinolones that is used to treat bacterial infections. Our results indicated that the absence of either whole Hfq or its C-terminal domain resulted in a more effective accumulation of ciprofloxacin, irrespective of the presence of the functional AcrAB-TolC efflux pump. However, overproduction of the MicF small regulatory RNA, which reduces the efficiency of expression of the ompF gene (coding for a porin involved in antibiotics influx) in a Hfq-dependent manner, resulted in impaired accumulation of ciprofloxacin. These results led us to propose potential mechanisms of action of Hfq in the regulation of fluoroquinolone fluxes across the E. coli envelope.
Collapse
Affiliation(s)
- Florian Turbant
- Synchrotron SOLEIL, L’Orme des Merisiers, Saint Aubin BP48, 91192 Gif-sur-Yvette, France; (F.T.); (F.W.); (H.C.)
- Laboratoire Léon Brillouin LLB, UMR12 CEA CNRS, CEA Saclay, 91191 Gif-sur-Yvette, France; (E.E.); (F.R.)
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland;
| | - Emeline Esnouf
- Laboratoire Léon Brillouin LLB, UMR12 CEA CNRS, CEA Saclay, 91191 Gif-sur-Yvette, France; (E.E.); (F.R.)
| | - Francois Rosaz
- Laboratoire Léon Brillouin LLB, UMR12 CEA CNRS, CEA Saclay, 91191 Gif-sur-Yvette, France; (E.E.); (F.R.)
| | - Frank Wien
- Synchrotron SOLEIL, L’Orme des Merisiers, Saint Aubin BP48, 91192 Gif-sur-Yvette, France; (F.T.); (F.W.); (H.C.)
| | - Grzegorz Węgrzyn
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland;
| | - Hugo Chauvet
- Synchrotron SOLEIL, L’Orme des Merisiers, Saint Aubin BP48, 91192 Gif-sur-Yvette, France; (F.T.); (F.W.); (H.C.)
| | - Véronique Arluison
- Laboratoire Léon Brillouin LLB, UMR12 CEA CNRS, CEA Saclay, 91191 Gif-sur-Yvette, France; (E.E.); (F.R.)
- UFR SDV, Université Paris Cité, 75013 Paris, France
| |
Collapse
|
4
|
Kerek Á, Török B, Laczkó L, Kardos G, Bányai K, Somogyi Z, Kaszab E, Bali K, Jerzsele Á. In Vitro Microevolution and Co-Selection Assessment of Florfenicol Impact on Escherichia coli Resistance Development. Antibiotics (Basel) 2023; 12:1728. [PMID: 38136762 PMCID: PMC10740648 DOI: 10.3390/antibiotics12121728] [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: 11/13/2023] [Revised: 12/08/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023] Open
Abstract
The issue of antimicrobial resistance is becoming an increasingly serious challenge in both human and veterinary medicine. Prudent antimicrobial use in veterinary medicine is warranted and supported by international guidelines, with the Antimicrobial Advice Ad Hoc Expert Group (AMEG) placing particular emphasis on the critically important group B antimicrobials. These antimicrobials are commonly employed, especially in the poultry and swine industry. The impact of florfenicol, a veterinary antibiotic, was studied on the resistance development of Escherichia coli. The aim of the study was to investigate the effect of the use of florfenicol on the development of phenotypic and genomic resistances, not only to the drug itself but also to other drugs. The minimum inhibitory concentrations (MICs) of the antibiotics were investigated at 1×, 10×, 100× and 1000× concentrations using the adapted Microbial Evolution and Growth Arena (MEGA-plate) method. The results demonstrate that florfenicol can select for resistance to fluoroquinolone antibiotics (167× MIC value increase) and cephalosporins (67× MIC value increase). A total of 44 antimicrobial resistance genes were identified, the majority of which were consistent across the samples. Chromosomal point mutations, including alterations in resistance-associated and regulatory genes (acrB, acrR, emrR and robA), are thought to trigger multiple drug efflux pump activations, leading to phenotypically increased resistance. The study underscores the impact of florfenicol and its role in the development of antimicrobial resistance, particularly concerning fluoroquinolone antibiotics and cephalosporins. This study is the first to report florfenicol's dose-dependent enhancement of other antibiotics' MICs, linked to mutations in SOS-box genes (mdtABC-tolC, emrAB-tolC and acrAB-tolC) and increased multidrug efflux pump genes. Mutations in the regulatory genes acrR, emrR and rpbA support the possibility of increased gene expression. The results are crucial for understanding antimicrobial resistance and its development, highlighting the promising potential of in vitro evolutionary and coselection studies for future research.
Collapse
Affiliation(s)
- Ádám Kerek
- Department of Pharmacology and Toxicology, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary; (B.T.); (K.B.); (Z.S.); (Á.J.)
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary; (G.K.); (E.K.)
| | - Bence Török
- Department of Pharmacology and Toxicology, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary; (B.T.); (K.B.); (Z.S.); (Á.J.)
| | - Levente Laczkó
- Institute of Metagenomics, University of Debrecen, 4032 Debrecen, Hungary;
| | - Gábor Kardos
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary; (G.K.); (E.K.)
- Institute of Metagenomics, University of Debrecen, 4032 Debrecen, Hungary;
| | - Krisztián Bányai
- Department of Pharmacology and Toxicology, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary; (B.T.); (K.B.); (Z.S.); (Á.J.)
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary; (G.K.); (E.K.)
- Veterinary Medical Research Institute, 1143 Budapest, Hungary;
| | - Zoltán Somogyi
- Department of Pharmacology and Toxicology, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary; (B.T.); (K.B.); (Z.S.); (Á.J.)
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary; (G.K.); (E.K.)
| | - Eszter Kaszab
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary; (G.K.); (E.K.)
- Veterinary Medical Research Institute, 1143 Budapest, Hungary;
| | - Krisztina Bali
- Veterinary Medical Research Institute, 1143 Budapest, Hungary;
| | - Ákos Jerzsele
- Department of Pharmacology and Toxicology, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary; (B.T.); (K.B.); (Z.S.); (Á.J.)
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary; (G.K.); (E.K.)
| |
Collapse
|
5
|
Lu H, Li P, Chen L, Chen G. Flectobacillus longus sp. nov. and Flectobacillus rivi sp. nov., isolated from streams in China and reclassification of Flectobacillus rhizosphaerae as a synonym of Flectobacillus roseus. Int J Syst Evol Microbiol 2023; 73. [PMID: 37917534 DOI: 10.1099/ijsem.0.006148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2023] Open
Abstract
Three Gram-stain-negative, aerobic, rod-shaped, non-motile strains (LYT7WT, DC10W and LFS242WT) were isolated from streams in PR China. Comparisons based on the 16S rRNA gene sequences showed that these three strains showed high 16S rRNA gene sequence similarity to Flectobacillus roseus GFA-11T (99.2, 98.8 and 99.8 %, respectively) and Flectobacillus rhizosphaerae JC289T (98.8, 98.6 and 99.4 %, respectively), and less than 96.6 % to other species of the genus Flectobacillus. The phylogenetic tree reconstructed based on the 16S rRNA gene sequences showed that strains LYT7WT, DC10W, LFS242WT, F. roseus GFA-11T and F. rhizosphaerae JC289T formed a tight cluster. The phylogenomic tree also supported the above robust phylogenetic relationships. The calculated OrthoANIu and digital DNA-DNA hybridization values between strains LYT7WT and DC10W were 95.2 % and 61.9 %, respectively. Although these values are located in the transition region for species demarcation, the similar physiological and genotypic characteristics supported that strains LYT7WT and DC10W should belong to the same species. The pairwise OrthoANIu and digital DNA-DNA hybridization values between strain LFS242WT and its related strains were less than 91.8 and 45.3 %, respectively, indicating that strain LFS242WT should represent an independent novel species of the genus Flectobacillus. It should be noticed that the pairwise OrthoANIu and digital DNA-DNA hybridization values between strains F. roseus LMG 24501T and F. rhizosphaerae KCTC 42575T were 96.9 and 73.1 %, respectively. The similar physiological and genotypic characteristics also supported that F. rhizosphaerae Ramaprasad et al. 2015 should be a synonym of F. roseus Sheu et al. 2009. Combining the above descriptions, strains LYT7WT, DC10W and LFS242WT should represent two novel species of the genus Flectobacillus, for which the names Flectobacillus longus sp. nov. (type strain LYT7WT=GDMCC 1.3222T=KCTC 92561T) and Flectobacillus rivi sp. nov. (type strain LFS242WT=GDMCC 1.3223T=KCTC 92562T) are proposed.
Collapse
Affiliation(s)
- Huibin Lu
- Yunnan Key Laboratory of Plateau Geographical Process and Environmental Changes, Faculty of Geography, Yunnan Normal University, Kunming 650500, PR China
| | - Ping Li
- College of Environment and Resource, Xichang University, Xichang 615000, PR China
| | - Li Chen
- Yunnan Key Laboratory of Plateau Geographical Process and Environmental Changes, Faculty of Geography, Yunnan Normal University, Kunming 650500, PR China
| | - Guangjie Chen
- Yunnan Key Laboratory of Plateau Geographical Process and Environmental Changes, Faculty of Geography, Yunnan Normal University, Kunming 650500, PR China
| |
Collapse
|
6
|
Islam MD, Harrison BD, Li JJY, McLoughlin AG, Court DA. Do mitochondria use efflux pumps to protect their ribosomes from antibiotics? MICROBIOLOGY (READING, ENGLAND) 2023; 169:001272. [PMID: 36748523 PMCID: PMC9993110 DOI: 10.1099/mic.0.001272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Fungal environments are rich in natural and engineered antimicrobials, and this, combined with the fact that fungal genomes are rich in coding sequences for transporters, suggests that fungi are an intriguing group in which to search for evidence of antimicrobial efflux pumps in mitochondria. Herein, the range of protective mechanisms used by fungi against antimicrobials is introduced, and it is hypothesized, based on the susceptibility of mitochondrial and bacterial ribosomes to the same antibiotics, that mitochondria might also contain pumps that efflux antibiotics from these organelles. Preliminary evidence of ethidium bromide efflux is presented and several candidate efflux pumps are identified in fungal mitochondrial proteomes.
Collapse
Affiliation(s)
- Md Deen Islam
- Department of Microbiology, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada
| | - Brian D Harrison
- Department of Microbiology, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada
| | - Judy J-Y Li
- Department of Microbiology, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada
| | - Austein G McLoughlin
- Department of Microbiology, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada
| | | |
Collapse
|
7
|
Kopf A, Bunk B, Coldewey SM, Gunzer F, Riedel T, Schröttner P. Comparative Genomic Analysis of the Human Pathogen Wohlfahrtiimonas Chitiniclastica Provides Insight Into the Identification of Antimicrobial Resistance Genotypes and Potential Virulence Traits. Front Cell Infect Microbiol 2022; 12:912427. [PMID: 35873140 PMCID: PMC9301364 DOI: 10.3389/fcimb.2022.912427] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 06/02/2022] [Indexed: 11/20/2022] Open
Abstract
Recent studies suggest that Wohlfahrtiimonas chitiniclastica may be the cause of several diseases in humans including sepsis and bacteremia making the bacterium as a previously underappreciated human pathogen. However, very little is known about the pathogenicity and genetic potential of W. chitiniclastica; therefore, it is necessary to conduct systematic studies to gain a deeper understanding of its virulence characteristics and treatment options. In this study, the entire genetic repertoire of all publicly available W. chitiniclastica genomes was examined including in silico characterization of bacteriophage content, antibiotic resistome, and putative virulence profile. The pan-genome of W. chitiniclastica comprises 3819 genes with 1622 core genes (43%) indicating a putative metabolic conserved species. Furthermore, in silico analysis indicated presumed resistome expansion as defined by the presence of genome-encoded transposons and bacteriophages. While macrolide resistance genes macA and macB are located within the core genome, additional antimicrobial resistance genotypes for tetracycline (tetH, tetB, and tetD), aminoglycosides (ant(2'')-Ia, aac(6')-Ia,aph(3'')-Ib, aph(3')-Ia, and aph(6)-Id)), sulfonamide (sul2), streptomycin (strA), chloramphenicol (cat3), and beta-lactamase (blaVEB) are distributed among the accessory genome. Notably, our data indicate that the type strain DSM 18708T does not encode any additional clinically relevant antibiotic resistance genes, whereas drug resistance is increasing within the W. chitiniclastica clade. This trend should be monitored with caution. To the best of our knowledge, this is the first comprehensive genome analysis of this species, providing new insights into the genome of this opportunistic human pathogen.
Collapse
Affiliation(s)
- Anna Kopf
- Medical Microbiology and Virology, University Hospital Carl Gustav Carus, Dresden, Germany
- Clinic for Hematology and Oncology, Carl-Thiem-Klinikum, Cottbus, Germany
| | - Boyke Bunk
- German Collection of Microorganisms and Cell Cultures GmbH, Leibniz Institute DSMZ, Braunschweig, Germany
| | - Sina M. Coldewey
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany
- Septomics Research Center, Jena University Hospital, Jena, Germany
| | - Florian Gunzer
- Department of Hospital Infection Control, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Thomas Riedel
- German Collection of Microorganisms and Cell Cultures GmbH, Leibniz Institute DSMZ, Braunschweig, Germany
- German Center for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Braunschweig, Germany
| | - Percy Schröttner
- Medical Microbiology and Virology, University Hospital Carl Gustav Carus, Dresden, Germany
| |
Collapse
|
8
|
Inactivation of a New Potassium Channel Increases Rifampicin Resistance and Induces Collateral Sensitivity to Hydrophilic Antibiotics in Mycobacterium smegmatis. Antibiotics (Basel) 2022; 11:antibiotics11040509. [PMID: 35453260 PMCID: PMC9025972 DOI: 10.3390/antibiotics11040509] [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: 02/28/2022] [Revised: 04/05/2022] [Accepted: 04/08/2022] [Indexed: 12/10/2022] Open
Abstract
Rifampicin is a critical first-line antibiotic for treating mycobacterial infections such as tuberculosis, one of the most serious infectious diseases worldwide. Rifampicin resistance in mycobacteria is mainly caused by mutations in the rpoB gene; however, some rifampicin-resistant strains showed no rpoB mutations. Therefore, alternative mechanisms must explain this resistance in mycobacteria. In this work, a library of 11,000 Mycobacterium smegmatis mc2 155 insertion mutants was explored to search and characterize new rifampicin-resistance determinants. A transposon insertion in the MSMEG_1945 gene modified the growth rate, pH homeostasis and membrane potential in M. smegmatis, producing rifampicin resistance and collateral susceptibility to other antitubercular drugs such as isoniazid, ethionamide and aminoglycosides. Our data suggest that the M. smegmatis MSMEG_1945 protein is an ion channel, dubbed MchK, essential for maintaining the cellular ionic balance and membrane potential, modulating susceptibility to antimycobacterial agents. The functions of this new gene point once again to potassium homeostasis impairment as a proxy to resistance to rifampicin. This study increases the known repertoire of mycobacterial ion channels involved in drug susceptibility/resistance to antimycobacterial drugs and suggests novel intervention opportunities, highlighting ion channels as druggable pathways.
Collapse
|
9
|
Kaur H, Kalia M, Singh V, Modgil V, Mohan B, Taneja N. In silico identification and characterization of promising drug targets in highly virulent uropathogenic Escherichia coli strain CFT073 by protein-protein interaction network analysis. INFORMATICS IN MEDICINE UNLOCKED 2021. [DOI: 10.1016/j.imu.2021.100704] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
|
10
|
The Great ESKAPE: Exploring the Crossroads of Bile and Antibiotic Resistance in Bacterial Pathogens. Infect Immun 2020; 88:IAI.00865-19. [PMID: 32661122 DOI: 10.1128/iai.00865-19] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Throughout the course of infection, many pathogens encounter bactericidal conditions that threaten the viability of the bacteria and impede the establishment of infection. Bile is one of the most innately bactericidal compounds present in humans, functioning to reduce the bacterial burden in the gastrointestinal tract while also aiding in digestion. It is becoming increasingly apparent that pathogens successfully resist the bactericidal conditions of bile, including bacteria that do not normally cause gastrointestinal infections. This review highlights the ability of Enterococcus, Staphylococcus, Klebsiella, Acinetobacter, Pseudomonas, Enterobacter (ESKAPE), and other enteric pathogens to resist bile and how these interactions can impact the sensitivity of bacteria to various antimicrobial agents. Given that pathogen exposure to bile is an essential component to gastrointestinal transit that cannot be avoided, understanding how bile resistance mechanisms align with antimicrobial resistance is vital to our ability to develop new, successful therapeutics in an age of widespread and increasing antimicrobial resistance.
Collapse
|
11
|
Wang Y, Alenzy R, Song D, Liu X, Teng Y, Mowla R, Ma Y, Polyak SW, Venter H, Ma S. Structural optimization of natural product nordihydroguaretic acid to discover novel analogues as AcrB inhibitors. Eur J Med Chem 2019; 186:111910. [PMID: 31801655 DOI: 10.1016/j.ejmech.2019.111910] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 11/12/2019] [Accepted: 11/24/2019] [Indexed: 12/27/2022]
Abstract
Drug efflux pumps confer multidrug resistance to dangerous bacterial pathogens which makes these proteins promising drug targets. Herein, we present initial chemical optimization and structure-activity relationship (SAR) data around a previously described efflux pump inhibitor, nordihydroguaretic acid (NDGA). Four series of novel NDGA analogues that target Escherichia coli AcrB were designed, synthesized and evaluated for their ability to potentiate the activity of antibiotics, to inhibit AcrB-mediated substrate efflux and reduce off-target activity. Nine novel structures were identified that increased the efficacy of a panel of antibiotics, inhibited drug efflux and reduced permeabilization of the bacterial outer and inner membranes. Among them, WA7, WB11 and WD6 possessing broad-spectrum antimicrobial sensitization activity were identified as NDGA analogues with favorable properties as potential AcrB inhibitors, demonstrating moderate improvement in potency as compared to NDGA. In particular, WD6 was the most broadly active analogue improving the activity of all four classes of antibacterials tested.
Collapse
Affiliation(s)
- Yinhu Wang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 West Culture Road, Jinan, 250012, China; School of Pharmacy, Liaocheng University, Liaocheng, China
| | - Rawaf Alenzy
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, 5000, Australia; Department of Medical Laboratory, College of Applied Medical Sciences-Shaqra, Shaqra University, 11961, Saudi Arabia
| | - Di Song
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 West Culture Road, Jinan, 250012, China
| | - Xingbang Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 West Culture Road, Jinan, 250012, China
| | - Yuetai Teng
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 West Culture Road, Jinan, 250012, China
| | - Rumana Mowla
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, 5000, Australia
| | - Yingang Ma
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 West Culture Road, Jinan, 250012, China
| | - Steven W Polyak
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, 5000, Australia
| | - Henrietta Venter
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, 5000, Australia.
| | - Shutao Ma
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 West Culture Road, Jinan, 250012, China.
| |
Collapse
|
12
|
Casaburi G, Duar RM, Vance DP, Mitchell R, Contreras L, Frese SA, Smilowitz JT, Underwood MA. Early-life gut microbiome modulation reduces the abundance of antibiotic-resistant bacteria. Antimicrob Resist Infect Control 2019; 8:131. [PMID: 31423298 PMCID: PMC6693174 DOI: 10.1186/s13756-019-0583-6] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 07/22/2019] [Indexed: 01/21/2023] Open
Abstract
Background Antibiotic-resistant (AR) bacteria are a global threat. AR bacteria can be acquired in early life and have long-term sequelae. Limiting the spread of antibiotic resistance without triggering the development of additional resistance mechanisms is of immense clinical value. Here, we show how the infant gut microbiome can be modified, resulting in a significant reduction of AR genes (ARGs) and the potentially pathogenic bacteria that harbor them. Methods The gut microbiome was characterized using shotgun metagenomics of fecal samples from two groups of healthy, term breastfed infants. One group was fed B. infantis EVC001 in addition to receiving lactation support (n = 29, EVC001-fed), while the other received lactation support alone (n = 31, controls). Coliforms were isolated from fecal samples and genome sequenced, as well as tested for minimal inhibitory concentrations against clinically relevant antibiotics. Results Infants fed B. infantis EVC001 exhibited a change to the gut microbiome, resulting in a 90% lower level of ARGs compared to controls. ARGs that differed significantly between groups were predicted to confer resistance to beta lactams, fluoroquinolones, or multiple drug classes, the majority of which belonged to Escherichia, Clostridium, and Staphylococcus. Minimal inhibitory concentration assays confirmed the resistance phenotypes among isolates with these genes. Notably, we found extended-spectrum beta lactamases among healthy, vaginally delivered breastfed infants who had never been exposed to antibiotics. Conclusions Colonization of the gut of breastfed infants by a single strain of B. longum subsp. infantis had a profound impact on the fecal metagenome, including a reduction in ARGs. This highlights the importance of developing novel approaches to limit the spread of these genes among clinically relevant bacteria. Future studies are needed to determine whether colonization with B. infantis EVC001 decreases the incidence of AR infections in breastfed infants. Trial registration This clinical trial was registered at ClinicalTrials.gov, NCT02457338.
Collapse
Affiliation(s)
| | | | | | | | | | - Steven A. Frese
- Evolve Biosystems, Inc, Davis, CA 95618 USA
- Department of Food Science and Technology, University of Nebraska, Lincoln, NE 68588 USA
| | - Jennifer T. Smilowitz
- Department of Food Science and Technology, University of California, Davis, CA 95616 USA
- Foods for Health Institute, University of California, Davis, CA 95616 USA
| | - Mark A. Underwood
- Foods for Health Institute, University of California, Davis, CA 95616 USA
- Department of Pediatrics, UC Davis Children’s Hospital, Sacramento, CA 95817 USA
| |
Collapse
|
13
|
Iyer R, Moussa SH, Tommasi R, Miller AA. Titrating Levels of TolC in E. coli: A Sensitive Approach to Quantifying Efflux. ACS Infect Dis 2019; 5:49-54. [PMID: 30489063 DOI: 10.1021/acsinfecdis.8b00273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The susceptibility of small molecules to Gram-negative bacterial efflux is typically evaluated using an antibacterial activity-based efflux ratio, which is computed as the ratio of the antibacterial activity for a wild-type strain and its isogenic efflux mutant (typically lacking genes encoding major efflux pumps). The magnitude of the ratio is often used as an efflux index. However, early in drug discovery, hits with suboptimal physicochemical properties often lack whole cell inhibition against wild-type strains, which makes efflux ratios indeterminable. To address this gap, we developed an assay to titrate levels of total efflux by varying the TolC expression using an arabinose-inducible promoter (pBAD) in an Escherichia coli Δ tolC strain. We provide a proof of concept for the assay using sets of related compounds from two antibiotic classes and show that the TolC titration provides a sensitive method for rank ordering compounds with respect to their efflux susceptibility.
Collapse
Affiliation(s)
- Ramkumar Iyer
- Entasis Therapeutics Inc., 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Samir H. Moussa
- Entasis Therapeutics Inc., 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Ruben Tommasi
- Entasis Therapeutics Inc., 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Alita A. Miller
- Entasis Therapeutics Inc., 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| |
Collapse
|
14
|
Aron Z, Opperman TJ. The hydrophobic trap-the Achilles heel of RND efflux pumps. Res Microbiol 2017; 169:393-400. [PMID: 29146106 DOI: 10.1016/j.resmic.2017.11.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 10/17/2017] [Accepted: 11/01/2017] [Indexed: 01/05/2023]
Abstract
Resistance-nodulation-division (RND) superfamily efflux pumps play a major role in multidrug resistance (MDR) of Gram-negative pathogens by extruding diverse classes of antibiotics from the cell. There has been considerable interest in developing efflux pump inhibitors (EPIs) of RND pumps as adjunctive therapies. The primary challenge in EPI discovery has been the highly hydrophobic, poly-specific substrate binding site of the target. Recent findings have identified the hydrophobic trap, a narrow phenylalanine-lined groove in the substrate-binding site, as the "Achilles heel" of the RND efflux pumps. In this review, we will examine the hydrophobic trap as an EPI target and two chemically distinct series of EPIs that bind there.
Collapse
|
15
|
Praski Alzrigat L, Huseby DL, Brandis G, Hughes D. Fitness cost constrains the spectrum of marR mutations in ciprofloxacin-resistant Escherichia coli. J Antimicrob Chemother 2017; 72:3016-3024. [PMID: 28962020 PMCID: PMC5890708 DOI: 10.1093/jac/dkx270] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 06/12/2017] [Accepted: 07/07/2017] [Indexed: 12/26/2022] Open
Abstract
OBJECTIVES To determine whether the spectrum of mutations in marR in ciprofloxacin-resistant clinical isolates of Escherichia coli shows evidence of selection bias, either to reduce fitness costs, or to increase drug resistance. MarR is a repressor protein that regulates, via MarA, expression of the Mar regulon, including the multidrug efflux pump AcrAB-TolC. METHODS Isogenic strains carrying 36 different marR alleles identified in resistant clinical isolates, or selected for resistance in vitro, were constructed. Drug susceptibility and relative fitness in growth competition assays were measured for all strains. The expression level of marA, and of various efflux pump components, as a function of specific mutations in marR, was measured by qPCR. RESULTS The spectrum of genetic alterations in marR in clinical isolates is strongly biased against inactivating mutations. In general, the alleles found in clinical isolates conferred a lower level of resistance and imposed a lower growth fitness cost than mutations selected in vitro. The level of expression of MarA correlated well with the MIC of ciprofloxacin. This supports the functional connection between mutations in marR and reduced susceptibility to ciprofloxacin. CONCLUSIONS Mutations in marR selected in ciprofloxacin-resistant clinical isolates are strongly biased against inactivating mutations. Selection favours mutant alleles that have the lowest fitness costs, even though these cause only modest reductions in drug susceptibility. This suggests that selection for high relative fitness is more important than selection for increased resistance in determining which alleles of marR will be selected in resistant clinical isolates.
Collapse
Affiliation(s)
- Lisa Praski Alzrigat
- Department of Medical Biochemistry and Microbiology, Uppsala University, Biomedical Center Box 582, Husargatan 3, S-75123 Uppsala, Sweden
| | - Douglas L Huseby
- Department of Medical Biochemistry and Microbiology, Uppsala University, Biomedical Center Box 582, Husargatan 3, S-75123 Uppsala, Sweden
| | - Gerrit Brandis
- Department of Medical Biochemistry and Microbiology, Uppsala University, Biomedical Center Box 582, Husargatan 3, S-75123 Uppsala, Sweden
| | - Diarmaid Hughes
- Department of Medical Biochemistry and Microbiology, Uppsala University, Biomedical Center Box 582, Husargatan 3, S-75123 Uppsala, Sweden
| |
Collapse
|
16
|
Mowla R, Wang Y, Ma S, Venter H. Kinetic analysis of the inhibition of the drug efflux protein AcrB using surface plasmon resonance. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1860:878-886. [PMID: 28890187 DOI: 10.1016/j.bbamem.2017.08.024] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 08/19/2017] [Accepted: 08/31/2017] [Indexed: 11/24/2022]
Abstract
Multidrug efflux protein complexes such as AcrAB-TolC from Escherichia coli are paramount in multidrug resistance in Gram-negative bacteria and are also implicated in other processes such as virulence and biofilm formation. Hence efflux pump inhibition, as a means to reverse antimicrobial resistance in clinically relevant pathogens, has gained increased momentum over the past two decades. Significant advances in the structural and functional analysis of AcrB have informed the selection of efflux pump inhibitors (EPIs). However, an accurate method to determine the kinetics of efflux pump inhibition was lacking. In this study we standardised and optimised surface plasmon resonance (SPR) to probe the binding kinetics of substrates and inhibitors to AcrB. The SPR method was also combined with a fluorescence drug binding method by which affinity of two fluorescent AcrB substrates were determined using the same conditions and controls as for SPR. Comparison of the results from the fluorescent assay to those of the SPR assay showed excellent correlation and provided validation for the methods and conditions used for SPR. The kinetic parameters of substrate (doxorubicin, novobiocin and minocycline) binding to AcrB were subsequently determined. Lastly, the kinetics of inhibition of AcrB were probed for two established inhibitors (phenylalanine arginyl β-naphthylamide and 1-1-naphthylmethyl-piperazine) and three novel EPIs: 4-isobutoxy-2-naphthamide (A2), 4-isopentyloxy-2-naphthamide (A3) and 4-benzyloxy-2-naphthamide (A9) have also been probed. The kinetic data obtained could be correlated with inhibitor efficacy and mechanism of action. This study is the first step in the quantitative analysis of the kinetics of inhibition of the clinically important RND-class of multidrug efflux pumps and will allow the design of improved and more potent inhibitors of drug efflux pumps. This article is part of a Special Issue entitled: Beyond the Structure-Function Horizon of Membrane Proteins edited by Ute Hellmich, Rupak Doshi and Benjamin McIlwain.
Collapse
Affiliation(s)
- Rumana Mowla
- School of Pharmacy and Medical Sciences, Sansom Institute for Health Research, University of South Australia, SA 5000, Australia
| | - Yinhu Wang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Shutao Ma
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Henrietta Venter
- School of Pharmacy and Medical Sciences, Sansom Institute for Health Research, University of South Australia, SA 5000, Australia.
| |
Collapse
|
17
|
Evaluation of a series of 2-napthamide derivatives as inhibitors of the drug efflux pump AcrB for the reversal of antimicrobial resistance. Bioorg Med Chem Lett 2017; 27:733-739. [DOI: 10.1016/j.bmcl.2017.01.042] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 01/13/2017] [Indexed: 11/18/2022]
|
18
|
Evidence of a Substrate-Discriminating Entrance Channel in the Lower Porter Domain of the Multidrug Resistance Efflux Pump AcrB. Antimicrob Agents Chemother 2016; 60:4315-23. [PMID: 27161641 DOI: 10.1128/aac.00314-16] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 05/01/2016] [Indexed: 11/20/2022] Open
Abstract
Efflux pumps of the resistance nodulation cell division (RND) transporter family, such as AcrB of Escherichia coli, play an important role in the development of multidrug resistance, but the molecular basis for their substrate promiscuity is not yet completely understood. From a collection of highly clarithromycin-resistant AcrB periplasmic domain mutants derived from in vitro random mutagenesis, we identified variants with an unusually altered drug resistance pattern characterized by increased susceptibility to many drugs of lower molecular weight, including fluoroquinolones, tetracyclines, and oxazolidinones, but unchanged or increased resistance to drugs of higher molecular weight, including macrolides. Sequencing of 14 such "divergent resistance" phenotype mutants and 15 control mutants showed that this unusual phenotype was associated with mutations at residues I38 and I671 predominantly to phenylalanine and threonine, respectively, both conferring a similar susceptibility pattern. Reconstructed I38F and I671T single mutants as well as an engineered I38F I671T double mutant with proved efflux competence revealed an equivalent phenotype with enhanced or unchanged resistance to many large AcrB substrates but increased susceptibility to several lower-molecular-weight drugs known to bind within the distal binding pocket. The two isoleucines located in close vicinity to each other in the lower porter domain of AcrB beneath the bottom of the proximal binding pocket may be part of a preferential small-drug entrance pathway that is compromised by the mutations. This finding supports recent indications of distinct entrance channels used by compounds with different physicochemical properties, of which molecular size appears to play a prominent role.
Collapse
|
19
|
Dreier J, Ruggerone P. Interaction of antibacterial compounds with RND efflux pumps in Pseudomonas aeruginosa. Front Microbiol 2015; 6:660. [PMID: 26217310 PMCID: PMC4495556 DOI: 10.3389/fmicb.2015.00660] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 06/16/2015] [Indexed: 01/14/2023] Open
Abstract
Pseudomonas aeruginosa infections are becoming increasingly difficult to treat due to intrinsic antibiotic resistance and the propensity of this pathogen to accumulate diverse resistance mechanisms. Hyperexpression of efflux pumps of the Resistance-Nodulation-Cell Division (RND)-type multidrug efflux pumps (e.g., MexAB-OprM), chromosomally encoded by mexAB-oprM, mexCD-oprJ, mexEF-oprN, and mexXY (-oprA) is often detected in clinical isolates and contributes to worrying multi-drug resistance phenotypes. Not all antibiotics are affected to the same extent by the aforementioned RND efflux pumps. The impact of efflux on antibiotic activity varies not only between different classes of antibiotics but also between members of the same family of antibiotics. Subtle differences in physicochemical features of compound-pump and compound-solvent interactions largely determine how compounds are affected by efflux activity. The combination of different high-resolution techniques helps to gain insight into the functioning of these molecular machineries. This review discusses substrate recognition patterns based on experimental evidence and computer simulations with a focus on MexB, the pump subunit of the main RND transporter in P. aeruginosa.
Collapse
Affiliation(s)
- Jürg Dreier
- Basilea Pharmaceutica International Ltd.,Basel, Switzerland
| | - Paolo Ruggerone
- Dipartimento di Fisica, Università di Cagliari – Cittadella UniversitariaMonserrato, Italy
| |
Collapse
|
20
|
Opperman TJ, Nguyen ST. Recent advances toward a molecular mechanism of efflux pump inhibition. Front Microbiol 2015; 6:421. [PMID: 25999939 PMCID: PMC4419859 DOI: 10.3389/fmicb.2015.00421] [Citation(s) in RCA: 175] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 04/21/2015] [Indexed: 12/25/2022] Open
Abstract
Multidrug resistance (MDR) in Gram-negative pathogens, such as the Enterobacteriaceae and Pseudomonas aeruginosa, poses a significant threat to our ability to effectively treat infections caused by these organisms. A major component in the development of the MDR phenotype in Gram-negative bacteria is overexpression of Resistance-Nodulation-Division (RND)-type efflux pumps, which actively pump antibacterial agents and biocides from the periplasm to the outside of the cell. Consequently, bacterial efflux pumps are an important target for developing novel antibacterial treatments. Potent efflux pump inhibitors (EPIs) could be used as adjunctive therapies that would increase the potency of existing antibiotics and decrease the emergence of MDR bacteria. Several potent inhibitors of RND-type efflux pump have been reported in the literature, and at least three of these EPI series were optimized in a pre-clinical development program. However, none of these compounds have been tested in the clinic. One of the major hurdles to the development of EPIs has been the lack of biochemical, computational, and structural methods that could be used to guide rational drug design. Here, we review recent reports that have advanced our understanding of the mechanism of action of several potent EPIs against RND-type pumps.
Collapse
|
21
|
Venter H, Mowla R, Ohene-Agyei T, Ma S. RND-type drug efflux pumps from Gram-negative bacteria: molecular mechanism and inhibition. Front Microbiol 2015; 6:377. [PMID: 25972857 PMCID: PMC4412071 DOI: 10.3389/fmicb.2015.00377] [Citation(s) in RCA: 186] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 04/12/2015] [Indexed: 11/13/2022] Open
Abstract
Drug efflux protein complexes confer multidrug resistance on bacteria by transporting a wide spectrum of structurally diverse antibiotics. Moreover, organisms can only acquire resistance in the presence of an active efflux pump. The substrate range of drug efflux pumps is not limited to antibiotics, but it also includes toxins, dyes, detergents, lipids, and molecules involved in quorum sensing; hence efflux pumps are also associated with virulence and biofilm formation. Inhibitors of efflux pumps are therefore attractive compounds to reverse multidrug resistance and to prevent the development of resistance in clinically relevant bacterial pathogens. Recent successes on the structure determination and functional analysis of the AcrB and MexB components of the AcrAB-TolC and MexAB-OprM drug efflux systems as well as the structure of the fully assembled, functional triparted AcrAB-TolC complex significantly contributed to our understanding of the mechanism of substrate transport and the options for inhibition of efflux. These data, combined with the well-developed methodologies for measuring efflux pump inhibition, could allow the rational design, and subsequent experimental verification of potential efflux pump inhibitors (EPIs). In this review we will explore how the available biochemical and structural information can be translated into the discovery and development of new compounds that could reverse drug resistance in Gram-negative pathogens. The current literature on EPIs will also be analyzed and the reasons why no compounds have yet progressed into clinical use will be explored.
Collapse
Affiliation(s)
- Henrietta Venter
- School of Pharmacy and Medical Sciences, Sansom Institute for Health Research, University of South Australia Adelaide, SA, Australia
| | - Rumana Mowla
- School of Pharmacy and Medical Sciences, Sansom Institute for Health Research, University of South Australia Adelaide, SA, Australia
| | | | - Shutao Ma
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Shandong University Jinan, China
| |
Collapse
|
22
|
Wang Z, Zhong M, Lu W, Chai Q, Wei Y. Repressive mutations restore function-loss caused by the disruption of trimerization in Escherichia coli multidrug transporter AcrB. Front Microbiol 2015; 6:4. [PMID: 25657644 PMCID: PMC4303003 DOI: 10.3389/fmicb.2015.00004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 01/03/2015] [Indexed: 11/13/2022] Open
Abstract
AcrAB-TolC and their homologs are major multidrug efflux systems in Gram-negative bacteria. The inner membrane component AcrB functions as a trimer. Replacement of Pro223 by Gly in AcrB decreases the trimer stability and drastically reduces the drug efflux activity. The goal of this study is to identify suppressor mutations that restore function to mutant AcrBP223G and explore the mechanism of function recovery. Two methods were used to introduce random mutations into the plasmid of AcrBP223G. Mutants with elevated drug efflux activity were identified, purified, and characterized to examine their expression level, trimer stability, interaction with AcrA, and substrate binding. Nine single-site repressor mutations were identified, including T199M, D256N, A209V, G257V, M662I, Q737L, D788K, P800S, and E810K. Except for M662I, all other mutations located in the docking region of the periplasmic domain. While three mutations, T199M, A209V, and D256N, significantly increased the trimer stability, none of them restored the trimer affinity to the wild type level. M662, the only site of mutation that located in the porter domain, was involved in substrate binding. Our results suggest that the function loss resulted from compromised AcrB trimerization could be restored through various mechanisms involving the compensation of trimer stability and substrate binding.
Collapse
Affiliation(s)
- Zhaoshuai Wang
- Department of Chemistry, University of Kentucky Lexington, KY, USA
| | - Meng Zhong
- Department of Chemistry, University of Kentucky Lexington, KY, USA
| | - Wei Lu
- Department of Chemistry, University of Kentucky Lexington, KY, USA
| | - Qian Chai
- Department of Chemistry, University of Kentucky Lexington, KY, USA
| | - Yinan Wei
- Department of Chemistry, University of Kentucky Lexington, KY, USA
| |
Collapse
|
23
|
Saidijam M, Patching SG. Amino acid composition analysis of secondary transport proteins from Escherichia coli with relation to functional classification, ligand specificity and structure. J Biomol Struct Dyn 2015; 33:2205-20. [DOI: 10.1080/07391102.2014.998283] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Massoud Saidijam
- Department of Molecular Medicine and Genetics, Research Centre for Molecular Medicine, School of Medicine, Hamadan University of Medical Sciences , Hamadan, Iran
| | - Simon G. Patching
- Department of Molecular Medicine and Genetics, Research Centre for Molecular Medicine, School of Medicine, Hamadan University of Medical Sciences , Hamadan, Iran
| |
Collapse
|
24
|
Folding energetics and oligomerization of polytopic α-helical transmembrane proteins. Arch Biochem Biophys 2014; 564:281-96. [DOI: 10.1016/j.abb.2014.07.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 06/26/2014] [Accepted: 07/14/2014] [Indexed: 01/06/2023]
|
25
|
Laehnemann D, Peña-Miller R, Rosenstiel P, Beardmore R, Jansen G, Schulenburg H. Genomics of rapid adaptation to antibiotics: convergent evolution and scalable sequence amplification. Genome Biol Evol 2014; 6:1287-301. [PMID: 24850796 PMCID: PMC4079197 DOI: 10.1093/gbe/evu106] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Evolutionary adaptation can be extremely fast, especially in response to high selection intensities. A prime example is the surge of antibiotic resistance in bacteria. The genomic underpinnings of such rapid changes may provide information on the genetic processes that enhance fast responses and the particular trait functions under selection. Here, we use experimentally evolved Escherichia coli for a detailed dissection of the genomics of rapid antibiotic resistance evolution. Our new analyses demonstrate that amplification of a sequence region containing several known antibiotic resistance genes represents a fast genomic response mechanism under high antibiotic stress, here exerted by drug combination. In particular, higher dosage of such antibiotic combinations coincided with higher copy number of the sequence region. The amplification appears to be evolutionarily costly, because amplification levels rapidly dropped after removal of the drugs. Our results suggest that amplification is a scalable process, as copy number rapidly changes in response to the selective pressure encountered. Moreover, repeated patterns of convergent evolution were found across the experimentally evolved bacterial populations, including those with lower antibiotic selection intensities. Intriguingly, convergent evolution was identified on different organizational levels, ranging from the above sequence amplification, high variant frequencies in specific genes, prevalence of individual nonsynonymous mutations to the unusual repeated occurrence of a particular synonymous mutation in Glycine codons. We conclude that constrained evolutionary trajectories underlie rapid adaptation to antibiotics. Of the identified genomic changes, sequence amplification seems to represent the most potent, albeit costly genomic response mechanism to high antibiotic stress.
Collapse
Affiliation(s)
- David Laehnemann
- Department of Evolutionary Ecology and Genetics, University of Kiel, Germany
| | - Rafael Peña-Miller
- Biosciences, Geoffrey Pope Building, University of Exeter, United KingdomDepartment of Zoology, University of Oxford, United Kingdom
| | - Philip Rosenstiel
- Institute for Clinical Molecular Biology, University of Kiel, Germany
| | - Robert Beardmore
- Biosciences, Geoffrey Pope Building, University of Exeter, United Kingdom
| | - Gunther Jansen
- Department of Evolutionary Ecology and Genetics, University of Kiel, Germany
| | - Hinrich Schulenburg
- Department of Evolutionary Ecology and Genetics, University of Kiel, Germany
| |
Collapse
|
26
|
Interaction between mutations and regulation of gene expression during development of de novo antibiotic resistance. Antimicrob Agents Chemother 2014; 58:4371-9. [PMID: 24841263 DOI: 10.1128/aac.02892-14] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Bacteria can become resistant not only by horizontal gene transfer or other forms of exchange of genetic information but also by de novo by adaptation at the gene expression level and through DNA mutations. The interrelationship between changes in gene expression and DNA mutations during acquisition of resistance is not well documented. In addition, it is not known whether the DNA mutations leading to resistance always occur in the same order and whether the final result is always identical. The expression of >4,000 genes in Escherichia coli was compared upon adaptation to amoxicillin, tetracycline, and enrofloxacin. During adaptation, known resistance genes were sequenced for mutations that cause resistance. The order of mutations varied within two sets of strains adapted in parallel to amoxicillin and enrofloxacin, respectively, whereas the buildup of resistance was very similar. No specific mutations were related to the rather modest increase in tetracycline resistance. Ribosome-sensed induction and efflux pump activation initially protected the cell through induction of expression and allowed it to survive low levels of antibiotics. Subsequently, mutations were promoted by the stress-induced SOS response that stimulated modulation of genetic instability, and these mutations resulted in resistance to even higher antibiotic concentrations. The initial adaptation at the expression level enabled a subsequent trial and error search for the optimal mutations. The quantitative adjustment of cellular processes at different levels accelerated the acquisition of antibiotic resistance.
Collapse
|
27
|
Cheng YA, Jee J, Hsu G, Huang Y, Chen C, Lin CP. A markerless protocol for genetic analysis of Aggregatibacter actinomycetemcomitans. J Formos Med Assoc 2014; 113:114-23. [PMID: 24530245 DOI: 10.1016/j.jfma.2012.05.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2012] [Revised: 05/09/2012] [Accepted: 05/15/2012] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND/PURPOSE The genomes of different Aggregatibacter actinomycetemcomitans (A actinomycetemcomitans) strains contain many strain-specific genes and genomic islands (defined as DNA found in some but not all strains) of unknown functions. Genetic analysis for the functions of these islands will be constrained by the limited availability of genetic markers and vectors for A actinomycetemcomitans. In this study, we tested a novel genetic approach of gene deletion and restoration in a naturally competent A actinomycetemcomitans strain D7S-1. METHODS Specific genes' deletion mutants and mutants restored with the deleted genes were constructed by a markerless loxP/Cre system. In mutants with sequential deletion of multiple genes loxP with different spacer regions were used to avoid unwanted recombinations between loxP sites. RESULTS Eight single-gene deletion mutants, four multiple-gene deletion mutants, and two mutants with restored genes were constructed. No unintended non-specific deletion mutants were generated by this protocol. The protocol did not negatively affect the growth and biofilm formation of A actinomycetemcomitans. CONCLUSION The protocol described in this study is efficient and specific for genetic manipulation of A actinomycetemcomitans, and will be amenable for functional analysis of multiple genes in A actinomycetemcomitans.
Collapse
Affiliation(s)
- Ya-An Cheng
- School of Dentistry and Graduate Institute of Clinical Dentistry, National Taiwan University, Taipei, Taiwan
| | - Jason Jee
- Division of Periodontology, Diagnostic Sciences and Dental Hygiene, Herman Ostrow School of Dentistry of the University of Southern California, Los Angeles, CA, USA
| | - Genie Hsu
- Division of Periodontology, Diagnostic Sciences and Dental Hygiene, Herman Ostrow School of Dentistry of the University of Southern California, Los Angeles, CA, USA
| | - Yanyan Huang
- Division of Periodontology, Diagnostic Sciences and Dental Hygiene, Herman Ostrow School of Dentistry of the University of Southern California, Los Angeles, CA, USA
| | - Casey Chen
- Division of Periodontology, Diagnostic Sciences and Dental Hygiene, Herman Ostrow School of Dentistry of the University of Southern California, Los Angeles, CA, USA
| | - Chun-Pin Lin
- School of Dentistry and Graduate Institute of Clinical Dentistry, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
28
|
Kapoor V, Wendell D. Engineering bacterial efflux pumps for solar-powered bioremediation of surface waters. NANO LETTERS 2013; 13:2189-2193. [PMID: 23581993 DOI: 10.1021/nl400691d] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Antibiotics are difficult to selectively remove from surface waters by present treatment methods. Bacterial efflux pumps have evolved the ability to discriminately expel antibiotics and other noxious agents via proton and ATP driven pathways. Here, we describe light-dependent removal of antibiotics by engineering the bacterial efflux pump AcrB into a proteovesicle system. We have created a chimeric protein with the requisite proton motive force by coupling AcrB to the light-driven proton pump Delta-rhodopsin (dR) via a glycophorin A transmembrane domain. This creates a solar powered protein material capable of selectively capturing antibiotics from bulk solutions. Using environmental water and direct sunlight, our AcrB-dR vesicles removed almost twice as much antibiotic as the treatment standard, activated carbon. Altogether, the AcrB-dR system provides an effective means of extracting antibiotics from surface waters as well as potential antibiotic recovery through vesicle solubilization.
Collapse
Affiliation(s)
- Vikram Kapoor
- School of Energy, Environmental, Biological and Medical Engineering, University of Cincinnati, 2901 Woodside Drive, 705 Engineering Research Center, Cincinnati, Ohio 45221, USA
| | | |
Collapse
|
29
|
Janganan TK, Bavro VN, Zhang L, Borges-Walmsley MI, Walmsley AR. Tripartite efflux pumps: energy is required for dissociation, but not assembly or opening of the outer membrane channel of the pump. Mol Microbiol 2013; 88:590-602. [PMID: 23565750 PMCID: PMC3664412 DOI: 10.1111/mmi.12211] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/19/2013] [Indexed: 11/26/2022]
Abstract
The MtrCDE multidrug pump, from Neisseria gonorrhoeae, is assembled from the inner and outer membrane proteins MtrD and MtrE, which are connected by the periplasmic membrane fusion protein MtrC. Although it is clear that MtrD delivers drugs to the channel of MtrE, it remains unclear how drug delivery and channel opening are connected. We used a vancomycin sensitivity assay to test for opening of the MtrE channel. Cells expressing MtrE or MtrE-E434K were insensitive to vancomycin; but became moderately and highly sensitive to vancomycin respectively, when coexpressed with MtrC, suggesting that the MtrE channel opening requires MtrC binding and is energy-independent. Cells expressing wild-type MtrD, in an MtrCE background, were vancomycin-insensitive, but moderately sensitive in an MtrCE-E434K background. The mutation of residues involved in proton translocation inactivated MtrD and abolished drug efflux, rendered both MtrE and MtrE-E434K vancomycin-insensitive; imply that the pump-component interactions are preserved, and that the complex is stable in the absence of proton flux, thus sealing the open end of MtrE. Following the energy-dependent dissociation of the tripartite complex, the MtrE channel is able to reseal, while MtrE-E434K is unable to do so, resulting in the vancomycin-sensitive phenotype. Thus, our findings suggest that opening of the OMP via interaction with the MFP is energy-independent, while both drug export and complex dissociation require active proton flux.
Collapse
Affiliation(s)
- Thamarai K Janganan
- School of Biological and Biomedical Sciences, Durham University, South Road, Durham, DH1 3LE, UK
| | | | | | | | | |
Collapse
|
30
|
Lacal J, Reyes-Darias JA, García-Fontana C, Ramos JL, Krell T. Tactic responses to pollutants and their potential to increase biodegradation efficiency. J Appl Microbiol 2012; 114:923-33. [PMID: 23163356 DOI: 10.1111/jam.12076] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Revised: 11/07/2012] [Accepted: 11/11/2012] [Indexed: 11/26/2022]
Abstract
A significant number of bacterial strains are able to use toxic aromatic hydrocarbons as carbon and energy sources. In a number of cases, the evolution of the corresponding degradation pathway was accompanied by the evolution of tactic behaviours either towards or away from these toxic carbon sources. Reports are reviewed which show that a chemoattraction to heterogeneously distributed aromatic pollutants increases the bioavailability of these compounds and their biodegradation efficiency. An extreme form of chemoattraction towards aromatic pollutants, termed 'hyperchemotaxis', was described for Pseudomonas putida DOT-T1E, which is based on the action of the plasmid-encoded McpT chemoreceptor. Cells with this phenotype were found of being able to approach and of establishing contact with undiluted crude oil samples. Although close McpT homologues are found on other degradation plasmids, the sequence of their ligand-binding domains does not share significant similarity with that of NahY, the other characterized chemoreceptor for aromatic hydrocarbons. This may suggest the existence of at least two families of chemoreceptors for aromatic pollutants. The use of receptor chimers comprising the ligand-binding region of McpT for biosensing purposes is discussed.
Collapse
Affiliation(s)
- J Lacal
- Department of Environmental Protection, Estación Experimental del Zaidín, Consejo Superior de Investigaciones Científicas, Granada, Spain
| | | | | | | | | |
Collapse
|
31
|
Lu W, Chai Q, Zhong M, Yu L, Fang J, Wang T, Li H, Zhu H, Wei Y. Assembling of AcrB trimer in cell membrane. J Mol Biol 2012; 423:123-34. [PMID: 22766312 PMCID: PMC5699209 DOI: 10.1016/j.jmb.2012.06.036] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2012] [Revised: 06/26/2012] [Accepted: 06/26/2012] [Indexed: 11/16/2022]
Abstract
Many membrane proteins exist and function as oligomers, but how monomers oligomerize in the cell membrane remains poorly understood. AcrB is an obligate homo-trimer. We previously found that the folding of individual subunit precedes oligomerization. Following folding, individual AcrB subunits must locate and interact with each other in order to dimerize and eventually trimerize. It has been unclear if AcrB trimerization is a spontaneous process following the "chance encounter and random assembling" mechanism. In other words, it is currently unknown whether monomeric subunits diffuse freely to "search" for each other after they are co-translationally inserted and folded into the cell membrane. Using four sets of experiments exploiting AcrB variants with different fusion tags, disulfide trapping, and activity measurement, here we showed that AcrB variants co-expressed in the same Escherichia coli cell did co-assemble into hybrid trimers in vivo. However, the level of co-assembly measured experimentally was not consistent with calculations derived from random assembling. The potential role of the polysome structure during protein translation and the resultant clustering effect were discussed as a potential explanation for the observed bias in AcrB subunit assembling in vivo. Our results provide new insights into the dynamic assembling and equilibration process of obligate homo-oligomeric membrane proteins in the cell membrane.
Collapse
Affiliation(s)
- Wei Lu
- Department of Chemistry, University of Kentucky, Lexington, KY 40506, USA
| | - Qian Chai
- Department of Chemistry, University of Kentucky, Lexington, KY 40506, USA
| | - Meng Zhong
- Department of Chemistry, University of Kentucky, Lexington, KY 40506, USA
| | - Linliang Yu
- Department of Chemistry, University of Kentucky, Lexington, KY 40506, USA
| | - Jun Fang
- Department of Chemistry, University of Kentucky, Lexington, KY 40506, USA
| | - Tong Wang
- Department of Biology, Brookhaven National Laboratory, Upton, NY 11973, USA
| | - Huilin Li
- Department of Biology, Brookhaven National Laboratory, Upton, NY 11973, USA
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Haining Zhu
- Department of Molecular and Cellular Biochemistry and Center for Structural Biology, University of Kentucky, Lexington, KY 40506, USA
| | - Yinan Wei
- Department of Chemistry, University of Kentucky, Lexington, KY 40506, USA
| |
Collapse
|
32
|
Ohene-Agyei T, Lea JD, Venter H. Mutations in MexB that affect the efflux of antibiotics with cytoplasmic targets. FEMS Microbiol Lett 2012; 333:20-7. [PMID: 22568688 DOI: 10.1111/j.1574-6968.2012.02594.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2012] [Revised: 04/24/2012] [Accepted: 05/06/2012] [Indexed: 11/30/2022] Open
Abstract
Drug efflux pumps such as MexAB-OprM from Pseudomonas aeruginosa confer resistance to a wide range of chemically different compounds. Within the tripartite assembly, the inner membrane protein MexB is mainly responsible for substrate recognition. Recently, considerable advances have been made in elucidating the drug efflux pathway through the large periplasmic domains of resistance-nodulation-division (RND) transporters. However, little is known about the role of amino acids in other parts of the protein. We have investigated the role of two conserved phenylalanine residues that are aligned around the cytoplasmic side of the central cavity of MexB. The two conserved phenylalanine residues have been mutated to alanine residues (FAFA MexB). The interaction of the wild-type and mutant proteins with a variety of drugs from different classes was investigated by assays of cytotoxicity and drug transport. The FAFA mutation affected the efflux of compounds that have targets inside the cell, but antibiotics that act on cell wall synthesis and membrane probes were unaffected. Combined, our results indicate the presence of a hitherto unidentified cytoplasmic-binding site in RND drug transporters and enhance our understanding of the molecular mechanisms that govern drug resistance in Gram-negative pathogens.
Collapse
|
33
|
Nikaido H, Pagès JM. Broad-specificity efflux pumps and their role in multidrug resistance of Gram-negative bacteria. FEMS Microbiol Rev 2011; 36:340-63. [PMID: 21707670 DOI: 10.1111/j.1574-6976.2011.00290.x] [Citation(s) in RCA: 510] [Impact Index Per Article: 36.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Antibiotic resistance mechanisms reported in Gram-negative bacteria are causing a worldwide health problem. The continuous dissemination of 'multidrug-resistant' (MDR) bacteria drastically reduces the efficacy of our antibiotic 'arsenal' and consequently increases the frequency of therapeutic failure. In MDR bacteria, the overexpression of efflux pumps that expel structurally unrelated drugs contributes to the reduced susceptibility by decreasing the intracellular concentration of antibiotics. During the last decade, several clinical data have indicated an increasing involvement of efflux pumps in the emergence and dissemination of resistant Gram-negative bacteria. It is necessary to clearly define the molecular, functional and genetic bases of the efflux pump in order to understand the translocation of antibiotic molecules through the efflux transporter. The recent investigation on the efflux pump AcrB at its structural and physiological levels, including the identification of drug affinity sites and kinetic parameters for various antibiotics, may pave the way towards the rational development of an improved new generation of antibacterial agents as well as efflux inhibitors in order to efficiently combat efflux-based resistance mechanisms.
Collapse
Affiliation(s)
- Hiroshi Nikaido
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | | |
Collapse
|
34
|
Perez C, Khafizov K, Forrest LR, Krämer R, Ziegler C. The role of trimerization in the osmoregulated betaine transporter BetP. EMBO Rep 2011; 12:804-10. [PMID: 21681199 DOI: 10.1038/embor.2011.102] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2011] [Revised: 05/04/2011] [Accepted: 05/06/2011] [Indexed: 11/09/2022] Open
Abstract
The osmoregulated betaine transporter BetP is a stable trimer. Structural studies have shown that individual protomers can adopt distinct transport conformations, implying a functional role for the trimeric state in transport, although the role of trimerization in regulation is not yet understood. We designed putative monomeric mutants by molecular-dynamics simulations and in silico alanine-scanning mutagenesis. Several mutants including BetP-W101A/T351A were monomeric in detergent as well as in the membrane, as shown by blue native gel electrophoresis, crosslinking and electron microscopy. This monomeric form retains the ability to accumulate betaine, but is no longer regulated by hyperosmotic shock.
Collapse
Affiliation(s)
- Camilo Perez
- Department of Structural Biology, Max-Planck Institute of Biophysics, Frankfurt am Main 60438, Germany
| | | | | | | | | |
Collapse
|
35
|
Schulz R, Vargiu AV, Ruggerone P, Kleinekathöfer U. Role of Water during the Extrusion of Substrates by the Efflux Transporter AcrB. J Phys Chem B 2011; 115:8278-87. [DOI: 10.1021/jp200996x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Robert Schulz
- School of Engineering and Science, Jacobs University Bremen, Campus Ring 1, 28759 Bremen,
Germany
| | - Attilio V. Vargiu
- CNR-IOM, Unità SLACS,
c/o Dipartimento di Fisica, Università di Cagliari, s.p. Monserrato-Sestu km 0.7, I-09042 Monserrato (CA), Italy
| | - Paolo Ruggerone
- CNR-IOM, Unità SLACS,
c/o Dipartimento di Fisica, Università di Cagliari, s.p. Monserrato-Sestu km 0.7, I-09042 Monserrato (CA), Italy
| | - Ulrich Kleinekathöfer
- School of Engineering and Science, Jacobs University Bremen, Campus Ring 1, 28759 Bremen,
Germany
| |
Collapse
|
36
|
Castaneda-Garcia A, Do TT, Blazquez J. The K+ uptake regulator TrkA controls membrane potential, pH homeostasis and multidrug susceptibility in Mycobacterium smegmatis. J Antimicrob Chemother 2011; 66:1489-98. [DOI: 10.1093/jac/dkr165] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
37
|
Amaral L, Cerca P, Spengler G, Machado L, Martins A, Couto I, Viveiros M, Fanning S, Pagès JM. Ethidium bromide efflux by Salmonella: modulation by metabolic energy, pH, ions and phenothiazines. Int J Antimicrob Agents 2011; 38:140-5. [PMID: 21565465 DOI: 10.1016/j.ijantimicag.2011.03.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2011] [Revised: 03/21/2011] [Accepted: 03/22/2011] [Indexed: 11/16/2022]
Abstract
The main efflux pump of Salmonella enterica serotype Enteritidis, which obtains its energy for the extrusion of noxious agents from the proton-motive force, was studied with the aid of an ethidium bromide (EtBr) semi-automated method under conditions that define the role of metabolic energy, ions and pH in the extrusion of the universal substrate EtBr. The results obtained in this study indicate that in minimal medium containing sodium at pH 5 efflux of EtBr is independent of glucose, whereas at pH 8 metabolic energy is an absolute requirement for the maintenance of efflux. In deionised water at pH 5.5, metabolic energy is required for the maintenance of efflux. The inhibitory effect of the ionophore carbonyl cyanide m-chlorophenylhydrazone (CCCP) on efflux is shown to be minimised by low pH, and at high pH by metabolic energy. Similarly, thioridazine, an inhibitor of metabolic enzymes, inhibits efflux of EtBr only at pH 8 and the degree of inhibition is lessened by the presence of metabolic energy.
Collapse
Affiliation(s)
- Leonard Amaral
- Grupo de Micobactérias, Unidade de Microbiologia Médica, Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa, Rua da Junqueira 100, 1349-008 Lisboa, Portugal.
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Bolla JM, Alibert-Franco S, Handzlik J, Chevalier J, Mahamoud A, Boyer G, Kieć-Kononowicz K, Pagès JM. Strategies for bypassing the membrane barrier in multidrug resistant Gram-negative bacteria. FEBS Lett 2011; 585:1682-90. [DOI: 10.1016/j.febslet.2011.04.054] [Citation(s) in RCA: 141] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2011] [Revised: 04/20/2011] [Accepted: 04/21/2011] [Indexed: 10/18/2022]
|
39
|
Barabote RD, Thekkiniath J, Strauss RE, Vediyappan G, Fralick JA, San Francisco MJ. Xenobiotic efflux in bacteria and fungi: a genomics update. ADVANCES IN ENZYMOLOGY AND RELATED AREAS OF MOLECULAR BIOLOGY 2011; 77:237-306. [PMID: 21692371 DOI: 10.1002/9780470920541.ch6] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Ravi D Barabote
- Department of Plant Sciences, University of California, Davis, California, USA
| | | | | | | | | | | |
Collapse
|
40
|
Mahamoud A, Chevalier J, Baitiche M, Adam E, Pagès JM. An alkylaminoquinazoline restores antibiotic activity in Gram-negative resistant isolates. MICROBIOLOGY-SGM 2010; 157:566-571. [PMID: 21071494 DOI: 10.1099/mic.0.045716-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
To date, various bacterial drug efflux pump inhibitors (EPIs) have been described. They exhibit variability in their activity spectrum with respect to antibiotic structural class and bacterial species. Among the various 4-alkylaminoquinazoline derivatives synthesized and studied in this work, one molecule, 1167, increased the susceptibility of important human-pathogenic, resistant, Gram-negative bacteria towards different antibiotic classes. This 4-(3-morpholinopropylamino)-quinazoline induced an increase in the activity of chloramphenicol, nalidixic acid, norfloxacin and sparfloxacin, which are substrates of the AcrAB-TolC and MexAB-OprM efflux pumps that act in these multidrug-resistant isolates. In addition, 1167 increased the intracellular concentration of chloramphenicol in efflux pump-overproducing strains. The rate of restoration depended on the structure of the antibiotic, suggesting that different sites in the efflux pumps may be involved. A molecule exhibiting a morpholine functional group and a propyl extension of the side chain was more active.
Collapse
Affiliation(s)
- Abdallah Mahamoud
- UMR-MD-1, Facultés de Médecine et de Pharmacie, Université de la Méditerranée, Marseille, France
| | - Jacqueline Chevalier
- UMR-MD-1, Facultés de Médecine et de Pharmacie, Université de la Méditerranée, Marseille, France
| | - Milad Baitiche
- UMR-MD-1, Facultés de Médecine et de Pharmacie, Université de la Méditerranée, Marseille, France
| | - Elissavet Adam
- UMR-MD-1, Facultés de Médecine et de Pharmacie, Université de la Méditerranée, Marseille, France
| | - Jean-Marie Pagès
- UMR-MD-1, Facultés de Médecine et de Pharmacie, Université de la Méditerranée, Marseille, France
| |
Collapse
|
41
|
Forrest LR, Krämer R, Ziegler C. The structural basis of secondary active transport mechanisms. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2010; 1807:167-88. [PMID: 21029721 DOI: 10.1016/j.bbabio.2010.10.014] [Citation(s) in RCA: 324] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 07/16/2010] [Revised: 10/13/2010] [Accepted: 10/15/2010] [Indexed: 12/22/2022]
Abstract
Secondary active transporters couple the free energy of the electrochemical potential of one solute to the transmembrane movement of another. As a basic mechanistic explanation for their transport function the model of alternating access was put forward more than 40 years ago, and has been supported by numerous kinetic, biochemical and biophysical studies. According to this model, the transporter exposes its substrate binding site(s) to one side of the membrane or the other during transport catalysis, requiring a substantial conformational change of the carrier protein. In the light of recent structural data for a number of secondary transport proteins, we analyze the model of alternating access in more detail, and correlate it with specific structural and chemical properties of the transporters, such as their assignment to different functional states in the catalytic cycle of the respective transporter, the definition of substrate binding sites, the type of movement of the central part of the carrier harboring the substrate binding site, as well as the impact of symmetry on fold-specific conformational changes. Besides mediating the transmembrane movement of solutes, the mechanism of secondary carriers inherently involves a mechanistic coupling of substrate flux to the electrochemical potential of co-substrate ions or solutes. Mainly because of limitations in resolution of available transporter structures, this important aspect of secondary transport cannot yet be substantiated by structural data to the same extent as the conformational change aspect. We summarize the concepts of coupling in secondary transport and discuss them in the context of the available evidence for ion binding to specific sites and the impact of the ions on the conformational state of the carrier protein, which together lead to mechanistic models for coupling.
Collapse
Affiliation(s)
- Lucy R Forrest
- Structural Biology Department, Max Planck Institute for Biophysics, Frankfurt, Germany
| | | | | |
Collapse
|
42
|
Kurumbang NP, Park JW, Yoon YJ, Liou K, Sohng JK. Heterologous production of ribostamycin derivatives in engineered Escherichia coli. Res Microbiol 2010; 161:526-33. [PMID: 20561584 DOI: 10.1016/j.resmic.2010.04.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2009] [Revised: 02/25/2010] [Accepted: 04/29/2010] [Indexed: 11/18/2022]
Abstract
Aminoglycosides are a class of important antibiotic compounds used for various therapeutic indications. In recent times, their efficacy has been curtailed due to the rapid development of bacterial resistance. There is a need to develop novel derivatives with an improved spectrum of activity and higher sensitivity against pathogenic bacteria. Although efforts have been focused on the development of newer therapeutic agents by chemical synthesis, to our knowledge, there has been no attempt to harness the potential of microorganisms for this purpose. Escherichia coli affords a widely studied cellular system that could be utilized not only for understanding but also for attempting to engineer the biosynthetic pathway of secondary metabolites. The primary metabolic pathway of E. coli can be engineered to divert the precursor pool required for the biosynthesis of secondary metabolites. Utilizing this approach previously, we engineered E. coli host and generated E. coli M1. Here, we produced a ribostamycin derivative in the engineered host by heterologous expression of the recombinants constructed from the genes encoding the biosynthetic pathway in aminoglycoside-producing strains. The products obtained from the transformants were isolated, analyzed and verified to be ribostamycin derivatives. The study further demonstrated the importance of E. coli as surrogate antibiotic producer and also offered future possibility for the production of other aminoglycoside derivatives through genetic engineering and expression in a heterologous background.
Collapse
Affiliation(s)
- Nagendra Prasad Kurumbang
- Institute of Biomolecule Reconstruction, Department of Pharmaceutical Engineering, Sun Moon University, 100, Kalsan-ri, Tangjeonmyun, Asansi, Chungnam 336-708, Republic of Korea
| | | | | | | | | |
Collapse
|
43
|
Schulz R, Vargiu AV, Collu F, Kleinekathöfer U, Ruggerone P. Functional rotation of the transporter AcrB: insights into drug extrusion from simulations. PLoS Comput Biol 2010; 6:e1000806. [PMID: 20548943 PMCID: PMC2883587 DOI: 10.1371/journal.pcbi.1000806] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2009] [Accepted: 05/05/2010] [Indexed: 01/16/2023] Open
Abstract
The tripartite complex AcrAB-TolC is the major efflux system in Escherichia coli. It extrudes a wide spectrum of noxious compounds out of the bacterium, including many antibiotics. Its active part, the homotrimeric transporter AcrB, is responsible for the selective binding of substrates and energy transduction. Based on available crystal structures and biochemical data, the transport of substrates by AcrB has been proposed to take place via a functional rotation, in which each monomer assumes a particular conformation. However, there is no molecular-level description of the conformational changes associated with the rotation and their connection to drug extrusion. To obtain insights thereon, we have performed extensive targeted molecular dynamics simulations mimicking the functional rotation of AcrB containing doxorubicin, one of the two substrates that were co-crystallized so far. The simulations, including almost half a million atoms, have been used to test several hypotheses concerning the structure-dynamics-function relationship of this transporter. Our results indicate that, upon induction of conformational changes, the substrate detaches from the binding pocket and approaches the gate to the central funnel. Furthermore, we provide strong evidence for the proposed peristaltic transport involving a zipper-like closure of the binding pocket, responsible for the displacement of the drug. A concerted opening of the channel between the binding pocket and the gate further favors the displacement of the drug. This microscopically well-funded information allows one to identify the role of specific amino acids during the transitions and to shed light on the functioning of AcrB. In nature, bacteria have to resist several toxic threats to be able to survive, from bile acids in intestines up to antibiotics. The Escherichia coli bacterium, which usually is a commensal inhabitant of human intestines, can also acquire pathogenic properties which would harm the human body. To dispose of toxic compounds, E. coli has developed a protein machinery which is called “efflux pump”. Here, we studied the dynamics of the transporter protein AcrB, a component of the E. coli major efflux system, in complex with an antibiotic (doxorubicin). We used computer simulations to complement the existing experimental data. Our purpose was to gain more detailed insights into the pumping mechanism at the molecular level. In our simulations the drug leaves the binding pocket upon induction of functional rotation in the protein, although a complete extrusion was never observed. A peristaltic motion, which starts with a zipper-like closure of the interior of the protein, is an important step for the extrusion of the drug. Interestingly, such a peristaltic mechanism of pumping has been suggested before on the basis of structural data. The molecular details obtained in this study shall deepen the understanding of the functioning of the efflux pump.
Collapse
Affiliation(s)
- Robert Schulz
- School of Engineering and Science, Jacobs University Bremen, Bremen, Germany
| | - Attilio V. Vargiu
- Istituto Officina dei Materiali del CNR, UOS SLACS and Dipartimento di Fisica, Universita' degli Studi di Cagliari, Monserrato, Italy
- * E-mail:
| | - Francesca Collu
- Istituto Officina dei Materiali del CNR, UOS SLACS and Dipartimento di Fisica, Universita' degli Studi di Cagliari, Monserrato, Italy
| | | | - Paolo Ruggerone
- Istituto Officina dei Materiali del CNR, UOS SLACS and Dipartimento di Fisica, Universita' degli Studi di Cagliari, Monserrato, Italy
| |
Collapse
|
44
|
Kim EH, Rensing C, McEvoy MM. Chaperone-mediated copper handling in the periplasm. Nat Prod Rep 2010; 27:711-9. [PMID: 20442961 DOI: 10.1039/b906681k] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Eun-Hae Kim
- Department of Soil, Water, and Environmental Science, University of Arizona, Tucson, AZ 85721, USA
| | | | | |
Collapse
|