1
|
Cartledge K, Short FL, Hall A, Lambert K, McDonald MJ, Lithgow T. Ethical bioprospecting and microbial assessments for sustainable solutions to the AMR crisis. IUBMB Life 2025; 77:e2931. [PMID: 39718471 PMCID: PMC11668235 DOI: 10.1002/iub.2931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 11/01/2024] [Indexed: 12/25/2024]
Abstract
Antimicrobial resistance (AMR) has been declared one of the top 10 global public health challenges of our age by the World Health Organization, and the World Bank describes AMR as a crisis affecting the finance, health, and agriculture sectors and a major threat to the attainment of Sustainable Development Goals. But what is AMR? It is a phenotype that evolves in microbes exposed to antimicrobial molecules and causes dangerous infections. This suggests that scientists and healthcare workers should be on the frontline in the search for sustainable solutions to AMR. Yet AMR is also a societal problem to be understood by everyone. This review aims to explore the need to address the problem of AMR through a coherent, international strategy with buy-in from all sectors of society. As reviewed here, the sustainable solutions to AMR will be driven by better understanding of AMR biology but will require more than this alone to succeed. Some advances on the horizon, such as the use of bacteriophage (phage) to treat AMR infections. However, many of the new technologies and new therapeutics to address AMR require access to biodiversity, where the custodians of that biodiversity-and the traditional knowledge required to access it-are needed as key partners in the scientific, clinical, biotechnological, and international ventures that would treat the problem of AMR and ultimately prevent its further evolution. Many of these advances will be built on microbial assessments to understand the extent of AMR in our environments and bioprospecting to identify microbes that may have beneficial uses. Genuine partnerships for access to this biodiversity and sharing of benefits accrued require a consideration of ethical practice and behavior. Behavior change is needed across all sectors of culturally diverse societies so that rapid deployment of solutions can be implemented for maximum effect against the impacts of AMR.
Collapse
Affiliation(s)
| | - Francesca L. Short
- Centre to Impact AMRMonash UniversityMelbourneAustralia
- Infection Program, Biomedicine Discovery Institute, Department of MicrobiologyMonash UniversityMelbourneAustralia
| | - Alex Hall
- Centre to Impact AMRMonash UniversityMelbourneAustralia
- Infection Program, Biomedicine Discovery Institute, Department of MicrobiologyMonash UniversityMelbourneAustralia
| | - Karen Lambert
- Centre to Impact AMRMonash UniversityMelbourneAustralia
- School of Curriculum, Teaching and Inclusive EducationMonash UniversityMelbourneAustralia
| | - Michael J. McDonald
- Centre to Impact AMRMonash UniversityMelbourneAustralia
- School of Biological SciencesMonash UniversityMelbourneAustralia
| | - Trevor Lithgow
- Centre to Impact AMRMonash UniversityMelbourneAustralia
- Infection Program, Biomedicine Discovery Institute, Department of MicrobiologyMonash UniversityMelbourneAustralia
| |
Collapse
|
2
|
Wharton T, Crawshay-Williams F, Schober T, Floto RA, Spring DR. Unlocking Amides: A General Method for the Self-Immolative Release of Amide-Containing Molecules. Angew Chem Int Ed Engl 2024; 63:e202402267. [PMID: 38411326 DOI: 10.1002/anie.202402267] [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: 01/31/2024] [Revised: 02/18/2024] [Accepted: 02/26/2024] [Indexed: 02/28/2024]
Abstract
The controlled liberation of molecules from a constructed framework is a subject of profound interest across various chemical fields. It allows for the masking of a molecule's properties and precise deployment upon a single controllable release event. While numerous methodologies have been developed for amines, alcohols, and thiols, approaches for utilising amides as payload-release handles are still in their early stages of development, despite the prevalence of amides in therapeutic compounds and materials. Herein, is presented a comprehensive strategy for the controlled and selective release of a diverse range of amides with stable linkers. The versatility of this approach is demonstrated by its successful application in the targeted release of various amide-containing drugs in their natural form via the use of commonly used trigger motifs, such as dipeptides or glycosides. As a proof of concept, the FDA-approved antibiotic linezolid has been successfully converted into a prodrug form and released selectively only in the presence of the trigger event. Significantly, in its prodrug state, no activity against Mycobacterium tuberculosis was exhibited. Linezolid's full potential was achieved only upon controlled release, where an equipotent efficacy to the free linezolid control was demonstrated, thus emphasising the immense potential of this method.
Collapse
Affiliation(s)
- Thomas Wharton
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, UK, CB2 1EW
| | - Felicity Crawshay-Williams
- University of Cambridge Molecular Immunity Unit, MRC Laboratory of Molecular Biology, Cambridge, UK, CB2 0QH
- Victor Phillip Dahdaleh Heart & Lung Research Institute, University of Cambridge, Cambridge, UK, CB2 0BB
| | - Tim Schober
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, UK, CB2 1EW
- Enamine Germany, Industriepark Hoechst G837, 65926, Frankfurt am Main, Germany
- Lumobiotics GmbH, Auerstrasse 2, 76227, Karlsruhe, Germany
| | - R Andres Floto
- University of Cambridge Molecular Immunity Unit, MRC Laboratory of Molecular Biology, Cambridge, UK, CB2 0QH
- Victor Phillip Dahdaleh Heart & Lung Research Institute, University of Cambridge, Cambridge, UK, CB2 0BB
| | - David R Spring
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, UK, CB2 1EW
| |
Collapse
|
3
|
Ampomah-Wireko M, Chen S, Li R, Gao C, Wang M, Qu Y, Kong H, Nininahazwe L, Zhang E. Recent advances in the exploration of oxazolidinone scaffolds from compound development to antibacterial agents and other bioactivities. Eur J Med Chem 2024; 269:116326. [PMID: 38513340 DOI: 10.1016/j.ejmech.2024.116326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/26/2024] [Accepted: 03/10/2024] [Indexed: 03/23/2024]
Abstract
Bacterial infections cause a variety of life-threatening diseases, and the continuous evolution of drug-resistant bacteria poses an increasing threat to current antimicrobial regimens. Gram-positive bacteria (GPB) have a wide range of genetic capabilities that allow them to adapt to and develop resistance to practically all existing antibiotics. Oxazolidinones, a class of potent bacterial protein synthesis inhibitors with a unique mechanism of action involving inhibition of bacterial ribosomal translation, has emerged as the antibiotics of choice for the treatment of drug-resistant GPB infections. In this review, we discussed the oxazolidinone antibiotics that are currently on the market and in clinical development, as well as an updated synopsis of current advances on their analogues, with an emphasis on innovative strategies for structural optimization of linezolid, structure-activity relationship (SAR), and safety properties. We also discussed recent efforts aimed at extending the activity of oxazolidinones to gram-negative bacteria (GNB), antitumor, and coagulation factor Xa. Oxazolidinone antibiotics can accumulate in GNB by a conjugation to siderophore-mediated β-lactamase-triggered release, making them effective against GNB.
Collapse
Affiliation(s)
- Maxwell Ampomah-Wireko
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, PR China
| | - Shengcong Chen
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, PR China
| | - Ruirui Li
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, PR China
| | - Chen Gao
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, PR China
| | - Meng Wang
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, PR China
| | - Ye Qu
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, PR China
| | - Hongtao Kong
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, PR China
| | - Lauraine Nininahazwe
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, PR China
| | - En Zhang
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, PR China; Pingyuan Laboratory (Zhengzhou University), PR China.
| |
Collapse
|
4
|
Natsheh IY, Alsaleh MM, Alkhawaldeh AK, Albadawi DK, Darwish MM, Shammout MJA. The dark side of drug repurposing. From clinical trial challenges to antimicrobial resistance: analysis based on three major fields. Drug Target Insights 2024; 18:8-19. [PMID: 38751378 PMCID: PMC11094707 DOI: 10.33393/dti.2024.3019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 04/18/2024] [Indexed: 05/18/2024] Open
Abstract
Drug repurposing is a strategic endeavor that entails the identification of novel therapeutic applications for pharmaceuticals that are already available in the market. Despite the advantageous nature of implementing this particular strategy owing to its cost-effectiveness and efficiency in reducing the time required for the drug discovery process, it is essential to bear in mind that there are various factors that must be meticulously considered and taken into account. Up to this point, there has been a noticeable absence of comprehensive analyses that shed light on the limitations of repurposing drugs. The primary aim of this review is to conduct a thorough illustration of the various challenges that arise when contemplating drug repurposing from a clinical perspective in three major fields-cardiovascular, cancer, and diabetes-and to further underscore the potential risks associated with the emergence of antimicrobial resistance (AMR) when employing repurposed antibiotics for the treatment of noninfectious and infectious diseases. The process of developing repurposed medications necessitates the application of creativity and innovation in designing the development program, as the body of evidence may differ for each specific case. In order to effectively repurpose drugs, it is crucial to consider the clinical implications and potential drawbacks that may arise during this process. By comprehensively analyzing these challenges, we can attain a deeper comprehension of the intricacies involved in drug repurposing, which will ultimately lead to the development of more efficacious and safe therapeutic approaches.
Collapse
Affiliation(s)
- Iyad Y. Natsheh
- Department of Medical Applied Sciences, Zarqa University College, Al-Balqa Applied University, Salt - Jordan
| | - Majd M. Alsaleh
- Department of Medical Applied Sciences, Zarqa University College, Al-Balqa Applied University, Salt - Jordan
- Department of Biology, School of Science, University of Jordan, Amman - Jordan
| | - Ahmad K. Alkhawaldeh
- Department of Medical Applied Sciences, Zarqa University College, Al-Balqa Applied University, Salt - Jordan
| | - Duaa K. Albadawi
- Department of Medical Applied Sciences, Zarqa University College, Al-Balqa Applied University, Salt - Jordan
| | - Maisa’ M. Darwish
- Department of Biology, School of Science, University of Jordan, Amman - Jordan
- National Agricultural Research Center, Amman - Jordan
| | | |
Collapse
|
5
|
Malik MS, Faazil S, Alsharif MA, Sajid Jamal QM, Al-Fahemi JH, Banerjee A, Chattopadhyay A, Pal SK, Kamal A, Ahmed SA. Antibacterial Properties and Computational Insights of Potent Novel Linezolid-Based Oxazolidinones. Pharmaceuticals (Basel) 2023; 16:516. [PMID: 37111273 PMCID: PMC10143092 DOI: 10.3390/ph16040516] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 03/23/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023] Open
Abstract
The mounting evidence of bacterial resistance against commonly prescribed antibiotics warrants the development of new antibacterial drugs on an urgent basis. Linezolid, an oxazolidinone antibiotic, is a lead molecule in designing new oxazolidinones as antibacterial agents. In this study, we report the antibacterial potential of the novel oxazolidinone-sulphonamide/amide conjugates that were recently reported by our research group. The antibacterial assays showed that, from the series, oxazolidinones 2 and 3a exhibited excellent potency (MIC of 1.17 μg/mL) against B. subtilis and P. aeruginosa strains, along with good antibiofilm activity. Docking studies revealed higher binding affinities of oxazolidinones 2 and 3a compared to linezolid, which were further validated by molecular dynamics simulations. In addition to this, other computational studies, one-descriptor (log P) analysis, ADME-T and drug likeness studies demonstrated the potential of these novel linezolid-based oxazolidinones to be taken forward for further studies.
Collapse
Affiliation(s)
- M. Shaheer Malik
- Department of Chemistry, Faculty of Applied Sciences, Umm Al-Qura University, Makkah 21955, Saudi Arabia; (M.A.A.)
| | - Shaikh Faazil
- Department of Chemistry, Poona College of Arts, Science and Commerce, Pune 411001, India
- Department of Medicinal Chemistry and Pharmacology, CSIR—Indian Institute of Chemical Technology, Hyderabad 500007, India
| | - Meshari A. Alsharif
- Department of Chemistry, Faculty of Applied Sciences, Umm Al-Qura University, Makkah 21955, Saudi Arabia; (M.A.A.)
| | - Qazi Mohammad Sajid Jamal
- Department of Health Informatics, College of Public Health and Health Informatics, Qassim University, Al Bukayriyah 52741, Saudi Arabia;
| | - Jabir H. Al-Fahemi
- Department of Chemistry, Faculty of Applied Sciences, Umm Al-Qura University, Makkah 21955, Saudi Arabia; (M.A.A.)
| | - Amrita Banerjee
- Department of Physics, Jadavpur University, 188, Raja S.C. Mallick Rd., Kolkata 700032, India;
- Technical Research Centre, S. N. Bose National Centre for Basic Sciences, Block JD, Sector III, Salt Lake, Kolkata 700106, India
| | - Arpita Chattopadhyay
- Department of Basic Science and Humanities, Techno International New Town, Block—DG 1/1, Action Area 1, New Town, Rajarhat, Kolkata 700156, India;
| | - Samir Kumar Pal
- Department of Chemical and Biological Sciences, S. N. Bose National Centre for Basic Sciences, Block JD, Sector 3, Salt Lake, Kolkata 700106, India;
| | - Ahmed Kamal
- Department of Medicinal Chemistry and Pharmacology, CSIR—Indian Institute of Chemical Technology, Hyderabad 500007, India
- Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad 500078, India
| | - Saleh A. Ahmed
- Department of Chemistry, Faculty of Applied Sciences, Umm Al-Qura University, Makkah 21955, Saudi Arabia; (M.A.A.)
- Chemistry Department, Faculty of Science, Assiut University, Assiut 71516, Egypt
| |
Collapse
|
6
|
Buckley ME, Ndukwe ARN, Nair PC, Rana S, Fairfull-Smith KE, Gandhi NS. Comparative Assessment of Docking Programs for Docking and Virtual Screening of Ribosomal Oxazolidinone Antibacterial Agents. Antibiotics (Basel) 2023; 12:463. [PMID: 36978331 PMCID: PMC10044086 DOI: 10.3390/antibiotics12030463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/21/2023] [Accepted: 02/23/2023] [Indexed: 03/03/2023] Open
Abstract
Oxazolidinones are a broad-spectrum class of synthetic antibiotics that bind to the 50S ribosomal subunit of Gram-positive and Gram-negative bacteria. Many crystal structures of the ribosomes with oxazolidinone ligands have been reported in the literature, facilitating structure-based design using methods such as molecular docking. It would be of great interest to know in advance how well docking methods can reproduce the correct ligand binding modes and rank these correctly. We examined the performance of five molecular docking programs (AutoDock 4, AutoDock Vina, DOCK 6, rDock, and RLDock) for their ability to model ribosomal-ligand interactions with oxazolidinones. Eleven ribosomal crystal structures with oxazolidinones as the ligands were docked. The accuracy was evaluated by calculating the docked complexes' root-mean-square deviation (RMSD) and the program's internal scoring function. The rankings for each program based on the median RMSD between the native and predicted were DOCK 6 > AD4 > Vina > RDOCK >> RLDOCK. Results demonstrate that the top-performing program, DOCK 6, could accurately replicate the ligand binding in only four of the eleven ribosomes due to the poor electron density of said ribosomal structures. In this study, we have further benchmarked the performance of the DOCK 6 docking algorithm and scoring in improving virtual screening (VS) enrichment using the dataset of 285 oxazolidinone derivatives against oxazolidinone binding sites in the S. aureus ribosome. However, there was no clear trend between the structure and activity of the oxazolidinones in VS. Overall, the docking performance indicates that the RNA pocket's high flexibility does not allow for accurate docking prediction, highlighting the need to validate VS. protocols for ligand-RNA before future use. Later, we developed a re-scoring method incorporating absolute docking scores and molecular descriptors, and the results indicate that the descriptors greatly improve the correlation of docking scores and pMIC values. Morgan fingerprint analysis was also used, suggesting that DOCK 6 underpredicted molecules with tail modifications with acetamide, n-methylacetamide, or n-ethylacetamide and over-predicted molecule derivatives with methylamino bits. Alternatively, a ligand-based approach similar to a field template was taken, indicating that each derivative's tail groups have strong positive and negative electrostatic potential contributing to microbial activity. These results indicate that one should perform VS. campaigns of ribosomal antibiotics with care and that more comprehensive strategies, including molecular dynamics simulations and relative free energy calculations, might be necessary in conjunction with VS. and docking.
Collapse
Affiliation(s)
- McKenna E. Buckley
- Centre for Genomics and Personalised Health, Queensland University of Technology, Brisbane, QLD 4059, Australia
- School of Chemistry and Physics, Queensland University of Technology, Brisbane, QLD 4000, Australia
| | - Audrey R. N. Ndukwe
- School of Chemistry and Physics, Queensland University of Technology, Brisbane, QLD 4000, Australia
- Centre for Materials Science, Queensland University of Technology, Brisbane, QLD 4000, Australia
| | - Pramod C. Nair
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Adelaide, SA 5042, Australia
- Flinders Health and Medical Research Institute (FHMRI), Flinders University, Adelaide, SA 5042, Australia
- South Australian Health and Medical Research Institute (SAHMRI), University of Adelaide, Adelaide, SA 5000, Australia
- Discipline of Medicine, Adelaide Medical School, The University of Adelaide, Adelaide, SA 5000, Australia
| | - Santu Rana
- Applied Artificial Intelligence Institute (A2I2), Deakin University, Geelong, VIC 3220, Australia
| | - Kathryn E. Fairfull-Smith
- School of Chemistry and Physics, Queensland University of Technology, Brisbane, QLD 4000, Australia
- Centre for Materials Science, Queensland University of Technology, Brisbane, QLD 4000, Australia
| | - Neha S. Gandhi
- Centre for Genomics and Personalised Health, Queensland University of Technology, Brisbane, QLD 4059, Australia
- School of Chemistry and Physics, Queensland University of Technology, Brisbane, QLD 4000, Australia
| |
Collapse
|
7
|
Cordeiro LV, Souza HDS, Sousa AP, Andrade Júnior FPDE, Figueiredo PTRDE, Oliveira RFDE, Athayde Filho PFDE, Oliveira-Filho AA, Lima EDEO. Effect of 2-chloro-N-(4-fluoro-3-nitrophenyl)acetamide in combination with antibacterial drugs against Klebsiella pneumoniae. AN ACAD BRAS CIENC 2023; 95:e20210141. [PMID: 36921147 DOI: 10.1590/0001-3765202320210141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 06/29/2021] [Indexed: 03/16/2023] Open
Abstract
Klebsiella pneumoniae is a species of Gram-negative bacteria related to a wide range of infections and high rates of drug resistance. The combined use of antibacterial agents is one of the strategies that has been analyzed in recent years as part of the alternatives in the treatment of drug-resistant infections. Recently, the antibacterial activity of of 2-chloro-N-(4-fluoro-3-nitrophenyl)acetamide has been demonstrated against K. pneumoniae, also indicating that this acetamide did not show significant cytotoxic potential in preliminary tests. Thus, it becomes an interesting substance for future studies that explore its antimicrobial capacity, including investigating its association with antibacterial drugs. Based on this, this research aimed to analyze the effects of the association of 2-chloro-N-(4-fluoro-3-nitrophenyl)acetamide (CFA) with ciprofloxacin, cefepime, ceftazidime, meropenem and imipenem against K. pneumoniae strains. The results showed additivity when the substance was combined with ciprofloxacin and cefepime, indifference when associated with ceftazidime and synergistic effect when combined with meropenem and imipenem. Thus, the acetamide was able to optimize the effects of antibacterial drugs, reducing the concentrations necessary to cause bacterial death. These data indicate a potential future clinical use of these combinations, and further studies are needed to analyze this viability.
Collapse
Affiliation(s)
- Laísa V Cordeiro
- Universidade Federal da Paraíba, Centro de Ciências da Saúde, Departamento de Ciências Farmacêuticas, Jardim Universitário, Cidade Universitária, s/n, Campus I, Castelo Branco, 58051-900 João Pessoa, PB, Brazil
| | - Helivaldo D S Souza
- Universidade Federal da Paraíba, Centro de Ciências Exatas e da Natureza, Departamento de Química, Jardim Universitário, Cidade Universitária, s/n, Campus I, Castelo Branco, 58051-900 João Pessoa, PB, Brazil
| | - Aleson P Sousa
- Universidade Federal da Paraíba, Centro de Ciências da Saúde, Departamento de Ciências Farmacêuticas, Jardim Universitário, Cidade Universitária, s/n, Campus I, Castelo Branco, 58051-900 João Pessoa, PB, Brazil
| | - Francisco P DE Andrade Júnior
- Universidade Federal da Paraíba, Centro de Ciências da Saúde, Departamento de Ciências Farmacêuticas, Jardim Universitário, Cidade Universitária, s/n, Campus I, Castelo Branco, 58051-900 João Pessoa, PB, Brazil
| | - Pedro T R DE Figueiredo
- Universidade Federal da Paraíba, Centro de Ciências da Saúde, Departamento de Ciências Farmacêuticas, Jardim Universitário, Cidade Universitária, s/n, Campus I, Castelo Branco, 58051-900 João Pessoa, PB, Brazil
| | - Rafael F DE Oliveira
- Universidade Federal da Paraíba, Centro de Ciências Exatas e da Natureza, Departamento de Química, Jardim Universitário, Cidade Universitária, s/n, Campus I, Castelo Branco, 58051-900 João Pessoa, PB, Brazil
| | - Petrônio F DE Athayde Filho
- Universidade Federal da Paraíba, Centro de Ciências Exatas e da Natureza, Departamento de Química, Jardim Universitário, Cidade Universitária, s/n, Campus I, Castelo Branco, 58051-900 João Pessoa, PB, Brazil
| | - Abrahão A Oliveira-Filho
- Universidade Federal de Campina Grande, Centro de Saúde e Tecnologia Rural, Unidade Acadêmica de Ciências Biológicas, Rua Aprígio Veloso, 882, Campus Patos, 58429-900 Campina Grande, PB, Brazil
| | - Edeltrudes DE O Lima
- Universidade Federal da Paraíba, Centro de Ciências da Saúde, Departamento de Ciências Farmacêuticas, Jardim Universitário, Cidade Universitária, s/n, Campus I, Castelo Branco, 58051-900 João Pessoa, PB, Brazil
| |
Collapse
|
8
|
Shchelik IS, Gademann K. Synthesis and Antimicrobial Evaluation of New Cephalosporin Derivatives Containing Cyclic Disulfide Moieties. ACS Infect Dis 2022; 8:2327-2338. [PMID: 36251034 DOI: 10.1021/acsinfecdis.2c00393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Due to a steady increase in microbial resistance, there is a need to increase the effectiveness of antibiotic performance by involving additional mechanisms of their penetration or retention for their better action. Cephalosporins are a successful group of antibiotics to combat pathogenic microorganisms, including drug-resistant strains. In this study, we investigated the effect of newly synthesized cephalosporin derivatives with cyclic disulfide modifications against several Gram-positive and Gram-negative strains as well as against biofilm formation. The incorporation of asparagusic acid was found to be effective in improving the activity of the drug against Gram-negative strains compared to the all carbon-control compounds. Furthermore, we could demonstrate the successful reduction of biofilm formation for Staphylococcus aureus and Pseudomonas aeruginosa at similar concentrations as obtained against planktonic cells. We propose that the incorporation of cyclic disulfides is one additional strategy to improve antibiotic activity and to combat bacterial infections.
Collapse
Affiliation(s)
- Inga S Shchelik
- Department of Chemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Karl Gademann
- Department of Chemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| |
Collapse
|
9
|
Wang J, Wu Y, Li Y. The crystal structure of ethyl 2,3,5-trifluoro-4-(4-oxo-3,4-dihydropyridin-1(2 H)-yl)benzoate, C 14H 12F 3NO 3. Z KRIST-NEW CRYST ST 2022. [DOI: 10.1515/ncrs-2022-0412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Abstract
C14H12F3NO3, monoclinic, P21/n (no. 14), a = 14.4583(12) Å, b = 6.6553(5) Å, c = 14.8395(11) Å, β = 113.733(3)°, V = 1307.16(18) Å3, Z = 4, Rgt
(F) = 0.0479, wRref
(F
2) = 0.1235, T = 170 K.
Collapse
Affiliation(s)
- Jingjing Wang
- Department of Student Affairs , Ningbo Polytechnic , No. 388, Lushan East Road, Ningbo Economic and Technological Development Zone, Beilun District , Ningbo City 315806 , Zhejiang Province , P. R. China
| | - Yundeng Wu
- Technique Center , Jinling Pharmaceutical Company Limited , Nanjing 210046 , Jiangsu Province , P. R. China
| | - Yong Li
- School of Chemical Engineering , Ningbo Polytechnic , No. 388, Lushan East Road, Ningbo Economic and Technological Development Zone, Beilun District , Ningbo City 315806 , Zhejiang Province , P. R. China
| |
Collapse
|
10
|
Perlaza-Jiménez L, Tan KS, Piper SJ, Johnson RM, Bamert RS, Stubenrauch CJ, Wright A, Lupton D, Lithgow T, Belousoff MJ. A Structurally Characterized Staphylococcus aureus Evolutionary Escape Route from Treatment with the Antibiotic Linezolid. Microbiol Spectr 2022; 10:e0058322. [PMID: 35736238 PMCID: PMC9431193 DOI: 10.1128/spectrum.00583-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 06/03/2022] [Indexed: 11/30/2022] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is a bacterial pathogen that presents great health concerns. Treatment requires the use of last-line antibiotics, such as members of the oxazolidinone family, of which linezolid is the first member to see regular use in the clinic. Here, we report a short time scale selection experiment in which strains of MRSA were subjected to linezolid treatment. Clonal isolates which had evolved a linezolid-resistant phenotype were characterized by whole-genome sequencing. Linezolid-resistant mutants were identified which had accumulated mutations in the ribosomal protein uL3. Multiple clones which had two mutations in uL3 exhibited resistance to linezolid, 2-fold higher than the clinical breakpoint. Ribosomes from this strain were isolated and subjected to single-particle cryo-electron microscopic analysis and compared to the ribosomes from the parent strain. We found that the mutations in uL3 lead to a rearrangement of a loop that makes contact with Helix 90, propagating a structural change over 15 Å away. This distal change swings nucleotide U2504 into the binding site of the antibiotic, causing linezolid resistance. IMPORTANCE Antibiotic resistance poses a critical problem to human health and decreases the utility of these lifesaving drugs. Of particular concern is the "superbug" methicillin-resistant Staphylococcus aureus (MRSA), for which treatment of infection requires the use of last-line antibiotics, including linezolid. In this paper, we characterize the atomic rearrangements which the ribosome, the target of linezolid, undergoes during its evolutionary journey toward becoming drug resistant. Using cryo-electron microscopy, we describe a particular molecular mechanism which MRSA uses to become resistant to linezolid.
Collapse
Affiliation(s)
- Laura Perlaza-Jiménez
- Centre to Impact AMR, Monash University, Clayton, Victoria, Australia
- Infection Program, Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Victoria, Australia
| | - Kher-Shing Tan
- Centre to Impact AMR, Monash University, Clayton, Victoria, Australia
- Infection Program, Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Victoria, Australia
| | - Sarah J. Piper
- Drug Development Biology, Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia
- Centre for Cryo-electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Rachel M. Johnson
- Drug Development Biology, Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia
- Centre for Cryo-electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Rebecca S. Bamert
- Centre to Impact AMR, Monash University, Clayton, Victoria, Australia
- Infection Program, Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Victoria, Australia
| | - Christopher J. Stubenrauch
- Centre to Impact AMR, Monash University, Clayton, Victoria, Australia
- Infection Program, Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Victoria, Australia
| | - Alexander Wright
- School of Chemistry, Monash University, Clayton, Victoria, Australia
| | - David Lupton
- School of Chemistry, Monash University, Clayton, Victoria, Australia
| | - Trevor Lithgow
- Centre to Impact AMR, Monash University, Clayton, Victoria, Australia
- Infection Program, Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Victoria, Australia
| | - Matthew J. Belousoff
- Infection Program, Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Victoria, Australia
- Drug Development Biology, Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia
- Centre for Cryo-electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| |
Collapse
|
11
|
Tsai K, Stojković V, Lee DJ, Young ID, Szal T, Klepacki D, Vázquez-Laslop N, Mankin AS, Fraser JS, Fujimori DG. Structural basis for context-specific inhibition of translation by oxazolidinone antibiotics. Nat Struct Mol Biol 2022; 29:162-171. [DOI: 10.1038/s41594-022-00723-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 01/05/2022] [Indexed: 01/02/2023]
|
12
|
Rosas NC, Lithgow T. Targeting bacterial outer-membrane remodelling to impact antimicrobial drug resistance. Trends Microbiol 2021; 30:544-552. [PMID: 34872824 DOI: 10.1016/j.tim.2021.11.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/29/2021] [Accepted: 11/03/2021] [Indexed: 12/14/2022]
Abstract
The cell envelope is essential for survival and adaptation of bacteria. Bacterial membrane proteins include the major porins that mediate the influx of nutrients and several classes of antimicrobial drugs. Consequently, membrane remodelling is closely linked to antimicrobial resistance (AMR). Knowledge of bacterial membrane protein biogenesis and turnover underpins our understanding of bacterial membrane remodelling and the consequences that this process have in the evolution of AMR phenotypes. At the population level, the evolution of phenotypes is a reversible process, and we can use these insights to deploy evolutionary principles to resensitize bacteria to existing antimicrobial drugs. In our opinion, fundamental knowledge is opening a new way of thinking towards sustainable solutions to the mounting crisis in AMR. Here we discuss what is known about outer-membrane remodelling in bacteria and how the process could be targeted as a means to restore sensitivity to antimicrobial drugs. Bacteriophages are highlighted as a powerful means to exert this control over membrane remodelling but they require careful selection so as to reverse, and not exacerbate, AMR phenotypes.
Collapse
Affiliation(s)
- Natalia C Rosas
- Centre to Impact AMR, Monash University, Melbourne, Australia; Infection and Immunity Program, Biomedicine Discovery Institute and Department of Microbiology, Monash University, Melbourne, Australia
| | - Trevor Lithgow
- Centre to Impact AMR, Monash University, Melbourne, Australia; Infection and Immunity Program, Biomedicine Discovery Institute and Department of Microbiology, Monash University, Melbourne, Australia.
| |
Collapse
|
13
|
Bellio MA, Young KC, Milberg J, Santos I, Abdullah Z, Stewart D, Arango A, Chen P, Huang J, Williams K, Kelly K, Sterling S, Khan A, Xu X, Shapiro GC, Mitrani MI. Amniotic fluid-derived extracellular vesicles: characterization and therapeutic efficacy in an experimental model of bronchopulmonary dysplasia. Cytotherapy 2021; 23:1097-1107. [PMID: 34538718 DOI: 10.1016/j.jcyt.2021.07.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 07/12/2021] [Accepted: 07/22/2021] [Indexed: 01/08/2023]
Abstract
BACKGROUND AIMS Extracellular vesicles (EVs) are being tested for their use as novel therapeutics. However, the optimal source of EVs is currently under investigation. Amniotic fluid (AF) is a natural source of EVs that can be easily obtained for use in regenerative medicine, yet AF-EV characterization has not been fully explored. METHODS Here the authors demonstrate AF as a rich source of EVs and identify the microRNA and proteomic cargo. Bioinformatics analysis of this cargo revealed multiple pathway targets, including immunomodulatory, anti-inflammatory and free radical scavenging networks. The authors further demonstrated the therapeutic potential of this EV product as a novel preventative agent for bronchopulmonary dysplasia (BPD). RESULTS Intra-tracheal administration of AF-EVs preserved alveolar development, attenuated vascular remodeling and pulmonary hypertension, decreased lung pro-inflammatory cytokine expression and reduced macrophage infiltration in an experimental BPD model. CONCLUSIONS The authors' results suggest that AF is a viable biological fluid for EV harvest and that AF-EVs have strong therapeutic potential for pulmonary diseases, such as BPD, warranting further development to transition this novel EV product into the clinic.
Collapse
Affiliation(s)
| | - Karen C Young
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Julian Milberg
- Organicell Regenerative Medicine, Inc, Miami, Florida, USA
| | - Ivan Santos
- Organicell Regenerative Medicine, Inc, Miami, Florida, USA
| | - Zanub Abdullah
- Organicell Regenerative Medicine, Inc, Miami, Florida, USA
| | | | - Alissa Arango
- Organicell Regenerative Medicine, Inc, Miami, Florida, USA
| | - Pingping Chen
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Jian Huang
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Kevin Williams
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Kaitlyn Kelly
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Shanique Sterling
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida, USA
| | | | - Xiumin Xu
- AssureImmune LLC, Miami, Florida, USA
| | | | | |
Collapse
|
14
|
Matsingos C, Al-Adhami T, Jamshidi S, Hind C, Clifford M, Mark Sutton J, Rahman KM. Synthesis, microbiological evaluation and structure activity relationship analysis of linezolid analogues with different C5-acylamino substituents. Bioorg Med Chem 2021; 49:116397. [PMID: 34619406 DOI: 10.1016/j.bmc.2021.116397] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 08/29/2021] [Accepted: 09/01/2021] [Indexed: 10/20/2022]
Abstract
Antimicrobial resistance and lack of new antibiotics to treat multidrug-resistant (MDR) bacteria is a significant public health problem. There is a discovery void and the pipeline of new classes of antibiotics in clinical development is almost empty. Therefore, it is important to understand the structure activity relationships (SAR) of current chemical classes as that can help the drug discovery community in their efforts to develop new antibiotics by modifying existing antibiotic classes. We studied the SAR of the C5-acylaminomethyl moiety of the linezolid, an oxazolidinone antibiotic, by synthesizing 25 compounds containing various aromatic, heteroaromatic and aliphatic substitutions. Our findings suggest that this position is highly important for the function of this antibiotic class, since only smaller non-polar fragments are tolerated at this position while larger and polar ones lead to a decrease in activity compared to linezolid. Our findings have led us to construct a structure activity relationship, around the C5-acylaminomethyl moiety of linezolid, that provides valuable insight into the function of the oxazolidinone class of antibiotics.
Collapse
Affiliation(s)
- Christos Matsingos
- Institute of Pharmaceutical Science, King's College London, London SE1 9NH, UK
| | - Taha Al-Adhami
- Institute of Pharmaceutical Science, King's College London, London SE1 9NH, UK
| | - Shirin Jamshidi
- Institute of Pharmaceutical Science, King's College London, London SE1 9NH, UK
| | - Charlotte Hind
- Public Health England, National Infections Service, Porton Down, Salisbury, Wiltshire SP4 0JG, UK
| | - Melanie Clifford
- Public Health England, National Infections Service, Porton Down, Salisbury, Wiltshire SP4 0JG, UK
| | - J Mark Sutton
- Public Health England, National Infections Service, Porton Down, Salisbury, Wiltshire SP4 0JG, UK.
| | | |
Collapse
|
15
|
Rekand IH, Brenk R. DrugPred_RNA-A Tool for Structure-Based Druggability Predictions for RNA Binding Sites. J Chem Inf Model 2021; 61:4068-4081. [PMID: 34286972 PMCID: PMC8389535 DOI: 10.1021/acs.jcim.1c00155] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
![]()
RNA is an emerging
target for drug discovery. However, like for
proteins, not all RNA binding sites are equally suited to be addressed
with conventional drug-like ligands. To this end, we have developed
the structure-based druggability predictor DrugPred_RNA to identify
druggable RNA binding sites. Due to the paucity of annotated RNA binding
sites, the predictor was trained on protein pockets, albeit using
only descriptors that can be calculated for both RNA and protein binding
sites. DrugPred_RNA performed well in discriminating druggable from
less druggable binding sites for the protein set and delivered predictions
for selected RNA binding sites that agreed with manual assignment.
In addition, most drug-like ligands contained in an RNA test set were
found in pockets predicted to be druggable, further adding confidence
to the performance of DrugPred_RNA. The method is robust against conformational
and sequence changes in the binding sites and can contribute to direct
drug discovery efforts for RNA targets.
Collapse
Affiliation(s)
- Illimar Hugo Rekand
- Department of Biomedicine, University of Bergen, Jonas Lies Vei, 5020 Bergen, Norway
| | - Ruth Brenk
- Department of Biomedicine, University of Bergen, Jonas Lies Vei, 5020 Bergen, Norway
| |
Collapse
|
16
|
Zhao Q, Xin L, Liu Y, Liang C, Li J, Jian Y, Li H, Shi Z, Liu H, Cao W. Current Landscape and Future Perspective of Oxazolidinone Scaffolds Containing Antibacterial Drugs. J Med Chem 2021; 64:10557-10580. [PMID: 34260235 DOI: 10.1021/acs.jmedchem.1c00480] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The widespread use of antibiotics has made the problem of bacterial resistance increasingly serious, and the study of new drug-resistant bacteria has become the main direction of antibacterial drug research. Among antibiotics, the fully synthetic oxazolidinone antibacterial drugs linezolid and tedizolid have been successfully marketed and have achieved good clinical treatment effects. Oxazolidinone antibacterial drugs have good pharmacokinetic and pharmacodynamic characteristics and unique antibacterial mechanisms, and resistant bacteria are sensitive to them. This Perspective focuses on reviewing oxazolidinones based on the structural modification of linezolid and new potential oxazolidinone drugs in the past 10 years, mainly describing their structure, antibacterial activity, safety, druggability, and so on, and discusses their structure-activity relationships, providing insight into the reasonable design of safer and more potent oxazolidinone antibacterial drugs.
Collapse
Affiliation(s)
- Qianqian Zhao
- Faculty of Pharmacy, Shaanxi University of Science & Technology, Xi'an 710021, P. R. China
| | - Liang Xin
- Faculty of Pharmacy, Shaanxi University of Science & Technology, Xi'an 710021, P. R. China.,Xi'an Xuri Shengchang Pharmaceutical Technology Co., Ltd., High-tech Zone, Xi'an 710075, P. R. China
| | - Yuzhi Liu
- Faculty of Pharmacy, Shaanxi University of Science & Technology, Xi'an 710021, P. R. China
| | - Chengyuan Liang
- Faculty of Pharmacy, Shaanxi University of Science & Technology, Xi'an 710021, P. R. China
| | - Jingyi Li
- Faculty of Pharmacy, Shaanxi University of Science & Technology, Xi'an 710021, P. R. China
| | - Yanlin Jian
- Faculty of Pharmacy, Shaanxi University of Science & Technology, Xi'an 710021, P. R. China
| | - Han Li
- Faculty of Pharmacy, Shaanxi University of Science & Technology, Xi'an 710021, P. R. China
| | - Zhenfeng Shi
- Department of Urology Surgery Center, Xinjiang Uyghur People's Hospital, Urumqi 830002, P. R. China
| | - Hong Liu
- Zhuhai Jinan Selenium Source Nanotechnology Co., Ltd., Hengqin New Area, Zhuhai 519030, P. R. China
| | - Wenqiang Cao
- Zhuhai Jinan Selenium Source Nanotechnology Co., Ltd., Hengqin New Area, Zhuhai 519030, P. R. China
| |
Collapse
|
17
|
Herrero del Valle A, Innis CA. Prospects for antimicrobial development in the cryo-EM era – a focus on the ribosome. FEMS Microbiol Rev 2020; 44:793-803. [DOI: 10.1093/femsre/fuaa032] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 07/25/2020] [Indexed: 12/12/2022] Open
Abstract
ABSTRACT
Resistance to antimicrobial drugs used to treat bacterial, viral, fungal and parasitic infections is a major health concern requiring a coordinated response across the globe. An important aspect in the fight against antimicrobial resistance is the development of novel drugs that are effective against resistant pathogens. Drug development is a complex trans-disciplinary endeavor, in which structural biology plays a major role by providing detailed functional and mechanistic information on an antimicrobial target and its interactions with small molecule inhibitors. Although X-ray crystallography and nuclear magnetic resonance have until now been the methods of choice to characterize microbial targets and drive structure-based drug development, cryo-electron microscopy is rapidly gaining ground in these areas. In this perspective, we will discuss how cryo-electron microscopy is changing our understanding of an established antimicrobial target, the ribosome, and how methodological developments could help this technique become an integral part of the antimicrobial drug discovery pipeline.
Collapse
Affiliation(s)
- Alba Herrero del Valle
- Institut Européen de Chimie et Biologie, Univ. Bordeaux, Institut National de la Santé et de la Recherche Médicale (U1212) and Centre National de la Recherche Scientifique (UMR 5320), 2 rue Robert Escarpit, 33607 Pessac, France
| | - C Axel Innis
- Institut Européen de Chimie et Biologie, Univ. Bordeaux, Institut National de la Santé et de la Recherche Médicale (U1212) and Centre National de la Recherche Scientifique (UMR 5320), 2 rue Robert Escarpit, 33607 Pessac, France
| |
Collapse
|
18
|
Wright A, Deane-Alder K, Marschall E, Bamert R, Venugopal H, Lithgow T, Lupton DW, Belousoff MJ. Characterization of the Core Ribosomal Binding Region for the Oxazolidone Family of Antibiotics Using Cryo-EM. ACS Pharmacol Transl Sci 2020; 3:425-432. [PMID: 32566908 DOI: 10.1021/acsptsci.0c00041] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Indexed: 01/02/2023]
Abstract
Linezolid and tedizolid are oxazolidinones with established clinical utility for the treatment of Gram-positive pathogens. Over time it has become apparent that even modest structural changes to the core phenyl oxazolidinone leads to drastic changes in biological activity. Consequently, the structure-activity relationship around the core oxazolidinone is constantly evolving, often reflected with new structural motifs present in nascent oxazolidinones. Herein we describe the use of cryo-electron microscopy to examine the differences in binding of several functionally different oxazolidinones in the hopes of enhanced understanding of their SAR. Tedizolid, radezolid, T145, and contezolid have been examined within the peptidyl transferase center (PTC) of the 50S ribosomal subunit from methicillin resistant Staphylococcus aureus. The ribosome-antibiotic complexes were resolved to a resolution of around 3 Å enabling unambiguous assignment of how each antibiotic interacts with the PTC.
Collapse
Affiliation(s)
- Alexander Wright
- School of Chemistry, Monash University, Wellington Road, Clayton, 3800 Victoria, Australia
| | - Kieran Deane-Alder
- Drug and Development Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, 3052 Victoria, Australia
| | - Edward Marschall
- Department of Biochemistry and Molecular Biology, Monash University, Wellington Road, Clayton, 3800 Victoria, Australia
| | - Rebecca Bamert
- Infection & Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Wellington Road, Clayton, 3800 Victoria, Australia
| | - Hari Venugopal
- Ramaciotti Center for Cryo-Electron Microscopy, Monash University, Wellington Road, Clayton, 3800 Victoria, Australia
| | - Trevor Lithgow
- Infection & Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Wellington Road, Clayton, 3800 Victoria, Australia
| | - David W Lupton
- School of Chemistry, Monash University, Wellington Road, Clayton, 3800 Victoria, Australia
| | - Matthew J Belousoff
- Drug and Development Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, 3052 Victoria, Australia
| |
Collapse
|
19
|
Characterization of the Self-Resistance Mechanism to Dityromycin in the Streptomyces Producer Strain. mSphere 2019; 4:4/5/e00554-19. [PMID: 31554724 PMCID: PMC6763770 DOI: 10.1128/msphere.00554-19] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Dityromycin is a peptide antibiotic isolated from the culture broth of the soil microorganism Streptomyces sp. strain AM-2504. Recent structural studies have shown that dityromycin targets the ribosomal protein S12 in the 30S ribosomal subunit, inhibiting translocation. Herein, by using in vitro protein synthesis assays, we identified the resistance mechanism of the producer strain to the secondary metabolite dityromycin. The results show that the self-resistance mechanism of the Streptomyces sp. strain AM-2504 is due to a specific modification of the ribosome. In particular, two amino acid substitutions, located in a highly conserved region of the S12 protein corresponding to the binding site of the antibiotic, were found. These mutations cause a substantial loss of affinity of the dityromycin for the 30S ribosomal subunit, protecting the producer strain from the toxic effect of the antibiotic. In addition to providing a detailed description of the first mechanism of self-resistance based on a mutated ribosomal protein, this work demonstrates that the molecular determinants of the dityromycin resistance identified in Streptomyces can be transferred to Escherichia coli ribosomes, where they can trigger the same antibiotic resistance mechanism found in the producer strain.IMPORTANCE The World Health Organization has identified antimicrobial resistance as a substantial threat to human health. Because of the emergence of pathogenic bacteria resistant to multiple antibiotics worldwide, there is a need to identify the mode of action of antibiotics and to unravel the basic mechanisms responsible for drug resistance. Antibiotic producers' microorganisms can protect themselves from the toxic effect of the drug using different strategies; one of the most common involves the modification of the antibiotic's target site. In this work, we report a detailed analysis of the molecular mechanism, based on protein modification, devised by the soil microorganism Streptomyces sp. strain AM-2504 to protect itself from the activity of the peptide antibiotic dityromycin. Furthermore, we demonstrate that this mechanism can be reproduced in E. coli, thereby eliciting antibiotic resistance in this human commensal bacterium.
Collapse
|