1
|
González-López A, Ge X, Larsson DSD, Sihlbom Wallem C, Sanyal S, Selmer M. Structural mechanism of FusB-mediated rescue from fusidic acid inhibition of protein synthesis. Nat Commun 2025; 16:3693. [PMID: 40251147 PMCID: PMC12008383 DOI: 10.1038/s41467-025-58902-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 04/04/2025] [Indexed: 04/20/2025] Open
Abstract
The antibiotic resistance protein FusB rescues protein synthesis from inhibition by fusidic acid (FA), which locks elongation factor G (EF-G) to the ribosome after GTP hydrolysis. Here, we present time-resolved single-particle cryo-EM structures explaining the mechanism of FusB-mediated rescue. FusB binds to the FA-trapped EF-G on the ribosome, causing large-scale conformational changes of EF-G that break interactions with the ribosome, tRNA, and mRNA. This leads to dissociation of EF-G from the ribosome, followed by FA release. We also observe two independent binding sites of FusB on the classical-state ribosome, overlapping with the binding site of EF-G to each of the ribosomal subunits, yet not inhibiting tRNA delivery. The affinity of FusB to the ribosome and the concentration of FusB in S. aureus during FusB-mediated resistance support that direct binding of FusB to ribosomes could occur in the cell. Our results reveal an intricate resistance mechanism involving specific interactions of FusB with both EF-G and the ribosome, and a non-canonical release pathway of EF-G.
Collapse
Affiliation(s)
- Adrián González-López
- Department of Cell and Molecular Biology, Uppsala University, BMC, Uppsala, Sweden
- Uppsala Antibiotic Center, Uppsala University, Uppsala, Sweden
| | - Xueliang Ge
- Department of Cell and Molecular Biology, Uppsala University, BMC, Uppsala, Sweden
| | - Daniel S D Larsson
- Department of Cell and Molecular Biology, Uppsala University, BMC, Uppsala, Sweden
| | - Carina Sihlbom Wallem
- Proteomics Core Facility, Scilifelab and University of Gothenburg, Gothenburg, Sweden
| | - Suparna Sanyal
- Department of Cell and Molecular Biology, Uppsala University, BMC, Uppsala, Sweden
| | - Maria Selmer
- Department of Cell and Molecular Biology, Uppsala University, BMC, Uppsala, Sweden.
- Uppsala Antibiotic Center, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
2
|
González-López A, Larsson DSD, Koripella RK, Cain BN, Chavez MG, Hergenrother PJ, Sanyal S, Selmer M. Structures of the Staphylococcus aureus ribosome inhibited by fusidic acid and fusidic acid cyclopentane. Sci Rep 2024; 14:14253. [PMID: 38902339 PMCID: PMC11190147 DOI: 10.1038/s41598-024-64868-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 06/13/2024] [Indexed: 06/22/2024] Open
Abstract
The antibiotic fusidic acid (FA) is used to treat Staphylococcus aureus infections. It inhibits protein synthesis by binding to elongation factor G (EF-G) and preventing its release from the ribosome after translocation. While FA, due to permeability issues, is only effective against gram-positive bacteria, the available structures of FA-inhibited complexes are from gram-negative model organisms. To fill this knowledge gap, we solved cryo-EM structures of the S. aureus ribosome in complex with mRNA, tRNA, EF-G and FA to 2.5 Å resolution and the corresponding complex structures with the recently developed FA derivative FA-cyclopentane (FA-CP) to 2.0 Å resolution. With both FA variants, the majority of the ribosomal particles are observed in chimeric state and only a minor population in post-translocational state. As expected, FA binds in a pocket between domains I, II and III of EF-G and the sarcin-ricin loop of 23S rRNA. FA-CP binds in an identical position, but its cyclopentane moiety provides additional contacts to EF-G and 23S rRNA, suggesting that its improved resistance profile towards mutations in EF-G is due to higher-affinity binding. These high-resolution structures reveal new details about the S. aureus ribosome, including confirmation of many rRNA modifications, and provide an optimal starting point for future structure-based drug discovery on an important clinical drug target.
Collapse
Affiliation(s)
- Adrián González-López
- Department of Cell and Molecular Biology, Uppsala University, BMC, P.O. Box 596, 75124, Uppsala, Sweden
| | - Daniel S D Larsson
- Department of Cell and Molecular Biology, Uppsala University, BMC, P.O. Box 596, 75124, Uppsala, Sweden
| | - Ravi Kiran Koripella
- Department of Cell and Molecular Biology, Uppsala University, BMC, P.O. Box 596, 75124, Uppsala, Sweden
- Robert P. Apkarian Integrated Electron Microscopy Core, Emory University, Atlanta, USA
| | - Brett N Cain
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Martin Garcia Chavez
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Paul J Hergenrother
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Suparna Sanyal
- Department of Cell and Molecular Biology, Uppsala University, BMC, P.O. Box 596, 75124, Uppsala, Sweden
| | - Maria Selmer
- Department of Cell and Molecular Biology, Uppsala University, BMC, P.O. Box 596, 75124, Uppsala, Sweden.
| |
Collapse
|
3
|
Sang M, Feng P, Chi LP, Zhang W. The biosynthetic logic and enzymatic machinery of approved fungi-derived pharmaceuticals and agricultural biopesticides. Nat Prod Rep 2024; 41:565-603. [PMID: 37990930 DOI: 10.1039/d3np00040k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2023]
Abstract
Covering: 2000 to 2023The kingdom Fungi has become a remarkably valuable source of structurally complex natural products (NPs) with diverse bioactivities. Since the revolutionary discovery and application of the antibiotic penicillin from Penicillium, a number of fungi-derived NPs have been developed and approved into pharmaceuticals and pesticide agents using traditional "activity-guided" approaches. Although emerging genome mining algorithms and surrogate expression hosts have brought revolutionary approaches to NP discovery, the time and costs involved in developing these into new drugs can still be prohibitively high. Therefore, it is essential to maximize the utility of existing drugs by rational design and systematic production of new chemical structures based on these drugs by synthetic biology. To this purpose, there have been great advances in characterizing the diversified biosynthetic gene clusters associated with the well-known drugs and in understanding the biosynthesis logic mechanisms and enzymatic transformation processes involved in their production. We describe advances made in the heterogeneous reconstruction of complex NP scaffolds using fungal polyketide synthases (PKSs), non-ribosomal peptide synthetases (NRPSs), PKS/NRPS hybrids, terpenoids, and indole alkaloids and also discuss mechanistic insights into metabolic engineering, pathway reprogramming, and cell factory development. Moreover, we suggest pathways for expanding access to the fungal chemical repertoire by biosynthesis of representative family members via common platform intermediates and through the rational manipulation of natural biosynthetic machineries for drug discovery.
Collapse
Affiliation(s)
- Moli Sang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, Shandong 266237, China.
| | - Peiyuan Feng
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, Shandong 266237, China.
| | - Lu-Ping Chi
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, Shandong 266237, China.
| | - Wei Zhang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, Shandong 266237, China.
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, Shandong 266071, China
| |
Collapse
|
4
|
Höger B, Peifer C, Beitz E. Cell-free production of fluorescent proteins for the discovery of novel ribosome-targeting antibiotics. J Microbiol Methods 2023; 213:106814. [PMID: 37652138 DOI: 10.1016/j.mimet.2023.106814] [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: 03/24/2023] [Revised: 08/14/2023] [Accepted: 08/29/2023] [Indexed: 09/02/2023]
Abstract
Various issues including the overuse of antibiotics has led to the development of threatening multidrug-resistant bacterial strains urging development of novel anti-infectives. One quarter of current clinical phase III antibiotic drug candidates address ribosomal protein translation as a target. Here, we describe an effective cell-free in vitro screening system for inhibitors of bacterial ribosome activity with direct fluorescence read-out. Using ribosomal S30 extracts from Escherichia coli, Salmonella enterica, and Pseudomonas putida, the validity of this system is demonstrated by concentration-dependent inhibition of translation by a set of different classes of translation-targeting drugs. The single-compartment cell-free translation reaction is compatible with multi-well formats. Fluorophore formation of green fluorescent protein or monomeric NeonGreen occurs in an hour time frame without the need of adding reagents for secondary enzymatic detection saving handling time, and prohibiting false positives. As label-free readout, the dose response further allows for IC50 determination in the same setup. Together, we show that cell-free production of fluorescent proteins for the discovery of ribosome-targeting antibiotics is feasible and amenable to high-throughput applications.
Collapse
Affiliation(s)
- Bastian Höger
- Department of Pharmaceutical and Medicinal Chemistry, Christian-Albrechts-University of Kiel, Gutenbergstraße 76, Kiel, Germany
| | - Christian Peifer
- Department of Pharmaceutical and Medicinal Chemistry, Christian-Albrechts-University of Kiel, Gutenbergstraße 76, Kiel, Germany
| | - Eric Beitz
- Department of Pharmaceutical and Medicinal Chemistry, Christian-Albrechts-University of Kiel, Gutenbergstraße 76, Kiel, Germany.
| |
Collapse
|
5
|
Douglas EJ, Laabei M. Staph wars: the antibiotic pipeline strikes back. MICROBIOLOGY (READING, ENGLAND) 2023; 169:001387. [PMID: 37656158 PMCID: PMC10569064 DOI: 10.1099/mic.0.001387] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 08/14/2023] [Indexed: 09/02/2023]
Abstract
Antibiotic chemotherapy is widely regarded as one of the most significant medical advancements in history. However, the continued misuse of antibiotics has contributed to the rapid rise of antimicrobial resistance (AMR) globally. Staphylococcus aureus, a major human pathogen, has become synonymous with multidrug resistance and is a leading antimicrobial-resistant pathogen causing significant morbidity and mortality worldwide. This review focuses on (1) the targets of current anti-staphylococcal antibiotics and the specific mechanisms that confirm resistance; (2) an in-depth analysis of recently licensed antibiotics approved for the treatment of S. aureus infections; and (3) an examination of the pre-clinical pipeline of anti-staphylococcal compounds. In addition, we examine the molecular mechanism of action of novel antimicrobials and derivatives of existing classes of antibiotics, collate data on the emergence of resistance to new compounds and provide an overview of key data from clinical trials evaluating anti-staphylococcal compounds. We present several successful cases in the development of alternative forms of existing antibiotics that have activity against multidrug-resistant S. aureus. Pre-clinical antimicrobials show promise, but more focus and funding are required to develop novel classes of compounds that can curtail the spread of and sustainably control antimicrobial-resistant S. aureus infections.
Collapse
Affiliation(s)
| | - Maisem Laabei
- Department of Life Sciences, University of Bath, Bath BA2 7AY, UK
| |
Collapse
|
6
|
Salimova EV, Mozgovoj OS, Efimova SS, Ostroumova OS, Parfenova LV. 3-Amino-Substituted Analogues of Fusidic Acid as Membrane-Active Antibacterial Compounds. MEMBRANES 2023; 13:309. [PMID: 36984696 PMCID: PMC10056636 DOI: 10.3390/membranes13030309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/27/2023] [Accepted: 03/03/2023] [Indexed: 06/18/2023]
Abstract
Fusidic acid (FA) is an antibiotic with high activity against Staphylococcus aureus; it has been used in clinical practice since the 1960s. However, the narrow antimicrobial spectrum of FA limits its application in the treatment of bacterial infections. In this regard, this work aims both at the study of the antimicrobial effect of a number of FA amines and at the identification of their potential biological targets. In this way, FA analogues containing aliphatic and aromatic amino groups and biogenic polyamine, spermine and spermidine, moieties at the C-3 atom, were synthesized (20 examples). Pyrazinecarboxamide-substituted analogues exhibit a high antibacterial activity against S. aureus (MRSA) with MIC ≤ 0.25 μg/mL. Spermine and spermidine derivatives, along with activity against S. aureus, also inhibit the growth and reproduction of Gram-negative bacteria Escherichia coli, Acinetobacter baumannii, and Pseudomonas aeruginosa, and have a high fungicidal effect against Candida albicans and Cryptococcus neoformans. The study of the membrane activity demonstrated that the spermidine- and spermine-containing compounds are able to immerse into membranes and disorder the lipidsleading to a detergent effect. Moreover, spermine-based compounds are also able to form ion-permeable pores in the lipid bilayers mimicking the bacterial membranes. Using molecular docking, inhibition of the protein synthesis elongation factor EF-G was proposed, and polyamine substituents were shown to make the greatest contribution to the stability of the complexes of fusidic acid derivatives with biological targets. This suggests that the antibacterial effect of the obtained compounds may be associated with both membrane activity and inhibition of the elongation factor EF-G.
Collapse
Affiliation(s)
- Elena V. Salimova
- Institute of Petrochemistry and Catalysis, Ufa Federal Research Center, Russian Academy of Sciences, 141 Prospect Oktyabrya, 450075 Ufa, Russia
| | - Oleg S. Mozgovoj
- Institute of Petrochemistry and Catalysis, Ufa Federal Research Center, Russian Academy of Sciences, 141 Prospect Oktyabrya, 450075 Ufa, Russia
| | - Svetlana S. Efimova
- Institute of Cytology of Russian Academy of Sciences, 4 Tikhoretsky Prospect, 194064 Saint Petersburg, Russia
| | - Olga S. Ostroumova
- Institute of Cytology of Russian Academy of Sciences, 4 Tikhoretsky Prospect, 194064 Saint Petersburg, Russia
| | - Lyudmila V. Parfenova
- Institute of Petrochemistry and Catalysis, Ufa Federal Research Center, Russian Academy of Sciences, 141 Prospect Oktyabrya, 450075 Ufa, Russia
| |
Collapse
|
7
|
Zheng W, Tu B, Zhang Z, Li J, Yan Z, Su K, Deng D, Sun Y, Wang X, Zhang B, Zhang K, Wong WL, Wu P, Hong WD, Ang S. Ligand and structure-based approaches for the exploration of structure-activity relationships of fusidic acid derivatives as antibacterial agents. Front Chem 2023; 10:1094841. [PMID: 36688047 PMCID: PMC9852990 DOI: 10.3389/fchem.2022.1094841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 12/19/2022] [Indexed: 01/09/2023] Open
Abstract
Introduction: Fusidic acid (FA) has been widely applied in the clinical prevention and treatment of bacterial infections. Nonetheless, its clinical application has been limited due to its narrow antimicrobial spectrum and some side effects. Purpose: Therefore, it is necessary to explore the structure-activity relationships of FA derivatives as antibacterial agents to develop novel ones possessing a broad antimicrobial spectrum. Methods and result: First, a pharmacophore model was established on the nineteen FA derivatives with remarkable antibacterial activities reported in previous studies. The common structural characteristics of the pharmacophore emerging from the FA derivatives were determined as those of six hydrophobic centers, two atom centers of the hydrogen bond acceptor, and a negative electron center around the C-21 field. Then, seven FA derivatives have been designed according to the reported structure-activity relationships and the pharmacophore characteristics. The designed FA derivatives were mapped on the pharmacophore model, and the Qfit values of all FA derivatives were over 50 and FA-8 possessed the highest value of 82.66. The molecular docking studies of the partial target compounds were conducted with the elongation factor G (EF-G) of S. aureus. Furthermore, the designed FA derivatives have been prepared and their antibacterial activities were evaluated by the inhibition zone test and the minimum inhibitory concentration (MIC) test. The derivative FA-7 with a chlorine group as the substituent group at C-25 of FA displayed the best antibacterial property with an MIC of 3.125 µM. Subsequently, 3D-QSAR was carried on all the derivatives by using the CoMSIA mode of SYBYL-X 2.0. Conclusion: Hence, a computer-aided drug design model was developed for FA, which can be further used to optimize FA derivatives as highly potent antibacterial agents.
Collapse
Affiliation(s)
- Wende Zheng
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China,International Healthcare Innovation Institute, Jiangmen, China
| | - Borong Tu
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China,International Healthcare Innovation Institute, Jiangmen, China
| | - Zhen Zhang
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China,International Healthcare Innovation Institute, Jiangmen, China
| | - Jinxuan Li
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China,International Healthcare Innovation Institute, Jiangmen, China
| | - Zhenping Yan
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China,International Healthcare Innovation Institute, Jiangmen, China
| | - Kaize Su
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China,International Healthcare Innovation Institute, Jiangmen, China
| | - Duanyu Deng
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China,International Healthcare Innovation Institute, Jiangmen, China
| | - Ying Sun
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China,International Healthcare Innovation Institute, Jiangmen, China
| | - Xu Wang
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China,International Healthcare Innovation Institute, Jiangmen, China
| | - Bingjie Zhang
- School of Biomedicine and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China
| | - Kun Zhang
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China,International Healthcare Innovation Institute, Jiangmen, China
| | - Wing-Leung Wong
- The State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, China
| | - Panpan Wu
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China,International Healthcare Innovation Institute, Jiangmen, China,*Correspondence: Panpan Wu, ; Weiqian David Hong, ; Song Ang,
| | - Weiqian David Hong
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China,International Healthcare Innovation Institute, Jiangmen, China,*Correspondence: Panpan Wu, ; Weiqian David Hong, ; Song Ang,
| | - Song Ang
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China,International Healthcare Innovation Institute, Jiangmen, China,*Correspondence: Panpan Wu, ; Weiqian David Hong, ; Song Ang,
| |
Collapse
|
8
|
Screening and Molecular Docking of Bioactive Metabolites of the Red Sea Sponge Callyspongia siphonella as Potential Antimicrobial Agents. Antibiotics (Basel) 2022; 11:antibiotics11121682. [PMID: 36551340 PMCID: PMC9774121 DOI: 10.3390/antibiotics11121682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/20/2022] [Accepted: 11/21/2022] [Indexed: 11/24/2022] Open
Abstract
Marine sponges create a wide range of bioactive secondary metabolites, as documented throughout the year. Several bioactive secondary metabolites were isolated from different members of Callyspongia siphonella species. This study aimed for isolation and structural elucidation of major metabolites in order to investigate their diverse bioactivities such as antimicrobial and anti-biofilm activities. Afterwards, a molecular docking study was conducted, searching for the possible mechanistic pathway of the most bioactive metabolites. Extraction, fractionation, and metabolomics analysis of different fractions was performed in order to obtain complete chemical profile. Moreover, in vitro assessment of different bioactivities was performed, using recent techniques. Additionally, purification, structural elucidation of high features using recent chromatographic and spectroscopic techniques was established. Finally, AutoDock Vina software was used for the Pharmacophore-based docking-based analysis. As a result, DCM (dichloromethane) fraction exerted the best antibacterial activity using disc diffusion method; particularly against S. aureus with an inhibition zone of 6.6 mm. Compound 11 displayed a considerable activity against both MRSA (Methicillin-resistant Staphyllococcus aureus) and Staphyllococcus aureus with inhibition ratios of 50.37 and 60.90%, respectively. Concerning anti-biofilm activity, compounds 1 and 2 displayed powerful activity with inhibition ratios ranging from 39.37% to 70.98%. Pharmacophore-based docking-based analysis suggested elongation factor G (EF-G) to be a probable target for compound 11 (siphonellinol C) that showed the best in vitro antibacterial activity, offering unexplored potential for new drugs and treatment candidates.
Collapse
|
9
|
Lade H, Joo HS, Kim JS. Molecular Basis of Non-β-Lactam Antibiotics Resistance in Staphylococcus aureus. Antibiotics (Basel) 2022; 11:1378. [PMID: 36290036 PMCID: PMC9598170 DOI: 10.3390/antibiotics11101378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 10/04/2022] [Accepted: 10/06/2022] [Indexed: 11/22/2022] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is one of the most successful human pathogens with the potential to cause significant morbidity and mortality. MRSA has acquired resistance to almost all β-lactam antibiotics, including the new-generation cephalosporins, and is often also resistant to multiple other antibiotic classes. The expression of penicillin-binding protein 2a (PBP2a) is the primary basis for β-lactams resistance by MRSA, but it is coupled with other resistance mechanisms, conferring resistance to non-β-lactam antibiotics. The multiplicity of resistance mechanisms includes target modification, enzymatic drug inactivation, and decreased antibiotic uptake or efflux. This review highlights the molecular basis of resistance to non-β-lactam antibiotics recommended to treat MRSA infections such as macrolides, lincosamides, aminoglycosides, glycopeptides, oxazolidinones, lipopeptides, and others. A thorough understanding of the molecular and biochemical basis of antibiotic resistance in clinical isolates could help in developing promising therapies and molecular detection methods of antibiotic resistance.
Collapse
Affiliation(s)
- Harshad Lade
- Department of Laboratory Medicine, Hallym University College of Medicine, Kangdong Sacred Heart Hospital, Seoul 05355, Korea
| | - Hwang-Soo Joo
- Department of Biotechnology, College of Engineering, Duksung Women’s University, Seoul 01369, Korea
| | - Jae-Seok Kim
- Department of Laboratory Medicine, Hallym University College of Medicine, Kangdong Sacred Heart Hospital, Seoul 05355, Korea
| |
Collapse
|
10
|
Tu B, Cao N, Zhang B, Zheng W, Li J, Tang X, Su K, Li J, Zhang Z, Yan Z, Li D, Zheng X, Zhang K, Hong WD, Wu P. Synthesis and Biological Evaluation of Novel Fusidic Acid Derivatives as Two-in-One Agent with Potent Antibacterial and Anti-Inflammatory Activity. Antibiotics (Basel) 2022; 11:antibiotics11081026. [PMID: 36009895 PMCID: PMC9405029 DOI: 10.3390/antibiotics11081026] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 07/24/2022] [Accepted: 07/28/2022] [Indexed: 12/10/2022] Open
Abstract
Fusidic acid (FA), a narrow-spectrum antibiotics, is highly sensitive to various Gram-positive cocci associated with skin infections. It has outstanding antibacterial effects against certain Gram-positive bacteria whilst no cross-resistance with other antibiotics. Two series of FA derivatives were synthesized and their antibacterial activities were tested. A new aromatic side-chain analog, FA-15 exhibited good antibacterial activity with MIC values in the range of 0.781–1.563 µM against three strains of Staphylococcus spp. Furthermore, through the assessment by the kinetic assay, similar characteristics of bacteriostasis by FA and its aromatic derivatives were observed. In addition, anti-inflammatory activities of FA and its aromatic derivatives were evaluated by using a 12-O-tetradecanoylphorbol-13-acetate (TPA) induced mouse ear edema model. The results also indicated that FA and its aromatic derivatives effectively reduced TPA-induced ear edema in a dose-dependent manner. Following, multiform computerized simulation, including homology modeling, molecular docking, molecular dynamic simulation and QSAR was conducted to clarify the mechanism and regularity of activities. Overall, the present work gave vital clues about structural modifications and has profound significance in deeply scouting for bioactive potentials of FA and its derivatives.
Collapse
Affiliation(s)
- Borong Tu
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China; (B.T.); (N.C.); (W.Z.); (J.L.); (X.T.); (K.S.); (J.L.); (Z.Z.); (Z.Y.); (D.L.); (X.Z.)
- International Healthcare Innovation Institute (Jiangmen), Jiangmen 529040, China
| | - Nana Cao
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China; (B.T.); (N.C.); (W.Z.); (J.L.); (X.T.); (K.S.); (J.L.); (Z.Z.); (Z.Y.); (D.L.); (X.Z.)
- International Healthcare Innovation Institute (Jiangmen), Jiangmen 529040, China
| | - Bingjie Zhang
- School of Biomedicine and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China;
| | - Wende Zheng
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China; (B.T.); (N.C.); (W.Z.); (J.L.); (X.T.); (K.S.); (J.L.); (Z.Z.); (Z.Y.); (D.L.); (X.Z.)
- International Healthcare Innovation Institute (Jiangmen), Jiangmen 529040, China
| | - Jiahao Li
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China; (B.T.); (N.C.); (W.Z.); (J.L.); (X.T.); (K.S.); (J.L.); (Z.Z.); (Z.Y.); (D.L.); (X.Z.)
- International Healthcare Innovation Institute (Jiangmen), Jiangmen 529040, China
| | - Xiaowen Tang
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China; (B.T.); (N.C.); (W.Z.); (J.L.); (X.T.); (K.S.); (J.L.); (Z.Z.); (Z.Y.); (D.L.); (X.Z.)
- International Healthcare Innovation Institute (Jiangmen), Jiangmen 529040, China
| | - Kaize Su
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China; (B.T.); (N.C.); (W.Z.); (J.L.); (X.T.); (K.S.); (J.L.); (Z.Z.); (Z.Y.); (D.L.); (X.Z.)
- International Healthcare Innovation Institute (Jiangmen), Jiangmen 529040, China
| | - Jinxuan Li
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China; (B.T.); (N.C.); (W.Z.); (J.L.); (X.T.); (K.S.); (J.L.); (Z.Z.); (Z.Y.); (D.L.); (X.Z.)
- International Healthcare Innovation Institute (Jiangmen), Jiangmen 529040, China
| | - Zhen Zhang
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China; (B.T.); (N.C.); (W.Z.); (J.L.); (X.T.); (K.S.); (J.L.); (Z.Z.); (Z.Y.); (D.L.); (X.Z.)
- International Healthcare Innovation Institute (Jiangmen), Jiangmen 529040, China
| | - Zhenping Yan
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China; (B.T.); (N.C.); (W.Z.); (J.L.); (X.T.); (K.S.); (J.L.); (Z.Z.); (Z.Y.); (D.L.); (X.Z.)
- International Healthcare Innovation Institute (Jiangmen), Jiangmen 529040, China
| | - Dongli Li
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China; (B.T.); (N.C.); (W.Z.); (J.L.); (X.T.); (K.S.); (J.L.); (Z.Z.); (Z.Y.); (D.L.); (X.Z.)
- International Healthcare Innovation Institute (Jiangmen), Jiangmen 529040, China
| | - Xi Zheng
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China; (B.T.); (N.C.); (W.Z.); (J.L.); (X.T.); (K.S.); (J.L.); (Z.Z.); (Z.Y.); (D.L.); (X.Z.)
- International Healthcare Innovation Institute (Jiangmen), Jiangmen 529040, China
| | - Kun Zhang
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China; (B.T.); (N.C.); (W.Z.); (J.L.); (X.T.); (K.S.); (J.L.); (Z.Z.); (Z.Y.); (D.L.); (X.Z.)
- International Healthcare Innovation Institute (Jiangmen), Jiangmen 529040, China
- School of Biomedicine and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China;
- Correspondence: (K.Z.); (W.D.H.); (P.W.); Tel.: +86-13822330019 (K.Z.); +44-7863354263 (W.D.H.); +86-18825179347 (P.W.)
| | - Weiqian David Hong
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China; (B.T.); (N.C.); (W.Z.); (J.L.); (X.T.); (K.S.); (J.L.); (Z.Z.); (Z.Y.); (D.L.); (X.Z.)
- International Healthcare Innovation Institute (Jiangmen), Jiangmen 529040, China
- Department of Chemistry, University of Liverpool, Liverpool L69 7ZD, UK
- Correspondence: (K.Z.); (W.D.H.); (P.W.); Tel.: +86-13822330019 (K.Z.); +44-7863354263 (W.D.H.); +86-18825179347 (P.W.)
| | - Panpan Wu
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China; (B.T.); (N.C.); (W.Z.); (J.L.); (X.T.); (K.S.); (J.L.); (Z.Z.); (Z.Y.); (D.L.); (X.Z.)
- International Healthcare Innovation Institute (Jiangmen), Jiangmen 529040, China
- School of Biomedicine and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China;
- Correspondence: (K.Z.); (W.D.H.); (P.W.); Tel.: +86-13822330019 (K.Z.); +44-7863354263 (W.D.H.); +86-18825179347 (P.W.)
| |
Collapse
|
11
|
Wieland M, Holm M, Rundlet EJ, Morici M, Koller TO, Maviza TP, Pogorevc D, Osterman IA, Müller R, Blanchard SC, Wilson DN. The cyclic octapeptide antibiotic argyrin B inhibits translation by trapping EF-G on the ribosome during translocation. Proc Natl Acad Sci U S A 2022; 119:e2114214119. [PMID: 35500116 PMCID: PMC9171646 DOI: 10.1073/pnas.2114214119] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 03/16/2022] [Indexed: 11/18/2022] Open
Abstract
Argyrins are a family of naturally produced octapeptides that display promising antimicrobial activity against Pseudomonas aeruginosa. Argyrin B (ArgB) has been shown to interact with an elongated form of the translation elongation factor G (EF-G), leading to the suggestion that argyrins inhibit protein synthesis by interfering with EF-G binding to the ribosome. Here, using a combination of cryo-electron microscopy (cryo-EM) and single-molecule fluorescence resonance energy transfer (smFRET), we demonstrate that rather than interfering with ribosome binding, ArgB rapidly and specifically binds EF-G on the ribosome to inhibit intermediate steps of the translocation mechanism. Our data support that ArgB inhibits conformational changes within EF-G after GTP hydrolysis required for translocation and factor dissociation, analogous to the mechanism of fusidic acid, a chemically distinct antibiotic that binds a different region of EF-G. These findings shed light on the mechanism of action of the argyrin-class antibiotics on protein synthesis as well as the nature and importance of rate-limiting, intramolecular conformational events within the EF-G-bound ribosome during late-steps of translocation.
Collapse
Affiliation(s)
- Maximiliane Wieland
- Institute for Biochemistry and Molecular Biology, University of Hamburg, 20146 Hamburg, Germany
| | - Mikael Holm
- St. Jude Children's Research Hospital, Memphis, TN 38105
| | - Emily J. Rundlet
- St. Jude Children's Research Hospital, Memphis, TN 38105
- Weill Cornell Medicine, Tri-Institutional PhD Program in Chemical Biology, New York, NY 10065
| | - Martino Morici
- Institute for Biochemistry and Molecular Biology, University of Hamburg, 20146 Hamburg, Germany
| | - Timm O. Koller
- Institute for Biochemistry and Molecular Biology, University of Hamburg, 20146 Hamburg, Germany
| | - Tinashe P. Maviza
- Center of Life Sciences, Skolkovo Institute of Science and Technology, 121205 Moscow, Russia
| | - Domen Pogorevc
- Department Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Centre for Infection Research, Saarland University, 66123 Saarbrücken,Germany
| | - Ilya A. Osterman
- Center of Life Sciences, Skolkovo Institute of Science and Technology, 121205 Moscow, Russia
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Rolf Müller
- Department Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Centre for Infection Research, Saarland University, 66123 Saarbrücken,Germany
| | | | - Daniel N. Wilson
- Institute for Biochemistry and Molecular Biology, University of Hamburg, 20146 Hamburg, Germany
| |
Collapse
|
12
|
Boumasmoud M, Dengler Haunreiter V, Schweizer TA, Meyer L, Chakrakodi B, Schreiber PW, Seidl K, Kühnert D, Kouyos RD, Zinkernagel AS. Genomic Surveillance of Vancomycin-Resistant Enterococcus faecium Reveals Spread of a Linear Plasmid Conferring a Nutrient Utilization Advantage. mBio 2022; 13:e0377121. [PMID: 35343787 PMCID: PMC9040824 DOI: 10.1128/mbio.03771-21] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 02/25/2022] [Indexed: 12/12/2022] Open
Abstract
Healthcare-associated outbreaks of vancomycin-resistant Enterococcus faecium (VREfm) are a worldwide problem with increasing prevalence. The genomic plasticity of this hospital-adapted pathogen contributes to its efficient spread despite infection control measures. Here, we aimed to identify the genomic and phenotypic determinants of health care-associated transmission of VREfm. We assessed the VREfm transmission networks at the tertiary-care University Hospital of Zurich (USZ) between October 2014 and February 2018 and investigated microevolutionary dynamics of this pathogen. We performed whole-genome sequencing for the 69 VREfm isolates collected during this time frame and assessed the population structure and variability of the vancomycin resistance transposon. Phylogenomic analysis allowed us to reconstruct transmission networks and to unveil external or wider transmission networks undetectable by routine surveillance. Notably, it unveiled a persistent clone, sampled 31 times over a 29-month period. Exploring the evolutionary dynamics of this clone and characterizing the phenotypic consequences revealed the spread of a variant with decreased daptomycin susceptibility and the acquired ability to utilize N-acetyl-galactosamine (GalNAc), one of the primary constituents of the human gut mucins. This nutrient utilization advantage was conferred by a novel plasmid, termed pELF_USZ, which exhibited a linear topology. This plasmid, which was harbored by two distinct clones, was transferable by conjugation. Overall, this work highlights the potential of combining epidemiological, functional genomic, and evolutionary perspectives to unveil adaptation strategies of VREfm. IMPORTANCE Sequencing microbial pathogens causing outbreaks has become a common practice to characterize transmission networks. In addition to the signal provided by vertical evolution, bacterial genomes harbor mobile genetic elements shared horizontally between clones. While macroevolutionary studies have revealed an important role of plasmids and genes encoding carbohydrate utilization systems in the adaptation of Enterococcus faecium to the hospital environment, mechanisms of dissemination and the specific function of many of these genetic determinants remain to be elucidated. Here, we characterize a plasmid providing a nutrient utilization advantage and show evidence for its clonal and horizontal spread at a local scale. Further studies integrating epidemiological, functional genomics, and evolutionary perspectives will be critical to identify changes shaping the success of this pathogen.
Collapse
Affiliation(s)
- Mathilde Boumasmoud
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Vanina Dengler Haunreiter
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Tiziano A. Schweizer
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Lilly Meyer
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Bhavya Chakrakodi
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Peter W. Schreiber
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Kati Seidl
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Denise Kühnert
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Roger D. Kouyos
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Annelies S. Zinkernagel
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
13
|
TIAN C, WANG K, ZHANG X, LI G, LOU HX. Old fusidane-type antibiotics for new challenges: Chemistry and biology. Chin J Nat Med 2022; 20:81-101. [DOI: 10.1016/s1875-5364(21)60114-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Indexed: 12/24/2022]
|
14
|
Long J, Ji W, Zhang D, Zhu Y, Bi Y. Bioactivities and Structure-Activity Relationships of Fusidic Acid Derivatives: A Review. Front Pharmacol 2021; 12:759220. [PMID: 34721042 PMCID: PMC8554340 DOI: 10.3389/fphar.2021.759220] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 09/27/2021] [Indexed: 12/22/2022] Open
Abstract
Fusidic acid (FA) is a natural tetracyclic triterpene isolated from fungi, which is clinically used for systemic and local staphylococcal infections, including methicillin-resistant Staphylococcus aureus and coagulase-negative staphylococci infections. FA and its derivatives have been shown to possess a wide range of pharmacological activities, including antibacterial, antimalarial, antituberculosis, anticancer, tumor multidrug resistance reversal, anti-inflammation, antifungal, and antiviral activity in vivo and in vitro. The semisynthesis, structural modification and biological activities of FA derivatives have been extensively studied in recent years. This review summarized the biological activities and structure-activity relationship (SAR) of FA in the last two decades. This summary can prove useful information for drug exploration of FA derivatives.
Collapse
Affiliation(s)
- Junjun Long
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Wentao Ji
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Doudou Zhang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Yifei Zhu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Yi Bi
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| |
Collapse
|
15
|
Song X, Lv J, Cao Z, Huang H, Chen G, Awakawa T, Hu D, Gao H, Abe I, Yao X. Extensive expansion of the chemical diversity of fusidane-type antibiotics using a stochastic combinational strategy. Acta Pharm Sin B 2021; 11:1676-1685. [PMID: 34221876 PMCID: PMC8245791 DOI: 10.1016/j.apsb.2020.12.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 11/11/2020] [Accepted: 12/04/2020] [Indexed: 12/12/2022] Open
Abstract
Fusidane-type antibiotics, represented by helvolic acid, fusidic acid and cephalosporin P1, are fungi-derived antimicrobials with little cross-resistance to commonly used antibiotics. Generation of new fusidane-type derivatives is therefore of great value, but this is hindered by available approaches. Here, we developed a stochastic combinational strategy by random assembly of all the post-tailoring genes derived from helvolic acid, fusidic acid, and cephalosporin P1 biosynthetic pathways in a strain that produces their common intermediate. Among a total of 27 gene combinations, 24 combinations produce expected products and afford 58 fusidane-type analogues, of which 54 are new compounds. Moreover, random gene combination can induce unexpected activity of some post-tailoring enzymes, leading to a further increase in chemical diversity. These newly generated derivatives provide new insights into the structure‒activity relationship of fusidane-type antibiotics. The stochastic combinational strategy established in this study proves to be a powerful approach for expanding structural diversity of natural products.
Collapse
|
16
|
Protein Expression Level Changes in Weissella koreensis during Garlic Media Fermentation. BIOLOGY 2021; 10:biology10060478. [PMID: 34071410 PMCID: PMC8226471 DOI: 10.3390/biology10060478] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/25/2021] [Accepted: 05/26/2021] [Indexed: 12/05/2022]
Abstract
Simple Summary Garlic is used in cooking and is known to have antibacterial properties due to various organic sulfur components. Weissella koreensis is one of the lactic acid bacteria used for fermentation with garlic and is a strain found in kimchi in Korea, one of foods to which garlic is added. In this study, the addition of garlic to the medium did not cause a difference in growth of W. koreensis. However, the addition of garlic to the growth medium did produce changes in the amount of specific W. koreensis proteins. This study provides basic information about the effect of garlic on fermentation using W. koreensis. Abstract This study investigated the changes in Weissella koreensis (WK) protein expression levels during fermentation in MRS medium supplemented with garlic of WK. WK was first discovered as lactic acid bacteria (LAB) in a Korean fermented cabbage dish known as kimchi. The number of WK cells in MRS medium with garlic (MBCG) and without (MB) after 7 days was 3.55 × 1010 and 2.55 × 1010 CFU/mL, respectively. To observe the changes in the carbon sources in the media, we measured the glucose, sucrose, lactic acid, and acetic acid levels in each medium (MB and MBCG). Thus, 67.2 ± 2.4 (MB) and 64.2 ± 4.7 (MBCG) mmol−1 of glucose were consumed. For sucrose, the level was 3.5 ± 2.2 (MB), and 3.4 ± 2.5 (MBCG) mmol−1. There was not much difference in the lactic acid and acetic acid levels at 160.8 ± 0.4 (MB) and 159.2 ± 0.2 (MBCG) and 2.4 ± 0.4 (MB) and 2.2 ± 8.1 (MBCG) mmol−1, respectively. After the 7-day fermentation period, two-dimensional electrophoresis (2DE) was used to confirm the protein expression pattern in the WK strain. The results show that the fusA and ssb1 proteins were reduced, and the clpP protein was increased. Afterwards, the expression patterns of the above proteins were confirmed through qRT-PCR. Thus, this study confirms the changes in protein expression levels in Weissella koreensis when garlic was added to the media. This study provides basic data for future studies on the major biosynthetic pathways of WK.
Collapse
|
17
|
Lee M, Matsunaga N, Akabane S, Yasuda I, Ueda T, Takeuchi-Tomita N. Reconstitution of mammalian mitochondrial translation system capable of correct initiation and long polypeptide synthesis from leaderless mRNA. Nucleic Acids Res 2021; 49:371-382. [PMID: 33300043 PMCID: PMC7797035 DOI: 10.1093/nar/gkaa1165] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 11/12/2020] [Accepted: 11/16/2020] [Indexed: 12/31/2022] Open
Abstract
Mammalian mitochondria have their own dedicated protein synthesis system, which produces 13 essential subunits of the oxidative phosphorylation complexes. We have reconstituted an in vitro translation system from mammalian mitochondria, utilizing purified recombinant mitochondrial translation factors, 55S ribosomes from pig liver mitochondria, and a tRNA mixture from either Escherichia coli or yeast. The system is capable of translating leaderless mRNAs encoding model proteins (DHFR and nanoLuciferase) or some mtDNA-encoded proteins. We show that a leaderless mRNA, encoding nanoLuciferase, is faithfully initiated without the need for any auxiliary factors other than IF-2mt and IF-3mt. We found that the ribosome-dependent GTPase activities of both the translocase EF-G1mt and the recycling factor EF-G2mt are insensitive to fusidic acid (FA), the translation inhibitor that targets bacterial EF-G homologs, and consequently the system is resistant to FA. Moreover, we demonstrate that a polyproline sequence in the protein causes 55S mitochondrial ribosome stalling, yielding ribosome nascent chain complexes. Analyses of the effects of the Mg concentration on the polyproline-mediated ribosome stalling suggested the unique regulation of peptide elongation by the mitoribosome. This system will be useful for analyzing the mechanism of translation initiation, and the interactions between the nascent peptide chain and the mitochondrial ribosome.
Collapse
Affiliation(s)
- Muhoon Lee
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5, Kashiwanoha, Kashiwa-shi, Chiba 277-8562, Japan
| | - Noriko Matsunaga
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5, Kashiwanoha, Kashiwa-shi, Chiba 277-8562, Japan
| | - Shiori Akabane
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5, Kashiwanoha, Kashiwa-shi, Chiba 277-8562, Japan.,Department of Life Science, Rikkyo University, Tokyo, 171-8501, Japan
| | - Ippei Yasuda
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5, Kashiwanoha, Kashiwa-shi, Chiba 277-8562, Japan
| | - Takuya Ueda
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5, Kashiwanoha, Kashiwa-shi, Chiba 277-8562, Japan.,Department of Integrative Bioscience and Biomedical Engineering, Graduate School of Science and Engineering, Waseda University, Tokyo, Shinjuku 162-8480, Japan
| | - Nono Takeuchi-Tomita
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5, Kashiwanoha, Kashiwa-shi, Chiba 277-8562, Japan
| |
Collapse
|
18
|
Boloki HA, Al-Musaileem WF, AlFouzan W, Verghese T, Udo EE. Fusidic Acid Resistance Determinants in Methicillin-Resistant Staphylococcus aureus Isolated in Kuwait Hospitals. Med Princ Pract 2021; 30:542-549. [PMID: 34348297 PMCID: PMC8740012 DOI: 10.1159/000518408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 07/09/2021] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE The aim of this study was to investigate the genetic determinants of fusidic acid (FA) resistance in MRSA isolated from patients in Kuwait hospitals. METHODS The minimum inhibitory concentration (MIC) of FA was tested with E-test strips. Genetic determinants of FA were determined by PCR and DNA microarray. Staphylococcal protein A gene (spa) typing and DNA microarray analysis were used to study their genetic backgrounds. RESULTS The FA MIC ranged from 2 mg/L to >256 mg/L. Of the 97 isolates, 79 (81.4%) harbored fusC, 14 isolates harbored fusA mutations (fusA), and 4 isolates harbored fusB. Isolates with fusA mutations expressed high FA MIC (MIC >256 mg/L), whereas those with fusC and fusB expressed low FA MIC (MIC 2-16 mg/L). The isolates belonged to 23 spa types and 12 clonal complexes (CCs). The major spa types were t688 (n = 25), t311 (n = 14), t860 (n = 8), and t127 (n = 6) which constituted 54.6% of the isolates. The 12 CCs were CC1, CC5, CC8, CC15, CC22, CC80, CC88, and CC97 with CC5 (45.6%) and CC97 (13.2%) as the dominant CCs. CONCLUSIONS The MRSA isolates belonged to diverse genetic backgrounds with the majority carrying the fusC resistance determinants. The high prevalence of FA resistance belonging to diverse genetic backgrounds warrants a review of FA usage in the country to preserve its therapeutic benefits.
Collapse
|
19
|
Tomlinson JH, Kalverda AP, Calabrese AN. Fusidic acid resistance through changes in the dynamics of the drug target. Proc Natl Acad Sci U S A 2020; 117:25523-25531. [PMID: 32999060 PMCID: PMC7568287 DOI: 10.1073/pnas.2008577117] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Antibiotic resistance in clinically important bacteria can be mediated by target protection mechanisms, whereby a protein binds to the drug target and protects it from the inhibitory effects of the antibiotic. The most prevalent source of clinical resistance to the antibiotic fusidic acid (FA) is expression of the FusB family of proteins that bind to the drug target (Elongation factor G [EF-G]) and promote dissociation of EF-G from FA-stalled ribosome complexes. FusB binding causes changes in both the structure and conformational flexibility of EF-G, but which of these changes drives FA resistance was not understood. We present here detailed characterization of changes in the conformational flexibility of EF-G in response to FusB binding and show that these changes are responsible for conferring FA resistance. Binding of FusB to EF-G causes a significant change in the dynamics of domain III of EF-GC3 that leads to an increase in a minor, more disordered state of EF-G domain III. This is sufficient to overcome the steric block of transmission of conformational changes within EF-G by which FA prevents release of EF-G from the ribosome. This study has identified an antibiotic resistance mechanism mediated by allosteric effects on the dynamics of the drug target.
Collapse
Affiliation(s)
- Jennifer H Tomlinson
- School of Molecular and Cellular Biology, University of Leeds, LS2 9JT Leeds, United Kingdom;
- Astbury Centre for Structural Molecular Biology, Faculty of Biological Sciences, University of Leeds, LS2 9JT Leeds, United Kingdom
| | - Arnout P Kalverda
- School of Molecular and Cellular Biology, University of Leeds, LS2 9JT Leeds, United Kingdom
- Astbury Centre for Structural Molecular Biology, Faculty of Biological Sciences, University of Leeds, LS2 9JT Leeds, United Kingdom
| | - Antonio N Calabrese
- School of Molecular and Cellular Biology, University of Leeds, LS2 9JT Leeds, United Kingdom
- Astbury Centre for Structural Molecular Biology, Faculty of Biological Sciences, University of Leeds, LS2 9JT Leeds, United Kingdom
| |
Collapse
|
20
|
Abstract
Antibiotic resistance is mediated through several distinct mechanisms, most of which are relatively well understood and the clinical importance of which has long been recognized. Until very recently, neither of these statements was readily applicable to the class of resistance mechanism known as target protection, a phenomenon whereby a resistance protein physically associates with an antibiotic target to rescue it from antibiotic-mediated inhibition. In this Review, we summarize recent progress in understanding the nature and importance of target protection. In particular, we describe the molecular basis of the known target protection systems, emphasizing that target protection does not involve a single, uniform mechanism but is instead brought about in several mechanistically distinct ways.
Collapse
|
21
|
Lozano‐Huntelman NA, Singh N, Valencia A, Mira P, Sakayan M, Boucher I, Tang S, Brennan K, Gianvecchio C, Fitz‐Gibbon S, Yeh P. Evolution of antibiotic cross-resistance and collateral sensitivity in Staphylococcus epidermidis using the mutant prevention concentration and the mutant selection window. Evol Appl 2020; 13:808-823. [PMID: 32211069 PMCID: PMC7086048 DOI: 10.1111/eva.12903] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 11/14/2019] [Indexed: 01/09/2023] Open
Abstract
In bacteria, evolution of resistance to one antibiotic is frequently associated with increased resistance (cross-resistance) or increased susceptibility (collateral sensitivity) to other antibiotics. Cross-resistance and collateral sensitivity are typically evaluated at the minimum inhibitory concentration (MIC). However, these susceptibility changes are not well characterized with respect to the mutant prevention concentration (MPC), the antibiotic concentration that prevents a single-step mutation from occurring. We measured the MIC and the MPC for Staphylococcus epidermidis and 14 single-drug resistant strains against seven antibiotics. We found that the MIC and the MPC were positively correlated but that this correlation weakened if cross-resistance did not evolve. If any type of resistance did evolve, the range of concentrations between the MIC and the MPC tended to shift right and widen. Similar patterns of cross-resistance and collateral sensitivity were observed at the MIC and MPC levels, though more symmetry was observed at the MIC level. Whole-genome sequencing revealed mutations in both known-target and nontarget genes. Moving forward, examining both the MIC and the MPC may lead to better predictions of evolutionary trajectories in antibiotic-resistant bacteria.
Collapse
Affiliation(s)
| | - Nina Singh
- Department of Ecology and Evolutionary BiologyUniversity of CaliforniaLos AngelesCAUSA
| | - Alondra Valencia
- Department of Ecology and Evolutionary BiologyUniversity of CaliforniaLos AngelesCAUSA
| | - Portia Mira
- Department of Ecology and Evolutionary BiologyUniversity of CaliforniaLos AngelesCAUSA
| | - Maral Sakayan
- Department of Ecology and Evolutionary BiologyUniversity of CaliforniaLos AngelesCAUSA
| | - Ian Boucher
- Department of Ecology and Evolutionary BiologyUniversity of CaliforniaLos AngelesCAUSA
| | - Sharon Tang
- Department of Ecology and Evolutionary BiologyUniversity of CaliforniaLos AngelesCAUSA
| | - Kelley Brennan
- Department of Ecology and Evolutionary BiologyUniversity of CaliforniaLos AngelesCAUSA
| | - Crystal Gianvecchio
- Department of Ecology and Evolutionary BiologyUniversity of CaliforniaLos AngelesCAUSA
| | - Sorel Fitz‐Gibbon
- Department of Molecular, Cell, Developmental BiologyUniversity of CaliforniaLos AngelesCAUSA
| | - Pamela Yeh
- Department of Ecology and Evolutionary BiologyUniversity of CaliforniaLos AngelesCAUSA
- Santa Fe InstituteSanta FeNMUSA
| |
Collapse
|
22
|
Synthesis, antifungal activity and potential mechanism of fusidic acid derivatives possessing amino-terminal groups. Future Med Chem 2020; 12:763-774. [PMID: 32208979 DOI: 10.4155/fmc-2019-0289] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Aim: Fusidic acid (FA) is a narrow-spectrum bacteriostatic antibiotic. We inadvertently discovered that a FA derivative modified by an amino-terminal group at the 3-OH position, namely 2, inhibited the growth of Cryptococcus neoformans. Methods & results: Multiscale molecular modeling approaches were used to analyze the binding modes of 2 with eEF2. FA derivatives modified at the 3-OH position were designed based on in silico models; seven derivatives possessing different amino-terminal groups were synthesized and tested in vitro for antifungal activity against C. neoformans. Conclusion: Compound 7 had the strongest minimum inhibitory concentration. Two protonated nitrogen atoms of 7 interacted with a negative electrostatic pocket of eEF2 likely explain the superiority of 7-2.
Collapse
|
23
|
Lim YJ, Hyun JE, Hwang CY. Identification of fusidic acid resistance in clinical isolates of Staphylococcus pseudintermedius from dogs in Korea. Vet Dermatol 2020; 31:267-e62. [PMID: 32115810 DOI: 10.1111/vde.12844] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/03/2020] [Indexed: 11/28/2022]
Abstract
BACKGROUND Staphylococcus pseudintermedius is a major bacterial species associated with canine pyoderma and otitis. Fusidic acid is used to treat skin infections caused by Gram-positive bacteria. The incidence of resistance to fusidic acid in S. pseudintermedius has importance in terms of limiting treatment options for bacterial infections. HYPOTHESIS/OBJECTIVES To evaluate the occurrence and mechanisms of fusidic acid resistance in clinical isolates of S. pseudintermedius. ANIMALS Fifty-two S. pseudintermedius isolates were collected from dogs with pyoderma (n = 36) or otitis (n = 16). METHODS AND MATERIALS The disk diffusion method determined that isolates <24 mm were resistant to fusidic acid. Minimum inhibitory concentrations (MICs) were measured by the E-test in those with confirmed resistance to fusidic acid by the disk diffusion method. Phenotypically fusidic acid resistant isolates were subjected to PCR to detect the presence of resistance-related genes (fusA, fusB, fusC and fusD) and fusA was further sequenced to identify point mutations. RESULTS Fourteen of 52 (27%) S. pseudintermedius isolates were resistant to fusidic acid and all of these showed low-level resistance. Among fusidic acid resistant isolates, fusA point mutations were confirmed in 11 isolates and amino acid substitutions were found in five. fusC was detected in seven isolates, but neither fusB nor fusD was detected. CONCLUSIONS AND CLINICAL IMPORTANCE This study demonstrates the occurrence and resistance mechanisms to fusidic acid in clinical isolates of S. pseudintermedius. Continuous monitoring for fusidic acid resistance is recommended.
Collapse
Affiliation(s)
- Yun-Ji Lim
- Laboratory of Veterinary Dermatology and the Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Jae-Eun Hyun
- Laboratory of Veterinary Dermatology and the Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Cheol-Yong Hwang
- Laboratory of Veterinary Dermatology and the Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| |
Collapse
|
24
|
Cao ZQ, Lv JM, Liu Q, Qin SY, Chen GD, Dai P, Zhong Y, Gao H, Yao XS, Hu D. Biosynthetic Study of Cephalosporin P 1 Reveals a Multifunctional P450 Enzyme and a Site-Selective Acetyltransferase. ACS Chem Biol 2020; 15:44-51. [PMID: 31860257 DOI: 10.1021/acschembio.9b00863] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Fusidane-type antibiotics are a group of triterpenoid antibiotics. They include helvolic acid, fusidic acid, and cephalosporin P1, among which fusidic acid has been used clinically. We have recently elucidated the biosynthesis of helvolic acid and fusidic acid, which share an early biosynthetic route involving six conserved enzymes. Here, we report two separate gene clusters for cephalosporin P1 biosynthesis. One consists of the six conserved genes, and the other contains three genes encoding a P450 enzyme (CepB4), an acetyltransferase (CepD2), and a short-chain dehydrogenase/reductase (CepC2). Introduction of these three genes into Aspergillus oryzae, which harbors the six conserved genes, produced cephalosporin P1. Stepwise introduction revealed that CepB4 not only catalyzes stereoselective dual oxidation of C6 and C7, but also monooxygenation of C6 or C7. This led to the generation of five new analogues. Using monohydroxylated products as substrates, we demonstrated that CepD2 specifically acetylates C6-OH, although both C6-OH and C7-OH acetylated analogues have been identified in nature.
Collapse
Affiliation(s)
- Zhi-Qin Cao
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou 510632, People’s Republic of China
- Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, People’s Republic of China
| | - Jian-Ming Lv
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou 510632, People’s Republic of China
| | - Qiu Liu
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou 510632, People’s Republic of China
| | - Sheng-Ying Qin
- Clinical Experimental Center, First Affiliated Hospital of Jinan University, Guangzhou 510630, People’s Republic of China
| | - Guo-Dong Chen
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou 510632, People’s Republic of China
| | - Ping Dai
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou 510632, People’s Republic of China
| | - Yue Zhong
- Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, People’s Republic of China
| | - Hao Gao
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou 510632, People’s Republic of China
| | - Xin-Sheng Yao
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou 510632, People’s Republic of China
| | - Dan Hu
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou 510632, People’s Republic of China
| |
Collapse
|
25
|
Cao Z, Li S, Lv J, Gao H, Chen G, Awakawa T, Abe I, Yao X, Hu D. Biosynthesis of clinically used antibiotic fusidic acid and identification of two short-chain dehydrogenase/reductases with converse stereoselectivity. Acta Pharm Sin B 2019; 9:433-442. [PMID: 30972287 PMCID: PMC6437595 DOI: 10.1016/j.apsb.2018.10.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 10/17/2018] [Accepted: 10/25/2018] [Indexed: 11/28/2022] Open
Abstract
Fusidic acid is the only fusidane-type antibiotic that has been clinically used. However, biosynthesis of this important molecule in fungi is poorly understood. We have recently elucidated the biosynthesis of fusidane-type antibiotic helvolic acid, which provides us with clues to identify a possible gene cluster for fusidic acid (fus cluster). This gene cluster consists of eight genes, among which six are conserved in the helvolic acid gene cluster except fusC1 and fusB1. Introduction of the two genes into the Aspergillus oryzae NSAR1 expressing the conserved six genes led to the production of fusidic acid. A stepwise introduction of fusC1 and fusB1 revealed that the two genes worked independently without a strict reaction order. Notably, we identified two short-chain dehydrogenase/reductase genes fusC1 and fusC2 in the fus cluster, which showed converse stereoselectivity in 3-ketoreduction. This is the first report on the biosynthesis and heterologous expression of fusidic acid.
Collapse
Affiliation(s)
- Zhiqin Cao
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou 510632, China
| | - Shaoyang Li
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou 510632, China
| | - Jianming Lv
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou 510632, China
| | - Hao Gao
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou 510632, China
| | - Guodong Chen
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou 510632, China
| | - Takayoshi Awakawa
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Ikuro Abe
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Xinsheng Yao
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou 510632, China
- Corresponding authors.
| | - Dan Hu
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou 510632, China
- Corresponding authors.
| |
Collapse
|
26
|
Foster TJ. Antibiotic resistance in Staphylococcus aureus. Current status and future prospects. FEMS Microbiol Rev 2018; 41:430-449. [PMID: 28419231 DOI: 10.1093/femsre/fux007] [Citation(s) in RCA: 428] [Impact Index Per Article: 61.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 02/12/2017] [Indexed: 12/11/2022] Open
Abstract
The major targets for antibiotics in staphylococci are (i) the cell envelope, (ii) the ribosome and (iii) nucleic acids. Several novel targets emerged from recent targeted drug discovery programmes including the ClpP protease and FtsZ from the cell division machinery. Resistance can either develop by horizontal transfer of resistance determinants encoded by mobile genetic elements viz plasmids, transposons and the staphylococcal cassette chromosome or by mutations in chromosomal genes. Horizontally acquired resistance can occur by one of the following mechanisms: (i) enzymatic drug modification and inactivation, (ii) enzymatic modification of the drug binding site, (iii) drug efflux, (iv) bypass mechanisms involving acquisition of a novel drug-resistant target, (v) displacement of the drug to protect the target. Acquisition of resistance by mutation can result from (i) alteration of the drug target that prevents the inhibitor from binding, (ii) derepression of chromosomally encoded multidrug resistance efflux pumps and (iii) multiple stepwise mutations that alter the structure and composition of the cell wall and/or membrane to reduce drug access to its target. This review focuses on development of resistance to currently used antibiotics and examines future prospects for new antibiotics and informed use of drug combinations.
Collapse
|
27
|
Effect of Fusidic Acid on the Kinetics of Molecular Motions During EF-G-Induced Translocation on the Ribosome. Sci Rep 2017; 7:10536. [PMID: 28874811 PMCID: PMC5585275 DOI: 10.1038/s41598-017-10916-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 08/16/2017] [Indexed: 11/08/2022] Open
Abstract
The translocation step of protein synthesis entails binding and dissociation of elongation factor G (EF-G), movements of the two tRNA molecules, and motions of the ribosomal subunits. The translocation step is targeted by many antibiotics. Fusidic acid (FA), an antibiotic that blocks EF-G on the ribosome, may also interfere with some of the ribosome rearrangements, but the exact timing of inhibition remains unclear. To follow in real-time the dynamics of the ribosome–tRNA–EF-G complex, we have developed a fluorescence toolbox which allows us to monitor the key molecular motions during translocation. Here we employed six different fluorescence observables to investigate how FA affects translocation kinetics. We found that FA binds to an early translocation intermediate, but its kinetic effect on tRNA movement is small. FA does not affect the synchronous forward (counterclockwise) movements of the head and body domains of the small ribosomal subunit, but exerts a strong effect on the rates of late translocation events, i.e. backward (clockwise) swiveling of the head domain and the transit of deacylated tRNA through the E′ site, in addition to blocking EF-G dissociation. The use of ensemble kinetics and numerical integration unraveled how the antibiotic targets molecular motions within the ribosome-EF-G complex.
Collapse
|
28
|
Abouelfetouh A, Kassem M, Naguib M, El-Nakeeb M. Investigation and Treatment of Fusidic Acid Resistance Among Methicillin-Resistant Staphylococcal Isolates from Egypt. Microb Drug Resist 2017; 23:8-17. [DOI: 10.1089/mdr.2015.0336] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Affiliation(s)
- Alaa Abouelfetouh
- Department of Microbiology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Mervat Kassem
- Department of Microbiology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Marwa Naguib
- Department of Microbiology, Faculty of Pharmacy, Damanhour University, Damanhour, Egypt
| | - Moustafa El-Nakeeb
- Department of Microbiology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| |
Collapse
|
29
|
Osterman IA, Komarova ES, Shiryaev DI, Korniltsev IA, Khven IM, Lukyanov DA, Tashlitsky VN, Serebryakova MV, Efremenkova OV, Ivanenkov YA, Bogdanov AA, Sergiev PV, Dontsova OA. Sorting Out Antibiotics' Mechanisms of Action: a Double Fluorescent Protein Reporter for High-Throughput Screening of Ribosome and DNA Biosynthesis Inhibitors. Antimicrob Agents Chemother 2016; 60:7481-7489. [PMID: 27736765 PMCID: PMC5119032 DOI: 10.1128/aac.02117-16] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 10/04/2016] [Indexed: 11/20/2022] Open
Abstract
In order to accelerate drug discovery, a simple, reliable, and cost-effective system for high-throughput identification of a potential antibiotic mechanism of action is required. To facilitate such screening of new antibiotics, we created a double-reporter system for not only antimicrobial activity detection but also simultaneous sorting of potential antimicrobials into those that cause ribosome stalling and those that induce the SOS response due to DNA damage. In this reporter system, the red fluorescent protein gene rfp was placed under the control of the SOS-inducible sulA promoter. The gene of the far-red fluorescent protein, katushka2S, was inserted downstream of the tryptophan attenuator in which two tryptophan codons were replaced by alanine codons, with simultaneous replacement of the complementary part of the attenuator to preserve the ability to form secondary structures that influence transcription termination. This genetically modified attenuator makes possible Katushka2S expression only upon exposure to ribosome-stalling compounds. The application of red and far-red fluorescent proteins provides a high signal-to-background ratio without any need of enzymatic substrates for detection of the reporter activity. This reporter was shown to be efficient in high-throughput screening of both synthetic and natural chemicals.
Collapse
Affiliation(s)
- Ilya A Osterman
- Lomonosov Moscow State University, Department of Chemistry and A. N. Belozersky Institute of Physico-Chemical Biology, Moscow, Russia
- Skolkovo Institute of Science and Technology, Skolkovo, Russia
| | - Ekaterina S Komarova
- Lomonosov Moscow State University, Faculty of Bioengineering and Bioinformatics, Moscow, Russia
| | - Dmitry I Shiryaev
- Lomonosov Moscow State University, Department of Chemistry and A. N. Belozersky Institute of Physico-Chemical Biology, Moscow, Russia
| | - Ilya A Korniltsev
- Lomonosov Moscow State University, Department of Chemistry and A. N. Belozersky Institute of Physico-Chemical Biology, Moscow, Russia
| | - Irina M Khven
- Lomonosov Moscow State University, Faculty of Bioengineering and Bioinformatics, Moscow, Russia
| | - Dmitry A Lukyanov
- Lomonosov Moscow State University, Department of Chemistry and A. N. Belozersky Institute of Physico-Chemical Biology, Moscow, Russia
| | - Vadim N Tashlitsky
- Lomonosov Moscow State University, Department of Chemistry and A. N. Belozersky Institute of Physico-Chemical Biology, Moscow, Russia
| | - Marina V Serebryakova
- Lomonosov Moscow State University, Department of Chemistry and A. N. Belozersky Institute of Physico-Chemical Biology, Moscow, Russia
| | - Olga V Efremenkova
- G. F. Gauze Institute for Search for New Antibiotics, Russian Academy of Medical Sciences, Moscow, Russia
| | - Yan A Ivanenkov
- Moscow Institute of Physics and Technology (State University), Moscow Region, Russia
| | - Alexey A Bogdanov
- Lomonosov Moscow State University, Department of Chemistry and A. N. Belozersky Institute of Physico-Chemical Biology, Moscow, Russia
| | - Petr V Sergiev
- Lomonosov Moscow State University, Department of Chemistry and A. N. Belozersky Institute of Physico-Chemical Biology, Moscow, Russia
- Skolkovo Institute of Science and Technology, Skolkovo, Russia
| | - Olga A Dontsova
- Lomonosov Moscow State University, Department of Chemistry and A. N. Belozersky Institute of Physico-Chemical Biology, Moscow, Russia
- Skolkovo Institute of Science and Technology, Skolkovo, Russia
| |
Collapse
|
30
|
Pushkin R, Iglesias-Ussel MD, Keedy K, MacLauchlin C, Mould DR, Berkowitz R, Kreuzer S, Darouiche R, Oldach D, Fernandes P. A Randomized Study Evaluating Oral Fusidic Acid (CEM-102) in Combination With Oral Rifampin Compared With Standard-of-Care Antibiotics for Treatment of Prosthetic Joint Infections: A Newly Identified Drug-Drug Interaction. Clin Infect Dis 2016; 63:1599-1604. [PMID: 27682068 DOI: 10.1093/cid/ciw665] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 09/14/2016] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Fusidic acid (FA) has been used for decades for bone infection, including prosthetic joint infection (PJI), often in combination with rifampin (RIF). An FA/RIF pharmacokinetic interaction has not previously been described. METHODS In a phase 2 open-label randomized study, we evaluated oral FA/RIF vs standard-of-care (SOC) intravenous antibiotics for treatment of hip or knee PJI. Outcome assessment occurred at reimplantation (week 12) for subjects with 2-stage exchange, and after 3 or 6 months of treatment for subjects with hip or knee debride and retain strategies, respectively. RESULTS Fourteen subjects were randomized 1:1 to FA/RIF or SOC. Pharmacokinetic profiles were obtained for 6 subjects randomized to FA/RIF. FA concentrations were lower than anticipated in all subjects during the first week of therapy, and at weeks 4 and 6, blood levels continued to decline. By week 6, FA exposures were 40%-45% lower than expected. CONCLUSIONS The sponsor elected to terminate this study due to a clearly illustrated drug-drug interaction between FA and RIF, which lowered FA levels to a degree that could influence subject outcomes. Optimization of FA exposure if used in combination with RIF should be a topic of future research. CLINICAL TRIALS REGISTRATION NCT01756924.
Collapse
Affiliation(s)
| | | | | | | | - Diane R Mould
- Projections Research Inc, Phoenixville, Pennsylvania
| | | | - Stephan Kreuzer
- Memorial Bone and Joint Clinic and University of Texas Health Science Center at Houston
| | - Rabih Darouiche
- Departments of Medicine, Surgery, and Physical Medicine and Rehabilitation, Michael E. DeBakey Veterans Affairs Medical Center and Baylor College of Medicine, Houston, Texas
| | | | | |
Collapse
|
31
|
Discovery and Analysis of Natural-Product Compounds Inhibiting Protein Synthesis in Pseudomonas aeruginosa. Antimicrob Agents Chemother 2016; 60:4820-9. [PMID: 27246774 DOI: 10.1128/aac.00800-16] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Accepted: 05/23/2016] [Indexed: 11/20/2022] Open
Abstract
Bacterial protein synthesis is the target for numerous natural and synthetic antibacterial agents. We have developed a poly(U) mRNA-directed aminoacylation/translation (A/T) protein synthesis system composed of phenylalanyl-tRNA synthetases (PheRS), ribosomes, and ribosomal factors from Pseudomonas aeruginosa This system has been used for high-throughput screening of a natural-compound library. Assays were developed for each component of the system to ascertain the specific target of inhibitory compounds. In high-throughput screens, 13 compounds were identified that inhibit protein synthesis with 50% inhibitory concentrations ranging from 0.3 to >80 μM. MICs were determined for the compounds against the growth of a panel of pathogenic organisms, including Enterococcus faecalis, Escherichia coli, Haemophilus influenzae, Moraxella catarrhalis, P. aeruginosa, Staphylococcus aureus, and Streptococcus pneumoniae Three of the compounds were observed to have broad-spectrum activity and inhibited a hypersensitive strain of P. aeruginosa with MICs of 8 to 16 μg/ml. The molecular target of each of the three compounds was determined to be PheRS. One compound was found to be bacteriostatic, and one compound was bactericidal against both Gram-positive and Gram-negative pathogens. The third compound was observed to be bacteriostatic against Gram-positive and bactericidal against Gram-negative bacteria. All three compounds were competitive with the substrate ATP; however, one compound was competitive, one was uncompetitive, and one noncompetitive with the amino acid substrate. Macromolecular synthesis assays confirm the compounds inhibit protein synthesis. The compounds were shown to be more than 25,000-fold less active than the control staurosporine in cytotoxicity MTT testing in human cell lines.
Collapse
|
32
|
Tomlinson JH, Thompson GS, Kalverda AP, Zhuravleva A, O'Neill AJ. A target-protection mechanism of antibiotic resistance at atomic resolution: insights into FusB-type fusidic acid resistance. Sci Rep 2016; 6:19524. [PMID: 26781961 PMCID: PMC4725979 DOI: 10.1038/srep19524] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 12/09/2015] [Indexed: 11/09/2022] Open
Abstract
Antibiotic resistance in clinically important bacteria can be mediated by proteins that physically associate with the drug target and act to protect it from the inhibitory effects of an antibiotic. We present here the first detailed structural characterization of such a target protection mechanism mediated through a protein-protein interaction, revealing the architecture of the complex formed between the FusB fusidic acid resistance protein and the drug target (EF-G) it acts to protect. Binding of FusB to EF-G induces conformational and dynamic changes in the latter, shedding light on the molecular mechanism of fusidic acid resistance.
Collapse
Affiliation(s)
- Jennifer H Tomlinson
- School of Molecular and Cellular Biology, Garstang Building, University of Leeds, Leeds, UK, LS2 9JT.,Astbury Centre for Structural Molecular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK, LS2 9JT
| | - Gary S Thompson
- School of Molecular and Cellular Biology, Garstang Building, University of Leeds, Leeds, UK, LS2 9JT.,Astbury Centre for Structural Molecular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK, LS2 9JT
| | - Arnout P Kalverda
- School of Molecular and Cellular Biology, Garstang Building, University of Leeds, Leeds, UK, LS2 9JT.,Astbury Centre for Structural Molecular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK, LS2 9JT
| | - Anastasia Zhuravleva
- School of Molecular and Cellular Biology, Garstang Building, University of Leeds, Leeds, UK, LS2 9JT.,Astbury Centre for Structural Molecular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK, LS2 9JT
| | - Alex J O'Neill
- School of Molecular and Cellular Biology, Garstang Building, University of Leeds, Leeds, UK, LS2 9JT.,Astbury Centre for Structural Molecular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK, LS2 9JT
| |
Collapse
|
33
|
Achenbach J, Nierhaus KH. The mechanics of ribosomal translocation. Biochimie 2015; 114:80-9. [DOI: 10.1016/j.biochi.2014.12.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 12/05/2014] [Indexed: 11/16/2022]
|
34
|
Structure of the ribosome with elongation factor G trapped in the pretranslocation state. Proc Natl Acad Sci U S A 2013; 110:20994-9. [PMID: 24324137 DOI: 10.1073/pnas.1311423110] [Citation(s) in RCA: 112] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
During protein synthesis, tRNAs and their associated mRNA codons move sequentially on the ribosome from the A (aminoacyl) site to the P (peptidyl) site to the E (exit) site in a process catalyzed by a universally conserved ribosome-dependent GTPase [elongation factor G (EF-G) in prokaryotes and elongation factor 2 (EF-2) in eukaryotes]. Although the high-resolution structure of EF-G bound to the posttranslocation ribosome has been determined, the pretranslocation conformation of the ribosome bound with EF-G and A-site tRNA has evaded visualization owing to the transient nature of this state. Here we use electron cryomicroscopy to determine the structure of the 70S ribosome with EF-G, which is trapped in the pretranslocation state using antibiotic viomycin. Comparison with the posttranslocation ribosome shows that the small subunit of the pretranslocation ribosome is rotated by ∼12° relative to the large subunit. Domain IV of EF-G is positioned in the cleft between the body and head of the small subunit outwardly of the A site and contacts the A-site tRNA. Our findings suggest a model in which domain IV of EF-G promotes the translocation of tRNA from the A to the P site as the small ribosome subunit spontaneously rotates back from the hybrid, rotated state into the nonrotated posttranslocation state.
Collapse
|
35
|
Palmer SO, Rangel EY, Hu Y, Tran AT, Bullard JM. Two homologous EF-G proteins from Pseudomonas aeruginosa exhibit distinct functions. PLoS One 2013; 8:e80252. [PMID: 24260360 PMCID: PMC3832671 DOI: 10.1371/journal.pone.0080252] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Accepted: 10/11/2013] [Indexed: 11/28/2022] Open
Abstract
Genes encoding two proteins corresponding to elongation factor G (EF-G) were cloned from Pseudomonas aeruginosa. The proteins encoded by these genes are both members of the EFG I subfamily. The gene encoding one of the forms of EF-G is located in the str operon and the resulting protein is referred to as EF-G1A while the gene encoding the other form of EF-G is located in another part of the genome and the resulting protein is referred to as EF-G1B. These proteins were expressed and purified to 98% homogeneity. Sequence analysis indicated the two proteins are 90/84% similar/identical. In other organisms containing multiple forms of EF-G a lower degree of similarity is seen. When assayed in a poly(U)-directed poly-phenylalanine translation system, EF-G1B was 75-fold more active than EF-G1A. EF-G1A pre-incubate with ribosomes in the presence of the ribosome recycling factor (RRF) decreased polymerization of poly-phenylalanine upon addition of EF-G1B in poly(U)-directed translation suggesting a role for EF-G1A in uncoupling of the ribosome into its constituent subunits. Both forms of P. aeruginosa EF-G were active in ribosome dependent GTPase activity. The kinetic parameters (KM) for the interaction of EF-G1A and EF-G1B with GTP were 85 and 70 μM, respectively. However, EF-G1B exhibited a 5-fold greater turnover number (observed kcat) for the hydrolysis of GTP than EF-G1A; 0.2 s-1 vs. 0.04 s-1. These values resulted in specificity constants (kcatobs/KM) for EF-G1A and EF-G1B of 0.5 x 103 s-1 M-1 and 3.0 x 103 s-1 M-1, respectively. The antibiotic fusidic acid (FA) completely inhibited poly(U)-dependent protein synthesis containing P. aeruginosa EF-G1B, but the same protein synthesis system containing EF-G1A was not affected. Likewise, the activity of EF-G1B in ribosome dependent GTPase assays was completely inhibited by FA, while the activity of EF-G1A was not affected.
Collapse
Affiliation(s)
- Stephanie O. Palmer
- The University of Texas-Pan American, Edinburg, Texas, United States of America
| | - Edna Y. Rangel
- The University of Texas-Pan American, Edinburg, Texas, United States of America
| | - Yanmei Hu
- The University of Texas-Pan American, Edinburg, Texas, United States of America
| | - Alexis T. Tran
- The University of Texas-Pan American, Edinburg, Texas, United States of America
| | - James M. Bullard
- The University of Texas-Pan American, Edinburg, Texas, United States of America
- * E-mail:
| |
Collapse
|
36
|
Gupta A, Mir SS, Saqib U, Biswas S, Vaishya S, Srivastava K, Siddiqi MI, Habib S. The effect of fusidic acid on Plasmodium falciparum elongation factor G (EF-G). Mol Biochem Parasitol 2013; 192:39-48. [PMID: 24211494 DOI: 10.1016/j.molbiopara.2013.10.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Revised: 10/29/2013] [Accepted: 10/29/2013] [Indexed: 11/30/2022]
Abstract
Inhibition of growth of the malaria parasite Plasmodium falciparum by known translation-inhibitory antibiotics has generated interest in understanding their action on the translation apparatus of the two genome containing organelles of the malaria parasite: the mitochondrion and the relic plastid (apicoplast). We report GTPase activity of recombinant EF-G proteins that are targeted to the organelles and further use these to test the effect of the EF-G inhibitor fusidic acid (FA) on the factor-ribosome interface. Our results monitoring locking of EF-G·GDP onto surrogate Escherichia coli ribosomes as well as multi-turnover GTP hydrolysis by the factor indicate that FA has a greater effect on apicoplast EF-G compared to the mitochondrial counterpart. Deletion of a three amino acid (GVG) sequence in the switch I loop that is conserved in proteins of the mitochondrial EF-G1 family and the Plasmodium mitochondrial factor, but is absent in apicoplast EF-G, demonstrated that this motif contributes to differential inhibition of the two EF-Gs by FA. Additionally, the drug thiostrepton, that is known to target the apicoplast and proteasome, enhanced retention of only mitochondrial EF-G on ribosomes providing support for the reported effect of the drug on parasite mitochondrial translation.
Collapse
Affiliation(s)
- Ankit Gupta
- Division of Molecular and Structural Biology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Snober S Mir
- Division of Molecular and Structural Biology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Uzma Saqib
- Division of Molecular and Structural Biology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Subir Biswas
- Division of Molecular and Structural Biology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Suniti Vaishya
- Division of Molecular and Structural Biology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Kumkum Srivastava
- Division of Parasitology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Mohammad Imran Siddiqi
- Division of Molecular and Structural Biology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Saman Habib
- Division of Molecular and Structural Biology, CSIR-Central Drug Research Institute, Lucknow, India.
| |
Collapse
|
37
|
Peske F, Wintermeyer W. Antibiotics Inhibiting the Translocation Step of Protein Elongation on the Ribosome. Antibiotics (Basel) 2013. [DOI: 10.1002/9783527659685.ch21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
|
38
|
Proof of Principle for a Real-Time Pathogen Isolation Media Diagnostic: The Use of Laser-Induced Breakdown Spectroscopy to Discriminate Bacterial Pathogens and Antimicrobial-Resistant Staphylococcus aureus Strains Grown on Blood Agar. J Pathog 2013; 2013:898106. [PMID: 24109513 PMCID: PMC3784155 DOI: 10.1155/2013/898106] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2012] [Revised: 04/29/2013] [Accepted: 05/16/2013] [Indexed: 11/17/2022] Open
Abstract
Laser-Induced Breakdown Spectroscopy (LIBS) is a rapid, in situ, diagnostic technique in which light emissions from a laser plasma formed on the sample are used for analysis allowing automated analysis results to be available in seconds to minutes. This speed of analysis coupled with little or no sample preparation makes LIBS an attractive detection tool. In this study, it is demonstrated that LIBS can be utilized to discriminate both the bacterial species and strains of bacterial colonies grown on blood agar. A discrimination algorithm was created based on multivariate regression analysis of spectral data. The algorithm was deployed on a simulated LIBS instrument system to demonstrate discrimination capability using 6 species. Genetically altered Staphylococcus aureus strains grown on BA, including isogenic sets that differed only by the acquisition of mutations that increase fusidic acid or vancomycin resistance, were also discriminated. The algorithm successfully identified all thirteen cultures used in this study in a time period of 2 minutes. This work provides proof of principle for a LIBS instrumentation system that could be developed for the rapid discrimination of bacterial species and strains demonstrating relatively minor genomic alterations using data collected directly from pathogen isolation media.
Collapse
|
39
|
Guo X, Peisker K, Bäckbro K, Chen Y, Koripella RK, Mandava CS, Sanyal S, Selmer M. Structure and function of FusB: an elongation factor G-binding fusidic acid resistance protein active in ribosomal translocation and recycling. Open Biol 2013; 2:120016. [PMID: 22645663 PMCID: PMC3352095 DOI: 10.1098/rsob.120016] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2012] [Accepted: 02/23/2012] [Indexed: 11/12/2022] Open
Abstract
Fusidic acid (FA) is a bacteriostatic antibiotic that locks elongation factor G (EF-G) to the ribosome after GTP hydrolysis during elongation and ribosome recycling. The plasmid pUB101-encoded protein FusB causes FA resistance in clinical isolates of Staphylococcus aureus through an interaction with EF-G. Here, we report 1.6 and 2.3 Å crystal structures of FusB. We show that FusB is a two-domain protein lacking homology to known structures, where the N-terminal domain is a four-helix bundle and the C-terminal domain has an alpha/beta fold containing a C4 treble clef zinc finger motif and two loop regions with conserved basic residues. Using hybrid constructs between S. aureus EF-G that binds to FusB and Escherichia coli EF-G that does not, we show that the sequence determinants for FusB recognition reside in domain IV and involve the C-terminal helix of S. aureus EF-G. Further, using kinetic assays in a reconstituted translation system, we demonstrate that FusB can rescue FA inhibition of tRNA translocation as well as ribosome recycling. We propose that FusB rescues S. aureus from FA inhibition by preventing formation or facilitating dissociation of the FA-locked EF-G–ribosome complex.
Collapse
Affiliation(s)
- Xiaohu Guo
- Department of Cell and Molecular Biology, BMC, P.O. Box 596, SE 751 24, Uppsala, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Zhou J, Lancaster L, Donohue JP, Noller HF. Crystal structures of EF-G-ribosome complexes trapped in intermediate states of translocation. Science 2013; 340:1236086. [PMID: 23812722 DOI: 10.1126/science.1236086] [Citation(s) in RCA: 189] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Translocation of messenger and transfer RNA (mRNA and tRNA) through the ribosome is a crucial step in protein synthesis, whose mechanism is not yet understood. The crystal structures of three Thermus ribosome-tRNA-mRNA-EF-G complexes trapped with β,γ-imidoguanosine 5'-triphosphate (GDPNP) or fusidic acid reveal conformational changes occurring during intermediate states of translocation, including large-scale rotation of the 30S subunit head and body. In all complexes, the tRNA acceptor ends occupy the 50S subunit E site, while their anticodon stem loops move with the head of the 30S subunit to positions between the P and E sites, forming chimeric intermediate states. Two universally conserved bases of 16S ribosomal RNA that intercalate between bases of the mRNA may act as "pawls" of a translocational ratchet. These findings provide new insights into the molecular mechanism of ribosomal translocation.
Collapse
Affiliation(s)
- Jie Zhou
- Center for Molecular Biology of RNA and Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA 95064, USA
| | | | | | | |
Collapse
|
41
|
Mutagenesis mapping of the protein-protein interaction underlying FusB-type fusidic acid resistance. Antimicrob Agents Chemother 2013; 57:4640-4. [PMID: 23836182 PMCID: PMC3811445 DOI: 10.1128/aac.00198-13] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
FusB-type proteins represent the predominant mechanism of resistance to fusidic acid in staphylococci and act by binding to and modulating the function of the drug target (elongation factor G [EF-G]). To gain further insight into this antibiotic resistance mechanism, we sought to identify residues important for the interaction of FusB with EF-G and thereby delineate the binding interface within the FusB–EF-G complex. Replacement with alanine of any one of four conserved residues within the C-terminal domain of FusB (F156, K184, Y187, and F208) abrogated the ability of the protein to confer resistance to fusidic acid; the purified mutant proteins also lost the ability to bind S. aureus EF-G in vitro. E. coli EF-G, which is not ordinarily able to bind FusB-type proteins, was rendered competent for binding to FusB following deletion of a 3-residue tract (529SNP531) from domain IV of the protein. This study has identified key regions of both FusB and EF-G that are important for the interaction between the proteins, findings which corroborate our previous in silico prediction for the architecture of the complex formed between the resistance protein and the drug target (G. Cox, G. S. Thompson, H. T. Jenkins, F. Peske, A. Savelsbergh, M. V. Rodnina, W. Wintermeyer, S. W. Homans, T. A. Edwards, and A. J. O'Neill, Proc. Natl. Acad. Sci. U. S. A. 109:2102-2107, 2012).
Collapse
|
42
|
Wang L, Wasserman MR, Feldman MB, Altman RB, Blanchard SC. Mechanistic insights into antibiotic action on the ribosome through single-molecule fluorescence imaging. Ann N Y Acad Sci 2013; 1241:E1-16. [PMID: 23419024 DOI: 10.1111/j.1749-6632.2012.06839.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Single-molecule fluorescence imaging has provided unprecedented access to the dynamics of ribosome function, revealing transient intermediate states that are critical to ribosome activity. Imaging platforms have now been developed that are capable of probing many hundreds of molecules simultaneously at temporal and spatial resolutions approaching the sub-millisecond time and the sub-nanometer scales. These advances enable both steady- and pre-steady state measurements of individual steps in the translation process as well as processive reactions. The data generated using these methods have yielded new, quantitative structural and kinetic insights into ribosomal activity. They have also shed light on the mechanisms of antibiotic targeting the translation apparatus, revealing features of the structure-function relationship that would be difficult to obtain by other means. This review provides an overview of the types of information that can be obtained using such imaging platforms and a blueprint for using the technique to assess how small-molecule antibiotics alter macromolecular functions.
Collapse
Affiliation(s)
- Leyi Wang
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, NY 10065, USA
| | | | | | | | | |
Collapse
|
43
|
Nyfeler B, Hoepfner D, Palestrant D, Kirby CA, Whitehead L, Yu R, Deng G, Caughlan RE, Woods AL, Jones AK, Barnes SW, Walker JR, Gaulis S, Hauy E, Brachmann SM, Krastel P, Studer C, Riedl R, Estoppey D, Aust T, Movva NR, Wang Z, Salcius M, Michaud GA, McAllister G, Murphy LO, Tallarico JA, Wilson CJ, Dean CR. Identification of elongation factor G as the conserved cellular target of argyrin B. PLoS One 2012; 7:e42657. [PMID: 22970117 PMCID: PMC3438169 DOI: 10.1371/journal.pone.0042657] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Accepted: 07/10/2012] [Indexed: 11/19/2022] Open
Abstract
Argyrins, produced by myxobacteria and actinomycetes, are cyclic octapeptides with antibacterial and antitumor activity. Here, we identify elongation factor G (EF-G) as the cellular target of argyrin B in bacteria, via resistant mutant selection and whole genome sequencing, biophysical binding studies and crystallography. Argyrin B binds a novel allosteric pocket in EF-G, distinct from the known EF-G inhibitor antibiotic fusidic acid, revealing a new mode of protein synthesis inhibition. In eukaryotic cells, argyrin B was found to target mitochondrial elongation factor G1 (EF-G1), the closest homologue of bacterial EF-G. By blocking mitochondrial translation, argyrin B depletes electron transport components and inhibits the growth of yeast and tumor cells. Further supporting direct inhibition of EF-G1, expression of an argyrin B-binding deficient EF-G1 L693Q variant partially rescued argyrin B-sensitivity in tumor cells. In summary, we show that argyrin B is an antibacterial and cytotoxic agent that inhibits the evolutionarily conserved target EF-G, blocking protein synthesis in bacteria and mitochondrial translation in yeast and mammalian cells.
Collapse
Affiliation(s)
- Beat Nyfeler
- Developmental and Molecular Pathways, Novartis Institutes for BioMedical Research, Cambridge, Massachussetts, United States of America
| | - Dominic Hoepfner
- Developmental and Molecular Pathways, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Deborah Palestrant
- Center for Proteomic Chemistry, Novartis Institutes for BioMedical Research, Cambridge, Massachussetts, United States of America
| | - Christina A. Kirby
- Center for Proteomic Chemistry, Novartis Institutes for BioMedical Research, Cambridge, Massachussetts, United States of America
| | - Lewis Whitehead
- Global Discovery Chemistry, Novartis Institutes for Biomedical Research, Cambridge, Massachussetts, United States of America
| | - Robert Yu
- Infectious Diseases, Novartis Institutes for BioMedical Research, Emeryville, California, United States of America
| | - Gejing Deng
- Infectious Diseases, Novartis Institutes for BioMedical Research, Emeryville, California, United States of America
| | - Ruth E. Caughlan
- Infectious Diseases, Novartis Institutes for BioMedical Research, Emeryville, California, United States of America
| | - Angela L. Woods
- Infectious Diseases, Novartis Institutes for BioMedical Research, Emeryville, California, United States of America
| | - Adriana K. Jones
- Infectious Diseases, Novartis Institutes for BioMedical Research, Emeryville, California, United States of America
| | - S. Whitney Barnes
- Novartis Institute for Functional Genomics, Novartis Institutes for Biomedical Research, San Diego, California, United States of America
| | - John R. Walker
- Novartis Institute for Functional Genomics, Novartis Institutes for Biomedical Research, San Diego, California, United States of America
| | - Swann Gaulis
- Disease Area Oncology, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Ervan Hauy
- Disease Area Oncology, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Saskia M. Brachmann
- Disease Area Oncology, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Philipp Krastel
- Center for Proteomic Chemistry, Natural Products Unit, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Christian Studer
- Developmental and Molecular Pathways, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Ralph Riedl
- Developmental and Molecular Pathways, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - David Estoppey
- Developmental and Molecular Pathways, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Thomas Aust
- Developmental and Molecular Pathways, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - N. Rao Movva
- Developmental and Molecular Pathways, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Zuncai Wang
- Developmental and Molecular Pathways, Novartis Institutes for BioMedical Research, Cambridge, Massachussetts, United States of America
| | - Michael Salcius
- Developmental and Molecular Pathways, Novartis Institutes for BioMedical Research, Cambridge, Massachussetts, United States of America
| | - Gregory A. Michaud
- Developmental and Molecular Pathways, Novartis Institutes for BioMedical Research, Cambridge, Massachussetts, United States of America
| | - Gregory McAllister
- Developmental and Molecular Pathways, Novartis Institutes for BioMedical Research, Cambridge, Massachussetts, United States of America
| | - Leon O. Murphy
- Developmental and Molecular Pathways, Novartis Institutes for BioMedical Research, Cambridge, Massachussetts, United States of America
| | - John A. Tallarico
- Developmental and Molecular Pathways, Novartis Institutes for BioMedical Research, Cambridge, Massachussetts, United States of America
| | - Christopher J. Wilson
- Developmental and Molecular Pathways, Novartis Institutes for BioMedical Research, Cambridge, Massachussetts, United States of America
| | - Charles R. Dean
- Infectious Diseases, Novartis Institutes for BioMedical Research, Emeryville, California, United States of America
- * E-mail:
| |
Collapse
|
44
|
Koripella RK, Chen Y, Peisker K, Koh CS, Selmer M, Sanyal S. Mechanism of elongation factor-G-mediated fusidic acid resistance and fitness compensation in Staphylococcus aureus. J Biol Chem 2012; 287:30257-67. [PMID: 22767604 DOI: 10.1074/jbc.m112.378521] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Antibiotic resistance in bacteria is often associated with fitness loss, which is compensated by secondary mutations. Fusidic acid (FA), an antibiotic used against pathogenic bacteria Staphylococcus aureus, locks elongation factor-G (EF-G) to the ribosome after GTP hydrolysis. To clarify the mechanism of fitness loss and compensation in relation to FA resistance, we have characterized three S. aureus EF-G mutants with fast kinetics and crystal structures. Our results show that a significantly slower tRNA translocation and ribosome recycling, plus increased peptidyl-tRNA drop-off, are the causes for fitness defects of the primary FA-resistant mutant F88L. The double mutant F88L/M16I is three to four times faster than F88L in both reactions and showed no tRNA drop-off, explaining its fitness compensatory phenotype. The M16I mutation alone showed hypersensitivity to FA, higher activity, and somewhat increased affinity to GTP. The crystal structures demonstrate that Phe-88 in switch II is a key residue for FA locking and also for triggering interdomain movements in EF-G essential for its function, explaining functional deficiencies in F88L. The mutation M16I loosens the hydrophobic core in the G domain and affects domain I to domain II contact, resulting in improved activity both in the wild-type and F88L background. Thus, FA-resistant EF-G mutations causing fitness loss and compensation operate by affecting the conformational dynamics of EF-G on the ribosome.
Collapse
Affiliation(s)
- Ravi Kiran Koripella
- Department of Cell and Molecular Biology, Uppsala University, 75124 Uppsala, Sweden
| | | | | | | | | | | |
Collapse
|
45
|
Characterization of fusidic acid-resistant Staphylococcus aureus isolates in the community of Casablanca (Morocco). Int J Med Microbiol 2012; 302:96-100. [DOI: 10.1016/j.ijmm.2011.10.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2011] [Revised: 10/20/2011] [Accepted: 10/23/2011] [Indexed: 11/21/2022] Open
|
46
|
Ribosome clearance by FusB-type proteins mediates resistance to the antibiotic fusidic acid. Proc Natl Acad Sci U S A 2012; 109:2102-7. [PMID: 22308410 DOI: 10.1073/pnas.1117275109] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Resistance to the antibiotic fusidic acid (FA) in the human pathogen Staphylococcus aureus usually results from expression of FusB-type proteins (FusB or FusC). These proteins bind to elongation factor G (EF-G), the target of FA, and rescue translation from FA-mediated inhibition by an unknown mechanism. Here we show that the FusB family are two-domain metalloproteins, the C-terminal domain of which contains a four-cysteine zinc finger with a unique structural fold. This domain mediates a high-affinity interaction with the C-terminal domains of EF-G. By binding to EF-G on the ribosome, FusB-type proteins promote the dissociation of stalled ribosome⋅EF-G⋅GDP complexes that form in the presence of FA, thereby allowing the ribosomes to resume translation. Ribosome clearance by these proteins represents a highly unusual antibiotic resistance mechanism, which appears to be fine-tuned by the relative abundance of FusB-type protein, ribosomes, and EF-G.
Collapse
|
47
|
Identification of fusB-mediated fusidic acid resistance islands in Staphylococcus epidermidis isolates. Antimicrob Agents Chemother 2011; 55:5842-9. [PMID: 21968364 DOI: 10.1128/aac.00592-11] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
To understand the high prevalence of fusB genes in fusidic acid-resistant Staphylococcus epidermidis, analysis of resistance elements in 34 isolates was performed. First, sequence analysis of the aj1-LP-fusB region indicated that at least three types were present. Type I contained full-length aj1, type II contained a partial aj1 truncated from nucleotide position 93 to 421, and type III contained a more truncated aj1 that retained only the last 37 bp. Isolates with type I or type II aj1 displayed slightly higher levels of resistance to fusidic acid (MICs, 8 to 32 μg/ml) than did those with type III aj1 (MICs, 4 to 16 μg/ml). Subsequent sequencing of the flanking regions of fusB from four selected isolates carrying different types of aj1-LP-fusB regions revealed that the fusB genes were all located on phage-related resistance islands (RIs), referred to as SeRI(fusB)(-2793), SeRI(fusB)(-704), SeRI(fusB)(-5907), and SeRI(fusB)(-7778), respectively. Among them, three islands (SeRI(fusB)(-2793), SeRI(fusB)(-704), and SeRI(fusB)(-5907)) were located downstream of groEL (corresponding to the 44-min position based on Staphylococcus aureus whole genomic sequences), and one (SeRI(fusB)(-7778)) was located downstream of rpsR (corresponding to the 8-min position). All of the RIs were inserted into integrase-recognized att sites. Among 34 isolates, the insertion sites of fusB RIs were mostly (28/34, 82%) located downstream of groEL and two were located downstream of rpsR, but four remained unidentified. The pulsotype distribution indicated that fusB-containing S. epidermidis isolates were heterogeneous. In conclusion, the fusB resistance determinant in S. epidermidis was highly associated with phage-related RIs. This is the first report of fusB RI in S. epidermidis.
Collapse
|
48
|
Chen CM, Huang M, Chen HF, Ke SC, Li CR, Wang JH, Wu LT. Fusidic acid resistance among clinical isolates of methicillin-resistant Staphylococcus aureus in a Taiwanese hospital. BMC Microbiol 2011; 11:98. [PMID: 21569422 PMCID: PMC3114704 DOI: 10.1186/1471-2180-11-98] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2010] [Accepted: 05/12/2011] [Indexed: 11/26/2022] Open
Abstract
Background The prevalence of resistance to fusidic acid of methicillin-resistant Staphylococcus aureus (MRSA) was increased each year in a Taiwan hospital. Thirty-four MRSA clinical isolates collected in 2007 and 2008 with reduced susceptibility to FA were selected for further evaluation the presence of resistance determinants. Results The most common resistance determinant was fusC, found in 25 of the 34 MRSA isolates. One of the 25 fusidic acid-resistant MRSA harboured both fusB and fusC, which is the first time this has been identified. Mutations in fusA were found in 10 strains, a total of 3 amino-acid substitutions in EF-G (fusA gene) were detected. Two substitutions with G556S and R659L were identified for the first time. Low-level resistance to fusidic acid (MICs, ≤ 32 μg/ml) was found in most our collection. All collected isolates carried type III SCCmec elements. MLST showed the isolates were MRSA ST239. PFGE revealed nine different pulsotypes in one cluster. Conclusions Our results indicate that the increase in the number of fusidic acid resistant among the MRSA isolates in this hospital is due mainly to the distribution of fusC determinants. Moreover, more than one fusidic acid-resistance mechanism was first detected in a same stain in our collection.
Collapse
Affiliation(s)
- Chih-Ming Chen
- Department of Internal Medicine, Tungs' Taichung MetroHarbor Hospital, and Department of Microbiology, China Medical University, Taichung, Taiwan
| | | | | | | | | | | | | |
Collapse
|
49
|
mRNA translocation occurs during the second step of ribosomal intersubunit rotation. Nat Struct Mol Biol 2011; 18:457-62. [PMID: 21399643 PMCID: PMC3079290 DOI: 10.1038/nsmb.2011] [Citation(s) in RCA: 109] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2010] [Accepted: 12/15/2010] [Indexed: 11/09/2022]
Abstract
During protein synthesis, mRNA and tRNA undergo coupled translocation through the ribosome in a process that is catalyzed by elongation factor G (EF-G). On the basis of cryo-EM reconstructions, counterclockwise and clockwise rotational movements between the large and small ribosomal subunits have been implicated in a proposed ratcheting mechanism to drive the unidirectional movement of translocation. We used a combination of two fluorescence-based approaches to study the timing of these events, intersubunit fluorescence resonance energy transfer measurements to observe relative rotational movement of the subunits, and a fluorescence quenching assay to monitor translocation of mRNA. Binding of EF-G-GTP first induces rapid counterclockwise intersubunit rotation, followed by a slower, clockwise reversal of the rotational movement. We compared the rates of these movements and found that mRNA translocation occurs during the second, clockwise rotation event, corresponding to the transition from the hybrid state to the classical state.
Collapse
|
50
|
Distribution of fusidic acid resistance determinants in methicillin-resistant Staphylococcus aureus. Antimicrob Agents Chemother 2010; 55:1173-6. [PMID: 21149625 DOI: 10.1128/aac.00817-10] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The prevalence of resistance to fusidic acid in clinical isolates of Staphylococcus aureus, including methicillin-resistant S. aureus (MRSA), has increased in the past 2 decades. However, there are limited data regarding the relative importance in this process of the different staphylococcal determinants that mediate resistance to fusidic acid. Furthermore, the roles played by clonal dissemination of fusidic acid-resistant strains versus horizontal transmission of fusidic acid resistance determinants have not been investigated in detail. To gain insight into both issues, we examined fusidic acid resistance in 1,639 MRSA isolates collected in Denmark between 2003 and 2005. Resistance to fusidic acid (MIC, >1 μg/ml) was exhibited by 291 (17.6%) isolates. For the majority of these isolates (∼87%), resistance was attributed to carriage of fusB or fusC, while the remainder harbored mutations in the gene (fusA) encoding the drug target (EF-G). The CC80-MRSA-IV clone carrying fusB accounted for ∼61% of the resistant isolates in this collection, while a single CC5 clone harboring fusC represented ∼12% of the resistant strains. These findings emphasize the importance of clonal dissemination of fusidic acid resistance within European MRSA strains. Nonetheless, the distribution of fusB and fusC across several genetic lineages, and their presence on multiple genetic elements, indicates that horizontal transmission of fusidic acid resistance genes has also played an important role in the increasing prevalence of fusidic acid resistance in MRSA.
Collapse
|