1
|
Henson BAB, Li F, Álvarez-Huerta JA, Wedamulla PG, Palacios AV, Scott MRM, Lim DTE, Scott WMH, Villanueva MTL, Ye E, Straus SK. Novel active Trp- and Arg-rich antimicrobial peptides with high solubility and low red blood cell toxicity designed using machine learning tools. Int J Antimicrob Agents 2025; 65:107399. [PMID: 39645171 DOI: 10.1016/j.ijantimicag.2024.107399] [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: 09/11/2024] [Revised: 11/07/2024] [Accepted: 11/29/2024] [Indexed: 12/09/2024]
Abstract
BACKGROUND Given the rising number of multidrug-resistant (MDR) bacteria, there is a need to design synthetic antimicrobial peptides (AMPs) that are highly active, non-hemolytic, and highly soluble. Machine learning tools allow the straightforward in silico identification of non-hemolytic antimicrobial peptides. METHODS Here, we utilized a number of these tools to rank the best peptides from two libraries comprised of: 1) a total of 8192 peptides with sequence bhxxbhbGAL, where b is the basic amino acid R or K, h is a hydrophobic amino acid, i.e. G, A, L, F, I, V, Y, or W and x is Q, S, A, or V; and 2) a total of 512 peptides with sequence RWhxbhRGWL, where b and h are as for the first library and x is Q, S, A, or G. The top 100 sequences from each library, as well as 10 peptides predicted to be active but hemolytic (for a total of 220 peptides), were SPOT synthesized and their IC50 values were determined against S. aureus USA 300 (MRSA). RESULTS Of these, 6 AMPs with low IC50's were characterized further in terms of: MICs against MRSA, E. faecalis, K. pneumoniae, E.coli and P. aeruginosa; RBC lysis; secondary structure in mammalian and bacterial model membranes; and activity against cancer cell lines HepG2, CHO, and PC-3. CONCLUSION Overall, the approach yielded a large family of active antimicrobial peptides with high solubility and low red blood cell toxicity. It also provides a framework for future designs and improved machine learning tools.
Collapse
Affiliation(s)
- Bridget A B Henson
- Department of Chemistry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Fucong Li
- Department of Chemistry, University of British Columbia, Vancouver, British Columbia, Canada
| | | | - Poornima G Wedamulla
- Department of Chemistry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Arianna Valdes Palacios
- Department of Chemistry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Max R M Scott
- Department of Chemistry, University of British Columbia, Vancouver, British Columbia, Canada
| | - David Thiam En Lim
- Department of Chemistry, University of British Columbia, Vancouver, British Columbia, Canada
| | - W M Hayden Scott
- Department of Chemistry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Monica T L Villanueva
- Department of Chemistry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Emily Ye
- Department of Chemistry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Suzana K Straus
- Department of Chemistry, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
2
|
Draveny M, Chauvet H, Rouam V, Jamme F, Masi M. Intracellular Quantification of an Antibiotic Metal Complex in Single Cells of Escherichia coli Using Cryo-X-ray Fluorescence Nanoimaging. ACS NANO 2024. [PMID: 39740123 DOI: 10.1021/acsnano.4c12664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
Abstract
Bacterial resistance is a major public health challenge. In Gram-negative bacteria, the synergy between multidrug efflux pumps and outer membrane impermeability determines the intracellular concentration of antibiotics. Consequently, it also dictates antibiotic activity on their respective targets. Previous research has employed spectrofluorimetry and synchrotron radiation-based DUV microscopy as tools for monitoring the accumulation of fluoroquinolone antibiotics in bacteria at population and single-cell scales, respectively. Here, we show that cryo-XRF nanoimaging allows intracellular localization and quantification of a fluoroquinolone metal complex accumulation in Escherichia coli with different efflux pump expression levels. This method offers a promising avenue for elucidating the intracellular behavior of a range of metallodrugs in bacteria and for designing novel agents with unique mechanisms of action.
Collapse
Affiliation(s)
- Margot Draveny
- Aix Marseille Univ, INSERM, SSA, MCT, 27 Bd Jean Moulin, Marseille 13005, France
- Synchrotron SOLEIL, L'Orme des Merisiers, Départementale 128, Saint-Aubin 91190, France
| | - Hugo Chauvet
- Synchrotron SOLEIL, L'Orme des Merisiers, Départementale 128, Saint-Aubin 91190, France
| | - Valérie Rouam
- Synchrotron SOLEIL, L'Orme des Merisiers, Départementale 128, Saint-Aubin 91190, France
| | - Frédéric Jamme
- Synchrotron SOLEIL, L'Orme des Merisiers, Départementale 128, Saint-Aubin 91190, France
| | - Muriel Masi
- Aix Marseille Univ, INSERM, SSA, MCT, 27 Bd Jean Moulin, Marseille 13005, France
- Synchrotron SOLEIL, L'Orme des Merisiers, Départementale 128, Saint-Aubin 91190, France
| |
Collapse
|
3
|
Koul V, Sharma A, Kumari D, Jamwal V, Palmo T, Singh K. Breaking the resistance: integrative approaches with novel therapeutics against Klebsiella pneumoniae. Arch Microbiol 2024; 207:18. [PMID: 39724243 DOI: 10.1007/s00203-024-04205-y] [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: 09/12/2024] [Revised: 11/14/2024] [Accepted: 11/18/2024] [Indexed: 12/28/2024]
Abstract
Klebsiella pneumoniae is a leading cause of anti-microbial resistance in healthcare-associated infections that have posed a severe threat to neonatal and wider community. The escalating crises of antibiotic resistance have compelled researchers to explore an innovative arsenal beginning from natural resources to chemical modifications in order to overcome the ever-increasing resistance issues. The present review highlights the drug discovery efforts with a special focus on cutting-edge strategies in the hunt for potential drug candidates against MDR/XDR Klebsiella pneumoniae. Nature's bounty constituting plant extracts, essential oils, fungal extracts, etc. holds promising anti-bacterial potential especially when combined with existing antibiotics. Further, enhancing these natural products with synthetic moieties has improved their effectiveness, creating a bridge between the natural and synthetic world. Conversely, the synthetically modified novel scaffolds have been also designed to meticulously target specific sites. Furthermore, we have also elaborated various emerging strategies for broad-spectrum infections caused by K. pneumoniae, which include anti-microbial peptides, nanotechnology, drug repurposing, bacteriophage, photodynamic, and multidrug therapies. This review further addresses the challenges confronted by the research community and the future way forward in the field of drug discovery against multi-resistant bacterial infections.
Collapse
Affiliation(s)
- Vimarishi Koul
- Infectious Diseases Division, CSIR- Indian Institute of Integrative Medicine, Jammu, 180001, India
- Department of Biological Sciences, Birla Institute of Technology and Sciences (BITS), Pilani campus, Pilani, Rajasthan, 333031, India
| | - Akshi Sharma
- Infectious Diseases Division, CSIR- Indian Institute of Integrative Medicine, Jammu, 180001, India
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, 201313, India
| | - Diksha Kumari
- Infectious Diseases Division, CSIR- Indian Institute of Integrative Medicine, Jammu, 180001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Vishwani Jamwal
- Infectious Diseases Division, CSIR- Indian Institute of Integrative Medicine, Jammu, 180001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Tashi Palmo
- Infectious Diseases Division, CSIR- Indian Institute of Integrative Medicine, Jammu, 180001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Kuljit Singh
- Infectious Diseases Division, CSIR- Indian Institute of Integrative Medicine, Jammu, 180001, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
4
|
Kubicskó A, Kamotsay K, Szabó D, Kocsis B. Analysis of molecular mechanisms of delafloxacin resistance in Escherichia coli. Sci Rep 2024; 14:26423. [PMID: 39488602 PMCID: PMC11531523 DOI: 10.1038/s41598-024-78124-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 10/29/2024] [Indexed: 11/04/2024] Open
Abstract
In this study delafloxacin resistance mechanisms in Escherichia coli strains were analyzed. Delafloxacin is a new fluoroquinolone, that is approved for clinical application however, resistance against this agent is scarcely reported. In our study 37 E. coli strains were included and antimicrobial susceptibility testing was performed for ciprofloxacin, delafloxacin, levofloxacin, moxifloxacin, ceftazidime, cefotaxime, imipenem. Six delafloxacin resistant E. coli strains were selected for whole-genome sequencing and all of them exhibited resistance to other fluoroquinonlones and showed an extended-spectrum beta-lactamase phenotype. The six delafloxacin resistant E. coli strains belonged to different sequence types (STs) namely, ST131 (2 strains), ST57 (2 strains), ST162 and ST15840. Each delafloxacin resistant strain possessed multiple mutations in quinolone resistance-determining regions (QRDRs). Notably, three mutations in gyrA Ser83Leu, Asp87Asn and parC Ser80Ile were in strains of ST162, ST57 and ST15840. However, the two strains of ST131 carried five combined mutations namely, gyrA Ser83Leu, Asp87Asn, parC Ser80Ile, Glu84Val, parE Ile549Leu. Association of delafloxacin resistance and production of CTX-M-15 in ST131, CMY-2 in ST162 and ST15840 was detected. In this study a new ST, ST15840 of clonal complex 69 was identified. Our results demonstrate, that at least three mutations in QRDRs are required for delafloxacin resistance in E. coli.
Collapse
Affiliation(s)
- András Kubicskó
- Institute of Medical Microbiology, Semmelweis University, 1089, Budapest, Hungary
| | - Katalin Kamotsay
- Central Microbiology Laboratory, National Institute of Hematology and Infectious Disease, Central Hospital of Southern-Pest, 1097, Budapest, Hungary
| | - Dóra Szabó
- Institute of Medical Microbiology, Semmelweis University, 1089, Budapest, Hungary
- HUN-REN-SU Human Microbiota Research Group, 1052, Budapest, Hungary
- Department of Neurosurgery and Neurointervention, Semmelweis University, 1083, Budapest, Hungary
| | - Béla Kocsis
- Institute of Medical Microbiology, Semmelweis University, 1089, Budapest, Hungary.
| |
Collapse
|
5
|
Chen B, Moriarty T, Steenackers H, Vles G, Onsea J, Vackier T, Spriet I, Lavigne R, Richards RG, Metsemakers WJ. Exploring the potential of naturally occurring antimicrobials for managing orthopedic-device-related infections. J Bone Jt Infect 2024; 9:249-260. [PMID: 39539734 PMCID: PMC11555427 DOI: 10.5194/jbji-9-249-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 08/29/2024] [Indexed: 11/16/2024] Open
Abstract
Orthopedic-device-related infections (ODRIs) are challenging clinical complications that are often exacerbated by antibiotic resistance and biofilm formation. This review explores the efficacy of naturally occurring antimicrobials - including agents sourced from bacteria, fungi, viruses, animals, plants and minerals - against pathogens common in ODRIs. The limitations of traditional antibiotic agents are presented, and innovative naturally occurring antimicrobials, such as bacteriophage therapy and antimicrobial peptides, are evaluated with respect to their interaction with conventional antibiotics and antibiofilm efficacy. The integration of these natural agents into clinical practice could revolutionize ODRI treatment strategies, offering effective alternatives to conventional antibiotics and mitigating resistance development. However, the translation of these compounds from research into the clinic may require the substantial investment of intellectual and financial resources.
Collapse
Affiliation(s)
- Baixing Chen
- Department of Trauma Surgery, University Hospitals Leuven, Leuven, Belgium
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | | | - Hans Steenackers
- Department of Microbial and Molecular Systems, Centre of Microbial and Plant Genetics (CMPG), KU Leuven, Leuven, Belgium
| | - Georges F. Vles
- Department of Orthopaedic Surgery, University Hospitals Leuven, Leuven, Belgium
- Institute for Orthopaedic Research and Training (IORT), KU Leuven, Leuven, Belgium
| | - Jolien Onsea
- Department of Trauma Surgery, University Hospitals Leuven, Leuven, Belgium
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Thijs Vackier
- Department of Microbial and Molecular Systems, Centre of Microbial and Plant Genetics (CMPG), KU Leuven, Leuven, Belgium
| | - Isabel Spriet
- Pharmacy Department, University Hospitals Leuven, Leuven, Belgium
- Pharmacotherapy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Belgium
| | - Rob Lavigne
- Laboratory of Gene Technology, Department of Biosystems, KU Leuven, Leuven, Belgium
| | | | - Willem-Jan Metsemakers
- Department of Trauma Surgery, University Hospitals Leuven, Leuven, Belgium
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| |
Collapse
|
6
|
Vladkova TG, Smani Y, Martinov BL, Gospodinova DN. Recent Progress in Terrestrial Biota Derived Antibacterial Agents for Medical Applications. Molecules 2024; 29:4889. [PMID: 39459256 PMCID: PMC11510244 DOI: 10.3390/molecules29204889] [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: 08/20/2024] [Revised: 10/07/2024] [Accepted: 10/13/2024] [Indexed: 10/28/2024] Open
Abstract
Conventional antibiotic and multidrug treatments are becoming less and less effective and the discovery of new effective and safe antibacterial agents is becoming a global priority. Returning to a natural antibacterial product is a relatively new current trend. Terrestrial biota is a rich source of biologically active substances whose antibacterial potential has not been fully utilized. The aim of this review is to present the current state-of-the-art terrestrial biota-derived antibacterial agents inspired by natural treatments. It summarizes the most important sources and newly identified or modified antibacterial agents and treatments from the last five years. It focuses on the significance of plant- animal- and bacteria-derived biologically active agents as powerful alternatives to antibiotics, as well as the advantages of utilizing natural antibacterial molecules alone or in combination with antibiotics. The main conclusion is that terrestrial biota-derived antibacterial products and substances open a variety of new ways for modern improved therapeutic strategies. New terrestrial sources of known antibacterial agents and new antibacterial agents from terrestrial biota were discovered during the last 5 years, which are under investigation together with some long-ago known but now experiencing their renaissance for the development of new medical treatments. The use of natural antibacterial peptides as well as combinational therapy by commercial antibiotics and natural products is outlined as the most promising method for treating bacterial infections. In vivo testing and clinical trials are necessary to reach clinical application.
Collapse
Affiliation(s)
- Todorka G. Vladkova
- Department of Polymer Engineering, University of Chemical Technology and Metallurgy, 8 “Kl. Ohridski” Blvd, 1756 Sofia, Bulgaria
| | - Younes Smani
- Andalusian Center of Developmental Biology, CSIC, Junta de Andalusia, University of Pablo de Olavide, 41013 Seville, Spain;
- Department of Molecular Biology and Biochemical Engineering, Andalusian Center of Developmental Biology, CSIC, Junta de Andalusia, University of Pablo de Olavide, 41013 Seville, Spain
| | - Boris L. Martinov
- Department of Biotechnology, University of Chemical Technology and Metallurgy, 8 “Kl. Ohridski” Blvd, 1756 Sofia, Bulgaria;
| | - Dilyana N. Gospodinova
- Faculty of Electrical Engineering, Technical University of Sofia, 8 “Kl. Ohridski” Blvd, 1756 Sofia, Bulgaria;
| |
Collapse
|
7
|
Ardila CM, Yadalam PK, González-Arroyave D. Integrating whole genome sequencing and machine learning for predicting antimicrobial resistance in critical pathogens: a systematic review of antimicrobial susceptibility tests. PeerJ 2024; 12:e18213. [PMID: 39399439 PMCID: PMC11470768 DOI: 10.7717/peerj.18213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 09/11/2024] [Indexed: 10/15/2024] Open
Abstract
Background Infections caused by antibiotic-resistant bacteria pose a major challenge to modern healthcare. This systematic review evaluates the efficacy of machine learning (ML) approaches in predicting antimicrobial resistance (AMR) in critical pathogens (CP), considering Whole Genome Sequencing (WGS) and antimicrobial susceptibility testing (AST). Methods The search covered databases including PubMed/MEDLINE, EMBASE, Web of Science, SCOPUS, and SCIELO, from their inception until June 2024. The review protocol was officially registered on PROSPERO (CRD42024543099). Results The review included 26 papers, analyzing data from 104,141 microbial samples. Random Forest (RF), XGBoost, and logistic regression (LR) emerged as the top-performing models, with mean Area Under the Receiver Operating Characteristic (AUC) values of 0.89, 0.87, and 0.87, respectively. RF showed superior performance with AUC values ranging from 0.66 to 0.97, while XGBoost and LR showed similar performance with AUC values ranging from 0.83 to 0.91 and 0.76 to 0.96, respectively. Most studies indicate that integrating WGS and AST data into ML models enhances predictive performance, improves antibiotic stewardship, and provides valuable clinical decision support. ML shows significant promise for predicting AMR by integrating WGS and AST data in CP. Standardized guidelines are needed to ensure consistency in future research.
Collapse
Affiliation(s)
- Carlos M. Ardila
- Basic Sciences Department, Faculty of Dentistry, Universidad de Antioquia, Medellin, Colombia
- CIFE University Center, Cuernavaca, Mexico
| | | | | |
Collapse
|
8
|
Ali-Khiavi P, Mohammadi M, Masoumi S, Saffarfar H, Kheradmand R, Mobed A, Hatefnia F. The Therapeutic Potential of Exosome Therapy in Sepsis Management: Addressing Complications and Improving Outcomes". Cell Biochem Biophys 2024:10.1007/s12013-024-01564-7. [PMID: 39363035 DOI: 10.1007/s12013-024-01564-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/17/2024] [Indexed: 10/05/2024]
Abstract
Infection occurs when pathogens penetrate tissues, reproduce, and trigger a host response to both the infectious agents and their toxins. A diverse array of pathogens, including viruses and bacteria, can cause infections. The host's immune system employs several mechanisms to combat these infections, typically involving an innate inflammatory response. Inflammation is a complex biological reaction that can affect various parts of the body and is a key component of the response to harmful stimuli. Sepsis arises when the body's response to infection leads to widespread damage to tissues and organs, potentially resulting in severe outcomes or death. The initial phase of sepsis involves immune system suppression. Early identification and targeted management are crucial for improving sepsis outcomes. Common treatment approaches include antibiotics, intravenous fluids, blood cultures, and monitoring urine output. This study explores the potential of exosome therapy in enhancing the management and alleviation of sepsis symptoms.
Collapse
Affiliation(s)
- Payam Ali-Khiavi
- Medical faculty, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahya Mohammadi
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sajjad Masoumi
- Department of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Hossein Saffarfar
- Cardiovascular Research Center, Tehran, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Kheradmand
- Social Determinants of Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahmad Mobed
- Social Determinants of Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Faezeh Hatefnia
- Social Determinants of Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
9
|
Schwenck LDC, Abreu PA, Nunes-da-Fonseca R. Spider's Silk as a Potential Source of Antibiotics: An Integrative Review. Probiotics Antimicrob Proteins 2024; 16:1608-1622. [PMID: 38460106 DOI: 10.1007/s12602-024-10241-3] [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] [Accepted: 02/27/2024] [Indexed: 03/11/2024]
Abstract
Spiders produce webs, which are still a largely unexplored source of antibacterial compounds, although the reports of its application in the medical field. Therefore, this study aims to present an integrative review of the antibacterial activity of spider webs. The research was conducted using Google Scholar, Scielo, Web of Science, PubMed, ScienceDirect, Medline EBSCO, LILACS, and Embase. The inclusion criteria were original articles written in English that studied the antibiotic properties of the web or isolated compounds tested. The studies were compared according to the spider species studied, the type of web, treatment of the sample, type of antimicrobial test, and the results obtained. Nine hundred and seventy-three publications were found, and after applying the inclusion and exclusion criteria, sixteen articles were selected. Bacterial inhibition was found in seven studies against various species of bacteria such as Escherichia coli, Klebsiella pneumoniae, Staphylococcus aureus, Salmonella Typhi, Bacillus megaterium, Listeria monocytogenes, Acinetobacter baumannii, Streptococcus pneumoniae, Pasteurella multocida, and Bacillus subtilis. Additionally, there was no apparent relationship between the proximity of the spider species evaluated in the studies and the presence or absence of activity. Methodological problems detected may affected the reproducibility and reliability of the results in some studies, such as the lack of description of the web or microorganism strain, as well as the absence of adequate controls and treatments to sterilize the sample. Spider webs can be a valuable source of antibiotics; however, more studies are needed to confirm the real activity of the web or components involved.
Collapse
Affiliation(s)
- Lucas da Costa Schwenck
- Instituto de Biodiversidade e Sustentabilidade-NUPEM, Universidade Federal do Rio de Janeiro (UFRJ), Av. São José do Barreto, 764, Macaé, Rio de Janeiro, CEP: 27920-560, Brazil
| | - Paula Alvarez Abreu
- Instituto de Biodiversidade e Sustentabilidade-NUPEM, Universidade Federal do Rio de Janeiro (UFRJ), Av. São José do Barreto, 764, Macaé, Rio de Janeiro, CEP: 27920-560, Brazil
| | - Rodrigo Nunes-da-Fonseca
- Instituto de Biodiversidade e Sustentabilidade-NUPEM, Universidade Federal do Rio de Janeiro (UFRJ), Av. São José do Barreto, 764, Macaé, Rio de Janeiro, CEP: 27920-560, Brazil.
| |
Collapse
|
10
|
Schmidt S, Täschner T, Nordholt N, Schreiber F. Differential Selection for Survival and for Growth in Adaptive Laboratory Evolution Experiments With Benzalkonium Chloride. Evol Appl 2024; 17:e70017. [PMID: 39399585 PMCID: PMC11470201 DOI: 10.1111/eva.70017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 06/14/2024] [Accepted: 09/01/2024] [Indexed: 10/15/2024] Open
Abstract
Biocides are used to control microorganisms across different applications, but emerging resistance may pose risks for those applications. Resistance to biocides has commonly been studied using adaptive laboratory evolution (ALE) experiments with growth at subinhibitory concentrations linked to serial subculturing. It has been shown recently that Escherichia coli adapts to repeated lethal stress imposed by the biocide benzalkonium chloride (BAC) by increased survival (i.e., tolerance) and not by evolving the ability to grow at increased concentrations (i.e., resistance). Here, we investigate the contributions of evolution for tolerance as opposed to resistance for the outcome of ALE experiments with E. coli exposed to BAC. We find that BAC concentrations close to the half maximal effective concentration (EC50, 4.36 μg mL-1) show initial killing (~40%) before the population resumes growth. This indicates that cells face a two-fold selection pressure: for increased survival and for increased growth. To disentangle the effects of both selection pressures, we conducted two ALE experiments: (i) one with initial killing and continued stress close to the EC50 during growth and (ii) another with initial killing and no stress during growth. Phenotypic characterization of adapted populations showed that growth at higher BAC concentrations was only selected for when BAC was present during growth. Whole genome sequencing revealed distinct differences in mutated genes across treatments. Treatments selecting for survival-only led to mutations in genes for metabolic regulation (cyaA) and cellular structure (flagella fliJ), while treatments selecting for growth and survival led to mutations in genes related to stress response (hslO and tufA). Our results demonstrate that serial subculture ALE experiments with an antimicrobial at subinhibitory concentrations can select for increased growth and survival. This finding has implications for the design of ALE experiments to assess resistance risks of antimicrobials in different scenarios such as disinfection, preservation, and environmental pollution.
Collapse
Affiliation(s)
- Selina B. I. Schmidt
- Department of Materials and the Environment, Division of Biodeterioration and Reference Organisms (4.1)Federal Institute for Materials Research and Testing (BAM)BerlinGermany
| | - Tom Täschner
- Department of Materials and the Environment, Division of Biodeterioration and Reference Organisms (4.1)Federal Institute for Materials Research and Testing (BAM)BerlinGermany
| | - Niclas Nordholt
- Department of Materials and the Environment, Division of Biodeterioration and Reference Organisms (4.1)Federal Institute for Materials Research and Testing (BAM)BerlinGermany
| | - Frank Schreiber
- Department of Materials and the Environment, Division of Biodeterioration and Reference Organisms (4.1)Federal Institute for Materials Research and Testing (BAM)BerlinGermany
| |
Collapse
|
11
|
Espino IN, Drolet J, Jones TN, Uwechue A, Koehler B, Beaird R, Maione S, Darrah C, Hijazi R, James C, Dupre A, Koscinski E, Creft L, Giampaolo M, Bernier A, Theisen KE. Computational docking of FtsZ: Survey of promising antibiotic compounds. Biochem Biophys Rep 2024; 39:101796. [PMID: 39687410 PMCID: PMC11647940 DOI: 10.1016/j.bbrep.2024.101796] [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: 01/26/2024] [Revised: 07/17/2024] [Accepted: 07/18/2024] [Indexed: 12/18/2024] Open
Abstract
The bacterial cell-division protein FtsZ has been a promising antibiotic target for over a decade now, but there is still a need for more work in this area. So far there are no FtsZ targeting drugs commercially available. We have analyzed a wide variety of prospective drugs and their interactions with multiple FtsZ species using both free and directed docking simulations. Our goal is to present a standardized computational screening method for potential drug compounds targeting FtsZ. Our work is an example of a way to compare many proposed drugs and FtsZ species combinations relatively quickly. A common method for comparison can yield new results that individual studies and varying methods might not show, as we demonstrate here. To our knowledge this is one of the first, if not the first, computational docking study on the new E. coli FtsZ structures obtained in 2020.
Collapse
Affiliation(s)
- Ileini N. Espino
- State University of New York at Plattsburgh, 101 Broad Street, Plattsburgh, 12901, NY, USA
| | - Julia Drolet
- State University of New York at Plattsburgh, 101 Broad Street, Plattsburgh, 12901, NY, USA
| | - Ty-niquia Jones
- State University of New York at Plattsburgh, 101 Broad Street, Plattsburgh, 12901, NY, USA
| | - Antonette Uwechue
- State University of New York at Plattsburgh, 101 Broad Street, Plattsburgh, 12901, NY, USA
| | - Brittany Koehler
- State University of New York at Plattsburgh, 101 Broad Street, Plattsburgh, 12901, NY, USA
| | - Raquel Beaird
- State University of New York at Plattsburgh, 101 Broad Street, Plattsburgh, 12901, NY, USA
| | - Sanni Maione
- State University of New York at Plattsburgh, 101 Broad Street, Plattsburgh, 12901, NY, USA
| | - Christine Darrah
- State University of New York at Plattsburgh, 101 Broad Street, Plattsburgh, 12901, NY, USA
| | - Rana Hijazi
- State University of New York at Plattsburgh, 101 Broad Street, Plattsburgh, 12901, NY, USA
| | - Christopher James
- State University of New York at Plattsburgh, 101 Broad Street, Plattsburgh, 12901, NY, USA
| | - Annabelle Dupre
- State University of New York at Plattsburgh, 101 Broad Street, Plattsburgh, 12901, NY, USA
| | - Ewa Koscinski
- State University of New York at Plattsburgh, 101 Broad Street, Plattsburgh, 12901, NY, USA
| | - Leilani Creft
- State University of New York at Plattsburgh, 101 Broad Street, Plattsburgh, 12901, NY, USA
| | - Michael Giampaolo
- State University of New York at Plattsburgh, 101 Broad Street, Plattsburgh, 12901, NY, USA
| | - Alexandre Bernier
- State University of New York at Plattsburgh, 101 Broad Street, Plattsburgh, 12901, NY, USA
| | - Kelly E. Theisen
- State University of New York at Plattsburgh, 101 Broad Street, Plattsburgh, 12901, NY, USA
| |
Collapse
|
12
|
Sher EK, Džidić-Krivić A, Sesar A, Farhat EK, Čeliković A, Beća-Zećo M, Pinjic E, Sher F. Current state and novel outlook on prevention and treatment of rising antibiotic resistance in urinary tract infections. Pharmacol Ther 2024; 261:108688. [PMID: 38972453 DOI: 10.1016/j.pharmthera.2024.108688] [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: 04/16/2024] [Accepted: 07/04/2024] [Indexed: 07/09/2024]
Abstract
Antibiotic-resistant bacteria are currently an important public health concern posing a serious threat due to their resistance to the current arsenal of antibiotics. Uropathogens Escherichia coli (UPEC), Proteus mirabilis, Klebsiella pneumoniae and Enterococcus faecalis, antibiotic-resistant gram-negative bacteria, cause serious cases of prolonged UTIs, increasing healthcare costs and potentially even leading to the death of an affected patient. This review discusses current knowledge about the increasing resistance to currently recommended antibiotics for UTI therapy, as well as novel therapeutic options. Traditional antibiotics are still a part of the therapy guidelines for UTIs, although they are often not effective and have serious side effects. Hence, novel drugs are being developed, such as combinations of β-lactam antibiotics with cephalosporins and carbapenems. Siderophoric cephalosporins, such as cefiderocol, have shown potential in the treatment of individuals with significant gram-negative bacterial infections, as well as aminoglycosides, fluoroquinolones and tetracyclines that are also undergoing clinical trials. The use of cranberry and probiotics is another potential curative and preventive method that has shown antimicrobial and anti-inflammatory effects. However, further studies are needed to assess the efficacy and safety of probiotics containing cranberry extract for UTI prevention and treatment. An emerging novel approach for UTI treatment is the use of immuno-prophylactic vaccines, as well as different nanotechnology solutions such as nanoparticles (NP). NP have the potential to be used as delivery systems for drugs to specific targets. Furthermore, nanotechnology could enable the development of nano antibiotics with improved features by the application of different NPs in their structure, such as gold and copper NPs. However, further high-quality research is required for the synthesis and testing of these novel molecules, such as safety evaluation and pharmacovigilance.
Collapse
Affiliation(s)
- Emina K Sher
- School of Science and Technology, Nottingham Trent University, Nottingham NG11 8NS, United Kingdom.
| | - Amina Džidić-Krivić
- Department of Neurology, Cantonal Hospital Zenica, Zenica 72000, Bosnia and Herzegovina; International Society of Engineering Science and Technology, Nottingham, United Kingdom
| | - Ana Sesar
- International Society of Engineering Science and Technology, Nottingham, United Kingdom; Faculty of Health Studies, Victoria International University, Mostar 88000, Bosnia and Herzegovina
| | - Esma K Farhat
- International Society of Engineering Science and Technology, Nottingham, United Kingdom; Faculty of Food Technology, Josip Juraj Strossmayer University of Osijek, Croatia
| | - Amila Čeliković
- International Society of Engineering Science and Technology, Nottingham, United Kingdom; Faculty of Medicine, University of Zenica, Zenica 71000, Bosnia and Herzegovina
| | - Merima Beća-Zećo
- International Society of Engineering Science and Technology, Nottingham, United Kingdom; Faculty of Health Studies, Victoria International University, Mostar 88000, Bosnia and Herzegovina
| | - Emma Pinjic
- Department of Radiology, Beth Israel Deaconess Medical Center (BIDMC), Boston, MA, United States
| | - Farooq Sher
- Department of Engineering, School of Science and Technology, Nottingham Trent University, Nottingham NG11 8NS, United Kingdom.
| |
Collapse
|
13
|
Labriola VF, Amaral LS, Perussi JR, Cavalheiro CCS, Azevedo EB. Nitrofurantoin removal by the photo-Fenton process: degradation, mineralization, and biological inactivation. ENVIRONMENTAL TECHNOLOGY 2024; 45:3418-3427. [PMID: 37204328 DOI: 10.1080/09593330.2023.2215940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 04/27/2023] [Indexed: 05/20/2023]
Abstract
Antibiotics may induce super-resistant bacteria if they are available in the environment. Therefore, the removal of aqueous nitrofurantoin (NFT), and more importantly, the removal of the remaining antimicrobial activity after treatment, by the photo-Fenton process, was herein studied. Degradation experiments were performed according to an experimental design (0.5% error; factors: concentrations of NFT, Fe3+, and H2O2). Degradation conditions were: 20 mg NFT L-1, 10 mg Fe3+ L-1, and 170 mg H2O2 L-1. Fixed parameters were: 100 mL of the NFT solution, pH 2.5, 15-min stirring, and 25.0 ± 0.5°C. The initial rate constant (k0) and the maximum oxidation capacity (MOC) of the system were 0.61 min-1 and 100%, respectively (R2 = 0.986). 97% of the NFT and 93% of the organic carbon initially present were removed. Five degradation products (DPs) were detected by HPLC-MS and their endpoints estimated by the ECOSAR (ECOlogical Structure-Activity Relationships) 2.0 software. NFT and its DPs presented no toxicity towards Lactuca sativa. The antimicrobial activity (Escherichia coli) of NFT and/or DPs was completely removed in 15 min. Structures were proposed for the detected DPs. In short, the tested advanced oxidation technology (AOP), besides being capable of removing and mineralizing aqueous NFT in a short time, 15 min, also rendered the treated water biologically inactive (no ecotoxicity, no antimicrobial activity).
Collapse
Affiliation(s)
- Vanessa Feltrin Labriola
- Laboratório de Desenvolvimento de Tecnologias Ambientais (LDTAmb), São Carlos Institute of Chemistry, University of São Paulo (USP), São Carlos/SP, Brazil
| | - Larissa Souza Amaral
- Grupo de Fotosensibilizadores, São Carlos Institute of Chemistry, University of São Paulo (USP), São Carlos/SP, Brazil
| | - Janice Rodrigues Perussi
- Grupo de Fotosensibilizadores, São Carlos Institute of Chemistry, University of São Paulo (USP), São Carlos/SP, Brazil
| | | | - Eduardo Bessa Azevedo
- Laboratório de Desenvolvimento de Tecnologias Ambientais (LDTAmb), São Carlos Institute of Chemistry, University of São Paulo (USP), São Carlos/SP, Brazil
| |
Collapse
|
14
|
Sanz-Gaitero M, De Maesschalck V, Patel A, Longin H, Van Noort V, Rodriguez-Rubio L, van Ryne M, Danis-Wlodarczyk K, Drulis-Kawa Z, Mesnage S, van Raaij M, Lavigne R. Structural and Biochemical Characterization of a New Phage-Encoded Muramidase, KTN6 Gp46. PHAGE (NEW ROCHELLE, N.Y.) 2024; 5:53-62. [PMID: 39119210 PMCID: PMC11304755 DOI: 10.1089/phage.2023.0040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Abstract
Background Endolysins are phage-encoded lytic enzymes that degrade bacterial peptidoglycan at the end of phage lytic cycles to release new phage particles. These enzymes are being explored as an alternative to small-molecule antibiotics. Methods The crystal structure of KTN6 Gp46 was determined and compared with a ColabFold model. Cleavage specificity was examined using a peptidoglycan digest and reversed-phase high-performance liquid chromatography coupled to mass spectrometry (HPLC/MS). Results The structure of KTN6 Gp46 could be determined at 1.4 Å resolution, and key differences in loops of the putative peptidoglycan binding domain were identified in comparison with its closest known homologue, the endolysin of phage SPN1S. Reversed-phase HPLC/MS analysis of the reaction products following peptidoglycan digestion confirmed the muramidase activity of Gp46, consistent with structural predictions. Conclusion These insights into the structure and function of endolysins further expand the toolbox for endolysin engineering and explore their potential in enzyme-based antibacterial design strategies.
Collapse
Affiliation(s)
- Marta Sanz-Gaitero
- Centro Nacional de Biotecnologia, Consejo Superior de Investigaciones Cientificas, Madrid, Spain
- Department of Biological Sciences, Munster Technological University, Cork, Ireland
| | | | - Ankur Patel
- School of Biosciences, University of Sheffield, Sheffield, United Kingdom
| | - Hannelore Longin
- Laboratory of Gene Technology, KU Leuven, Leuven, Belgium
- Laboratory of Computational Systems Biology, KU Leuven, Leuven, Belgium
| | - Vera Van Noort
- Laboratory of Computational Systems Biology, KU Leuven, Leuven, Belgium
| | | | | | - Katarzyna Danis-Wlodarczyk
- Laboratory of Gene Technology, KU Leuven, Leuven, Belgium
- Department of Pathogen Biology and Immunology, University of Wroclaw, Wroclaw, Poland
| | - Zuzanna Drulis-Kawa
- Department of Pathogen Biology and Immunology, University of Wroclaw, Wroclaw, Poland
| | - Stephane Mesnage
- School of Biosciences, University of Sheffield, Sheffield, United Kingdom
| | - Mark van Raaij
- Centro Nacional de Biotecnologia, Consejo Superior de Investigaciones Cientificas, Madrid, Spain
| | - Rob Lavigne
- Laboratory of Gene Technology, KU Leuven, Leuven, Belgium
| |
Collapse
|
15
|
Bahatheg G, Kuppusamy R, Yasir M, Bridge S, Mishra SK, Cranfield CG, StC Black D, Willcox M, Kumar N. Dimeric peptoids as antibacterial agents. Bioorg Chem 2024; 147:107334. [PMID: 38583251 DOI: 10.1016/j.bioorg.2024.107334] [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: 01/29/2024] [Revised: 03/31/2024] [Accepted: 04/02/2024] [Indexed: 04/09/2024]
Abstract
Building upon our previous study on peptoid-based antibacterials which showed good activity against Gram-positive bacteria only, herein we report the synthesis of 34 dimeric peptoid compounds and the investigation of their activity against Gram-positive and Gram-negative pathogens. The newly designed peptoids feature a di-hydrophobic moiety incorporating phenyl, bromo-phenyl, and naphthyl groups, combined with variable lengths of cationic units such as amino and guanidine groups. The study also underscores the pivotal interplay between hydrophobicity and cationicity in optimizing efficacy against specific bacteria. The bromophenyl dimeric guanidinium peptoid compound 10j showed excellent activity against S. aureus 38 and E. coli K12 with MIC of 0.8 μg mL-1 and 6.2 μg mL-1, respectively. Further investigation into the mechanism of action revealed that the antibacterial effect might be attributed to the disruption of bacterial cell membranes, as suggested by tethered bilayer lipid membranes (tBLMs) and cytoplasmic membrane permeability studies. Notably, these promising antibacterial agents exhibited negligible toxicity against mammalian red blood cells. Additionally, the study explored the potential of 12 active compounds to disrupt established biofilms of S. aureus 38. The most effective biofilm disruptors were ethyl and octyl-naphthyl guanidinium peptoids (10c and 10 k). These compounds 10c and 10 k disrupted the established biofilms of S. aureus 38 with 51 % at 4x MIC (MIC = 17.6 μg mL-1 and 11.2 μg mL-1) and 56 %-58 % at 8x MIC (MIC = 35.2 μg mL-1 and 22.4 μg mL-1) respectively. Overall, this research contributes insights into the design principles of cationic dimeric peptoids and their antibacterial activity, with implications for the development of new antibacterial compounds.
Collapse
Affiliation(s)
- Ghayah Bahatheg
- School of Chemistry, The University of New South Wales (UNSW), Sydney, NSW 2052, Australia; Department of Chemistry, Faculty of Science, University of Jeddah, Jeddah 21589, Saudi Arabia
| | - Rajesh Kuppusamy
- School of Chemistry, The University of New South Wales (UNSW), Sydney, NSW 2052, Australia; School of Optometry and Vision Science, The University of New South Wales (UNSW), Sydney, NSW 2052, Australia.
| | - Muhammad Yasir
- School of Optometry and Vision Science, The University of New South Wales (UNSW), Sydney, NSW 2052, Australia
| | - Samara Bridge
- School of Life Sciences, University of Technology Sydney, PO Box 123, Ultimo 2007, Australia
| | - Shyam K Mishra
- School of Optometry and Vision Science, The University of New South Wales (UNSW), Sydney, NSW 2052, Australia
| | - Charles G Cranfield
- School of Life Sciences, University of Technology Sydney, PO Box 123, Ultimo 2007, Australia
| | - David StC Black
- School of Chemistry, The University of New South Wales (UNSW), Sydney, NSW 2052, Australia
| | - Mark Willcox
- School of Optometry and Vision Science, The University of New South Wales (UNSW), Sydney, NSW 2052, Australia
| | - Naresh Kumar
- School of Chemistry, The University of New South Wales (UNSW), Sydney, NSW 2052, Australia.
| |
Collapse
|
16
|
Kurnia D, Padilah R, Apriyanti E, Dharsono HDA. Phytochemical Analysis and Anti-Biofilm Potential That Cause Dental Caries from Black Cumin Seeds ( Nigella sativa Linn.). Drug Des Devel Ther 2024; 18:1917-1932. [PMID: 38828022 PMCID: PMC11144408 DOI: 10.2147/dddt.s454217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 04/23/2024] [Indexed: 06/05/2024] Open
Abstract
The oral cavity is an excellent place for various microorganisms to grow. Spectrococcus mutans and Spectrococcus sanguinis are Gram-negative bacteria found in the oral cavity as pioneer biofilm formers on the tooth surface that cause caries. Caries treatment has been done with antibiotics and therapeutics, but the resistance level of S. mutans and S. sanguinis bacteria necessitates the exploration of new drug compounds. Black cumin (Nigella sativa Linn.) is known to contain secondary metabolites that have antioxidant, antibacterial, anti-biofilm, anti-inflammatory and antifungal activities. The purpose of this review article is to present data on the potential of Nigella sativa Linn seeds as anti-biofilm. This article will discuss biofilm-forming bacteria, the resistance mechanism of antibiotics, the bioactivity of N. sativa extracts and seed isolates together with the Structure Activity Relationship (SAR) review of N. sativa compound isolates. We collected data from reliable references that will illustrate the potential of N. sativa seeds as anti-biofilm drug.
Collapse
Affiliation(s)
- Dikdik Kurnia
- Department of Chemistry, Faculty of Mathematics and Natural Science, Universitas Padjadjaran, Sumedang, Indonesia
| | - Rizal Padilah
- Department of Chemistry, Faculty of Mathematics and Natural Science, Universitas Padjadjaran, Sumedang, Indonesia
| | - Eti Apriyanti
- Department of Chemistry, Faculty of Mathematics and Natural Science, Universitas Padjadjaran, Sumedang, Indonesia
| | - Hendra Dian Adhita Dharsono
- Department of Conservative Dentistry, Faculty of Dentistry, Universitas Padjadjaran, Bandung, Jawa Barat, Indonesia
| |
Collapse
|
17
|
Pogostin BH, Wu SX, Swierczynski MJ, Pennington C, Li SY, Vohidova D, Seeley EH, Agrawal A, Tang C, Cabler J, Dey A, Veiseh O, Nuermberger EL, Ball ZT, Hartgerink JD, McHugh KJ. Enhanced dynamic covalent chemistry for the controlled release of small molecules and biologics from a nanofibrous peptide hydrogel platform. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.21.595134. [PMID: 38826442 PMCID: PMC11142141 DOI: 10.1101/2024.05.21.595134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Maintaining safe and potent pharmaceutical drug levels is often challenging. Multidomain peptides (MDPs) assemble into supramolecular hydrogels with a well-defined, highly porous nanostructure that makes them attractive for drug delivery, yet their ability to extend release is typically limited by rapid drug diffusion. To overcome this challenge, we developed self-assembling boronate ester release (SABER) MDPs capable of engaging in dynamic covalent bonding with payloads containing boronic acids (BAs). As examples, we demonstrate that SABER hydrogels can prolong the release of five BA-containing small-molecule drugs as well as BA-modified insulin and antibodies. Pharmacokinetic studies revealed that SABER hydrogels extended the therapeutic effect of ganfeborole from days to weeks, preventing Mycobacterium tuberculosis growth better than repeated oral administration in an infection model. Similarly, SABER hydrogels extended insulin activity, maintaining normoglycemia for six days in diabetic mice after a single injection. These results suggest that SABER hydrogels present broad potential for clinical translation.
Collapse
|
18
|
Funke FJ, Schlee S, Sterner R. Validation of aminodeoxychorismate synthase and anthranilate synthase as novel targets for bispecific antibiotics inhibiting conserved protein-protein interactions. Appl Environ Microbiol 2024; 90:e0057224. [PMID: 38700332 PMCID: PMC11107160 DOI: 10.1128/aem.00572-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 04/08/2024] [Indexed: 05/05/2024] Open
Abstract
Multi-resistant bacteria are a rapidly emerging threat to modern medicine. It is thus essential to identify and validate novel antibacterial targets that promise high robustness against resistance-mediating mutations. This can be achieved by simultaneously targeting several conserved function-determining protein-protein interactions in enzyme complexes from prokaryotic primary metabolism. Here, we selected two evolutionary related glutamine amidotransferase complexes, aminodeoxychorismate synthase and anthranilate synthase, that are required for the biosynthesis of folate and tryptophan in most prokaryotic organisms. Both enzymes rely on the interplay of a glutaminase and a synthase subunit that is conferred by a highly conserved subunit interface. Consequently, inhibiting subunit association in both enzymes by one competing bispecific inhibitor has the potential to suppress bacterial proliferation. We comprehensively verified two conserved interface hot-spot residues as potential inhibitor-binding sites in vitro by demonstrating their crucial role in subunit association and enzymatic activity. For in vivo target validation, we generated genomically modified Escherichia coli strains in which subunit association was disrupted by modifying these central interface residues. The growth of such strains was drastically retarded on liquid and solid minimal medium due to a lack of folate and tryptophan. Remarkably, the bacteriostatic effect was observed even in the presence of heat-inactivated human plasma, demonstrating that accessible host metabolite concentrations do not compensate for the lack of folate and tryptophan within the tested bacterial cells. We conclude that a potential inhibitor targeting both enzyme complexes will be effective against a broad spectrum of pathogens and offer increased resilience against antibiotic resistance. IMPORTANCE Antibiotics are indispensable for the treatment of bacterial infections in human and veterinary medicine and are thus a major pillar of modern medicine. However, the exposure of bacteria to antibiotics generates an unintentional selective pressure on bacterial assemblies that over time promotes the development or acquisition of resistance mechanisms, allowing pathogens to escape the treatment. In that manner, humanity is in an ever-lasting race with pathogens to come up with new treatment options before resistances emerge. In general, antibiotics with novel modes of action require more complex pathogen adaptations as compared to chemical derivates of existing entities, thus delaying the emergence of resistance. In this contribution, we use modified Escherichia coli strains to validate two novel targets required for folate and tryptophan biosynthesis that can potentially be targeted by one and the same bispecific protein-protein interaction inhibitor and promise increased robustness against bacterial resistances.
Collapse
Affiliation(s)
- Franziska Jasmin Funke
- Institute of Biophysics and Physical Biochemistry, Regensburg Center for Biochemistry, University of Regensburg, Regensburg, Germany
| | - Sandra Schlee
- Institute of Biophysics and Physical Biochemistry, Regensburg Center for Biochemistry, University of Regensburg, Regensburg, Germany
| | - Reinhard Sterner
- Institute of Biophysics and Physical Biochemistry, Regensburg Center for Biochemistry, University of Regensburg, Regensburg, Germany
| |
Collapse
|
19
|
Galani I, Souli M, Katsala D, Karaiskos I, Giamarellou H, Antoniadou A. In vitro activity of apramycin (EBL-1003) in combination with colistin, meropenem, minocycline or sulbactam against XDR/PDR Acinetobacter baumannii isolates from Greece. J Antimicrob Chemother 2024; 79:1101-1108. [PMID: 38501368 PMCID: PMC11062935 DOI: 10.1093/jac/dkae077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 03/01/2024] [Indexed: 03/20/2024] Open
Abstract
OBJECTIVES To evaluate the in vitro activity of the combination of apramycin with colistin, meropenem, minocycline or sulbactam, against some well-characterized XDR Acinetobacter baumannii clinical isolates from Greece, to understand how apramycin can be best incorporated into clinical practice and optimize effectiveness. METHODS In vitro interactions of apramycin (0.5×, 1× and 2× the MIC value) with colistin (2 mg/L), meropenem (30 mg/L), minocycline (3.5 mg/L) or sulbactam (24 mg/L) were tested using time-kill methodology. Twenty-one clinical A. baumannii isolates were chosen, exhibiting apramycin MICs of 4-16 mg/L, which were at or below the apramycin preliminary epidemiological cut-off value of 16 mg/L. These isolates were selected for a range of colistin (4-32 mg/L), meropenem (16-256 mg/L), minocycline (8-32 mg/L) and sulbactam (8-32 mg/L) MICs across the resistant range. Synergy was defined as a ≥2 log10 cfu/mL reduction compared with the most active agent. RESULTS The combination of apramycin with colistin, meropenem, minocycline or sulbactam was synergistic, at least at one of the concentrations of apramycin (0.5×, 1× or 2× MIC), against 83.3%, 90.5%, 90.9% or 92.3% of the tested isolates, respectively. Apramycin alone was bactericidal at 24 h against 9.5% and 33.3% of the tested isolates at concentrations equal to 1× and 2× MIC, while the combination of apramycin at 2× MIC with colistin, meropenem or sulbactam was bactericidal against all isolates tested (100%). The apramycin 2× MIC/minocycline combination had bactericidal activity against 90.9% of the tested isolates. CONCLUSIONS Apramycin combinations may have potential as a treatment option for XDR/pandrug-resistant (PDR) A. baumannii infections and warrant validation in the clinical setting, when this new aminoglycoside is available for clinical use.
Collapse
Affiliation(s)
- I Galani
- Infectious Diseases Laboratory, 4th Department of Internal Medicine, National and Kapodistrian University of Athens, School of Medicine, University General Hospital ‘ATTIKON’, Rimini 1, 124 62 Chaidari, Athens, Greece
| | - M Souli
- Duke Clinical Research Institute, Duke University Medical Center, Durham, NC, USA
| | - D Katsala
- Infectious Diseases Laboratory, 4th Department of Internal Medicine, National and Kapodistrian University of Athens, School of Medicine, University General Hospital ‘ATTIKON’, Rimini 1, 124 62 Chaidari, Athens, Greece
| | - I Karaiskos
- 1st Department of Internal Medicine & Infectious Diseases, Hygeia General Hospital, Athens, Greece
| | - H Giamarellou
- 1st Department of Internal Medicine & Infectious Diseases, Hygeia General Hospital, Athens, Greece
| | - A Antoniadou
- Infectious Diseases Laboratory, 4th Department of Internal Medicine, National and Kapodistrian University of Athens, School of Medicine, University General Hospital ‘ATTIKON’, Rimini 1, 124 62 Chaidari, Athens, Greece
- University of Cyprus, Medical School, Nicosia, Cyprus
| |
Collapse
|
20
|
Zhou Y, Song Y, Zhang Y, Liu X, Liu L, Bao Y, Wang J, Yang L. Azalomycin F4a targets peptidoglycan synthesis of Gram-positive bacteria revealed by high-throughput CRISPRi-seq analysis. Microbiol Res 2024; 280:127584. [PMID: 38157688 DOI: 10.1016/j.micres.2023.127584] [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: 11/18/2023] [Revised: 12/11/2023] [Accepted: 12/14/2023] [Indexed: 01/03/2024]
Abstract
Azalomycin F4a is a promising 36-membered polyhydroxy macrolide that shows antibacterial activity against drug-resistant Gram-positive bacteria, but its exact working mechanism remains to be elusive. Here, we isolated the azalomycin F4a product from a Streptomyces solisilvae and demonstrated its antibacterial activity against Gram-positive pathogens including Streptococcus pneumoniae, Staphylococcus aureus and methicillin-resistant Staphylococcus aureus (MRSA). We further showed that combination of azalomycin F4a with methicillin has an additive antimicrobial effect on MRSA, where the minimal inhibitory concentrations (MIC) of methicillin to MRSA was decreased by 1000-fold in the presence of sublethal concentration of azalomycin F4a. A CRISPRi-seq based whole genome screen was employed to identify the potential targets of azalomycin F4a, which revealed that peptidoglycan synthesis (PGS) was inhibited by azalomycin F4a. Furthermore, azalomycin F4a treatment could significantly impair S. aureus biofilm formation. Our research highlights that cell wall synthesis is an additional target for novel classes of macrolide besides ribosome.
Collapse
Affiliation(s)
- Yachun Zhou
- School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; Shenzhen Third People's Hospital, National Clinical Research Centre for Infectious Disease, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen 518112, China
| | - Yue Song
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology/Guangdong Key Laboratory of Marine Materia Medica, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yu Zhang
- Department of Pathogen Biology, Base for International Science and Technology Cooperation: Carson Cancer Stem Cell Vaccines R&D Centre, International Cancer Centre, Shenzhen University Health Science Centre, Shenzhen 518055, China
| | - Xue Liu
- Department of Pathogen Biology, Base for International Science and Technology Cooperation: Carson Cancer Stem Cell Vaccines R&D Centre, International Cancer Centre, Shenzhen University Health Science Centre, Shenzhen 518055, China
| | - Lei Liu
- Shenzhen Third People's Hospital, National Clinical Research Centre for Infectious Disease, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen 518112, China.
| | - Yanmin Bao
- Department of Respiratory Diseases, Shenzhen Children's Hospital, Shenzhen 518031, Guangdong, China
| | - Junfeng Wang
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology/Guangdong Key Laboratory of Marine Materia Medica, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Liang Yang
- School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; Shenzhen Third People's Hospital, National Clinical Research Centre for Infectious Disease, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen 518112, China.
| |
Collapse
|
21
|
Robotham JV, Tacconelli E, Vella V, de Kraker MEA. Synthesizing pathogen- and infection-specific estimates of the burden of antimicrobial resistance in Europe for health-technology assessment: gaps, heterogeneity, and bias. Clin Microbiol Infect 2024; 30 Suppl 1:S1-S3. [PMID: 38007386 DOI: 10.1016/j.cmi.2023.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 10/01/2023] [Accepted: 10/05/2023] [Indexed: 11/27/2023]
Affiliation(s)
- Julie V Robotham
- Healthcare Associated Infection, Fungal, Antimicrobial Resistance, Antimicrobial Usage & Sepsis Division, United Kingdom Health Security Agency, London, UK
| | - Evelina Tacconelli
- Infectious Diseases, Department of Diagnostic and Public Health, University of Verona, Verona, Italy
| | | | - Marlieke E A de Kraker
- Infection Control Program, Geneva University Hospitals and Faculty of Medicine, WHO Collaborating Center, Geneva, Switzerland.
| |
Collapse
|
22
|
Zhao S, Wang D, Zhou Q, Wang B, Tong Z, Tian H, Li J, Zhang Y. Nanozyme-based inulin@nanogold for adhesive and antibacterial agent with enhanced biosafety. Int J Biol Macromol 2024; 262:129207. [PMID: 38185305 DOI: 10.1016/j.ijbiomac.2024.129207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 10/26/2023] [Accepted: 01/01/2024] [Indexed: 01/09/2024]
Abstract
Nanozymes with oxidase or peroxidase-mimicking activity have emerged as a promising alternative for disinfecting resistant pathogens. However, further research and clinical applications of nanozymes are hampered by their low in vivo biosafety and biocompatibility. In this study, inulin-confined gold nanoparticles (IN@AuNP) are synthesized as an antibacterial agent via a straightforward in situ reduction of Au3+ ions by the hydroxyl groups in inulin. The IN@AuNP exhibits both peroxidase-mimicking and oxidase-mimicking catalytic activities, of which the maximum reaction velocity (Vmax) for H2O2 is 2.66 times higher than that of horseradish peroxidase. IN@AuNP can catalyze the production of reactive oxygen species (ROS), resulting in effective antibacterial behavior against both Gram-negative (Escherichia coli) and Gram-positive (Staphylococcus aureus) bacteria. Abundant hydroxyl groups retained in inulin endow the nanozyme with high adhesion to bacteria, reducing the distance between the captured bacteria and ROS, achieving an antibacterial ratio of 100 % within 1 h. Importantly, due to the natural biosafety and non-absorption of the dietary fiber inulin, as well as the inability of inulin-trapped AuNP to diffuse, the IN@AuNP exhibits high biosafety and biocompatibility under physiological conditions. This work is expected to open a new avenue for nanozymes with great clinical application value.
Collapse
Affiliation(s)
- Shiwen Zhao
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China; University Key Laboratory of Food Processing Byproducts for Advanced Development and High Value Utilization, Xi'an 710119, China
| | - Danyang Wang
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China; University Key Laboratory of Food Processing Byproducts for Advanced Development and High Value Utilization, Xi'an 710119, China
| | | | - Beibei Wang
- Xi'an Aerospace Chemical Propulsion Co., Ltd., Xi'an 710025, China
| | - Zhao Tong
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Honglei Tian
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China.
| | - Jianke Li
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China; University Key Laboratory of Food Processing Byproducts for Advanced Development and High Value Utilization, Xi'an 710119, China.
| | - Yuhuan Zhang
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China; University Key Laboratory of Food Processing Byproducts for Advanced Development and High Value Utilization, Xi'an 710119, China.
| |
Collapse
|
23
|
Wang M, Zhang J, Wei J, Jiang L, Jiang L, Sun Y, Zeng Z, Wang Z. Phage-inspired strategies to combat antibacterial resistance. Crit Rev Microbiol 2024; 50:196-211. [PMID: 38400715 DOI: 10.1080/1040841x.2023.2181056] [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: 12/09/2022] [Accepted: 02/07/2023] [Indexed: 02/25/2023]
Abstract
Antimicrobial resistance (AMR) in clinically priority pathogensis now a major threat to public health worldwide. Phages are bacterial parasites that efficiently infect or kill specific strains and represent the most abundant biological entities on earth, showing great attraction as potential antibacterial therapeutics in combating AMR. This review provides a summary of phage-inspired strategies to combat AMR. We firstly cover the phage diversity, and then explain the biological principles of phage therapy that support the use of phages in the post-antimicrobial era. Furthermore, we state the versatility methods of phage therapy both from direct access as well as collateral access. Among the direct access approaches, we discuss the use of phage cocktail therapy, phage-encoded endolysins and the bioengineering for function improvement of used phages or endolysins. On the other hand, we introduce the collateral access, including the phages antimicrobial immunity combined therapy and phage-based novel antibacterial mimic molecules. Nowadays, more and more talented and enthusiastic scientist, doctors, pharmacists, media, authorities, and industry are promoting the progress of phage therapy, and proposed more phages-inspired strategy to make them more tractable to combat AMR and benefit more people, more animal and diverse environment in "one health" framework.
Collapse
Affiliation(s)
- Mianzhi Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Junxuan Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - Jingyi Wei
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Lei Jiang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Li Jiang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Yongxue Sun
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - Zhenling Zeng
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - Zhiqiang Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
- International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou, China
| |
Collapse
|
24
|
De K, Dey R, Acharya Y, Aswal VK, Haldar J. Cleavable Amphiphilic Biocides with Ester-Bearing Moieties: Aggregation Properties and Antibacterial Activity. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024. [PMID: 38324708 DOI: 10.1021/acs.langmuir.3c02771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
The rise of multidrug-resistant bacterial infections and the dwindling supply of newly approved antibiotics have emerged as a grave threat to public health. Toward the ever-growing necessity of the development of novel antimicrobial agents, herein, we synthesized a series of cationic amphiphilic biocides featuring two cationic headgroups separated by different hydrophobic spacers, accompanied by the inclusion of two lipophilic tails through cleavable ester functionality. The detailed aggregation properties offered by these biocides were investigated by small-angle neutron scattering (SANS) and conductivity. The critical micellar concentration of the biocides and the size and shape of the micellar aggregates differed with variation of pendant and spacer hydrophobicity. Furthermore, the aggregation number and size of the micelles were found to vary with changing concentration and temperature. These easily synthesized biocides exhibited potent antibacterial properties against various multidrug-resistant bacteria. The optimized biocides with minimum hematotoxicity and potent antibacterial activity against methicillin-resistant Staphylococcus aureus and Acinetobacter baumannii exhibited rapid killing kinetics against planktonic bacteria. Also, these membrane-active agents were able to eradicate preformed biofilms. The enzymatic and acidic degradation profile further offered proof of gradual degradation. Collectively, these cleavable amphiphilic biocides demonstrated excellent potency for combating the multidrug-resistant bacterial infection.
Collapse
Affiliation(s)
- Kathakali De
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur, Bengaluru 560064, Karnataka, India
| | - Rajib Dey
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur, Bengaluru 560064, Karnataka, India
| | - Yash Acharya
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur, Bengaluru 560064, Karnataka, India
| | - Vinod Kumar Aswal
- Solid State Physics Division, Bhabha Atomic Research Centre (BARC), Mumbai 400085, Maharashtra, India
| | - Jayanta Haldar
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur, Bengaluru 560064, Karnataka, India
- School of Advanced Materials, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur, Bengaluru 560064, Karnataka, India
| |
Collapse
|
25
|
Abraham E, Lawther HA, Wang Y, Zarins-Tutt JS, Rivera GS, Wu C, Connolly JA, Florence G, Agbo M, Gao H, Goss RJM. The Identification and Heterologous Expression of the Biosynthetic Gene Cluster Encoding the Antibiotic and Anticancer Agent Marinomycin. Biomolecules 2024; 14:117. [PMID: 38254717 PMCID: PMC10813093 DOI: 10.3390/biom14010117] [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: 12/19/2023] [Revised: 12/31/2023] [Accepted: 01/04/2024] [Indexed: 01/24/2024] Open
Abstract
With the rise in antimicrobial resistance, there is an urgent need for new classes of antibiotic with which to treat infectious disease. Marinomycin, a polyene antibiotic from a marine microbe, has been shown capable of killing methicillin-resistant Staphylococcus aureus (MRSA) and vancomycin-resistant Enterococcus faecium (VREF), as well as having promising activity against melanoma. An attractive solution to the photoprotection of this antibiotic has been demonstrated. Here, we report the identification and analysis of the marinomycin biosynthetic gene cluster (BGC), and the biosynthetic assembly of the macrolide. The marinomycin BGC presents a challenge in heterologous expression due to its large size and high GC content, rendering the cluster prone to rearrangement. We demonstrate the transformation of Streptomyces lividans using a construct containing the cluster, and the heterologous expression of the encoded biosynthetic machinery and production of marinomycin B.
Collapse
Affiliation(s)
- Emily Abraham
- Department of Chemistry & BSRC, University of St. Andrews, St. Andrews KY16 9ST, UK; (E.A.); (J.A.C.)
| | - Hannah A. Lawther
- Department of Chemistry & BSRC, University of St. Andrews, St. Andrews KY16 9ST, UK; (E.A.); (J.A.C.)
| | - Yunpeng Wang
- Department of Chemistry & BSRC, University of St. Andrews, St. Andrews KY16 9ST, UK; (E.A.); (J.A.C.)
| | - Joseph S. Zarins-Tutt
- Department of Chemistry & BSRC, University of St. Andrews, St. Andrews KY16 9ST, UK; (E.A.); (J.A.C.)
| | | | - Chengcang Wu
- Intact Genomics, St. Louis, MO 63132, USA (C.W.)
| | - Jack A. Connolly
- Department of Chemistry & BSRC, University of St. Andrews, St. Andrews KY16 9ST, UK; (E.A.); (J.A.C.)
| | - Gordon Florence
- Department of Chemistry & BSRC, University of St. Andrews, St. Andrews KY16 9ST, UK; (E.A.); (J.A.C.)
| | - Matthias Agbo
- Department of Chemistry & BSRC, University of St. Andrews, St. Andrews KY16 9ST, UK; (E.A.); (J.A.C.)
| | - Hong Gao
- Department of Chemistry & BSRC, University of St. Andrews, St. Andrews KY16 9ST, UK; (E.A.); (J.A.C.)
| | - Rebecca J. M. Goss
- Department of Chemistry & BSRC, University of St. Andrews, St. Andrews KY16 9ST, UK; (E.A.); (J.A.C.)
| |
Collapse
|
26
|
Nazli A, Tao W, You H, He X, He Y. Treatment of MRSA Infection: Where are We? Curr Med Chem 2024; 31:4425-4460. [PMID: 38310393 DOI: 10.2174/0109298673249381231130111352] [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: 02/19/2023] [Revised: 07/10/2023] [Accepted: 10/10/2023] [Indexed: 02/05/2024]
Abstract
Staphylococcus aureus is a leading cause of septicemia, endocarditis, pneumonia, skin and soft tissue infections, bone and joint infections, and hospital-acquired infections. In particular, methicillin-resistant Staphylococcus aureus (MRSA) is associated with high morbidity and mortality, and continues to be a major public health problem. The emergence of multidrug-resistant MRSA strains along with the wide consumption of antibiotics has made anti-MRSA treatment a huge challenge. Novel treatment strategies (e.g., novel antimicrobials and new administrations) against MRSA are urgently needed. In the past decade, pharmaceutical companies have invested more in the research and development (R&D) of new antimicrobials and strategies, spurred by favorable policies. All research articles were collected from authentic online databases, including Google Scholar, PubMed, Scopus, and Web of Science, by using different combinations of keywords, including 'anti-MRSA', 'antibiotic', 'antimicrobial', 'clinical trial', 'clinical phase', clinical studies', and 'pipeline'. The information extracted from articles was compared to information provided on the drug manufacturer's website and Clinical Trials.gov (https://clinicaltrials.gov/) to confirm the latest development phase of anti-MRSA agents. The present review focuses on the current development status of new anti-MRSA strategies concerning chemistry, pharmacological target(s), indications, route of administration, efficacy and safety, pharmacokinetics, and pharmacodynamics, and aims to discuss the challenges and opportunities in developing drugs for anti-MRSA infections.
Collapse
Affiliation(s)
- Adila Nazli
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, China
| | - Wenlan Tao
- Chongqing School, University of Chinese Academy of Sciences (UCAS Chongqing), Chongqing, 400714, China
| | - Hengyao You
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, China
| | - Xiaoli He
- Chongqing School, University of Chinese Academy of Sciences (UCAS Chongqing), Chongqing, 400714, China
| | - Yun He
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, China
| |
Collapse
|
27
|
Faleye OS, Boya BR, Lee JH, Choi I, Lee J. Halogenated Antimicrobial Agents to Combat Drug-Resistant Pathogens. Pharmacol Rev 2023; 76:90-141. [PMID: 37845080 DOI: 10.1124/pharmrev.123.000863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 08/07/2023] [Accepted: 09/29/2023] [Indexed: 10/18/2023] Open
Abstract
Antimicrobial resistance presents us with a potential global crisis as it undermines the abilities of conventional antibiotics to combat pathogenic microbes. The history of antimicrobial agents is replete with examples of scaffolds containing halogens. In this review, we discuss the impacts of halogen atoms in various antibiotic types and antimicrobial scaffolds and their modes of action, structure-activity relationships, and the contributions of halogen atoms in antimicrobial activity and drug resistance. Other halogenated molecules, including carbohydrates, peptides, lipids, and polymeric complexes, are also reviewed, and the effects of halogenated scaffolds on pharmacokinetics, pharmacodynamics, and factors affecting antimicrobial and antivirulence activities are presented. Furthermore, the potential of halogenation to circumvent antimicrobial resistance and rejuvenate impotent antibiotics is addressed. This review provides an overview of the significance of halogenation, the abilities of halogens to interact in biomolecular settings and enhance pharmacological properties, and their potential therapeutic usages in preventing a postantibiotic era. SIGNIFICANCE STATEMENT: Antimicrobial resistance and the increasing impotence of antibiotics are critical threats to global health. The roles and importance of halogen atoms in antimicrobial drug scaffolds have been established, but comparatively little is known of their pharmacological impacts on drug resistance and antivirulence activities. This review is the first to extensively evaluate the roles of halogen atoms in various antibiotic classes and pharmacological scaffolds and to provide an overview of their ability to overcome antimicrobial resistance.
Collapse
Affiliation(s)
- Olajide Sunday Faleye
- School of Chemical Engineering (O.S.F., B.R.B., J.-H.L., J.L.) and Department of Medical Biotechnology (I.C.), Yeungnam University, Gyeongsan, Republic of Korea
| | - Bharath Reddy Boya
- School of Chemical Engineering (O.S.F., B.R.B., J.-H.L., J.L.) and Department of Medical Biotechnology (I.C.), Yeungnam University, Gyeongsan, Republic of Korea
| | - Jin-Hyung Lee
- School of Chemical Engineering (O.S.F., B.R.B., J.-H.L., J.L.) and Department of Medical Biotechnology (I.C.), Yeungnam University, Gyeongsan, Republic of Korea
| | - Inho Choi
- School of Chemical Engineering (O.S.F., B.R.B., J.-H.L., J.L.) and Department of Medical Biotechnology (I.C.), Yeungnam University, Gyeongsan, Republic of Korea
| | - Jintae Lee
- School of Chemical Engineering (O.S.F., B.R.B., J.-H.L., J.L.) and Department of Medical Biotechnology (I.C.), Yeungnam University, Gyeongsan, Republic of Korea
| |
Collapse
|
28
|
Weng C, Tan YLK, Koh WG, Ang WH. Harnessing Transition Metal Scaffolds for Targeted Antibacterial Therapy. Angew Chem Int Ed Engl 2023; 62:e202310040. [PMID: 37621226 DOI: 10.1002/anie.202310040] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/24/2023] [Accepted: 08/24/2023] [Indexed: 08/26/2023]
Abstract
Antimicrobial resistance, caused by persistent adaptation and growing resistance of pathogenic bacteria to overprescribed antibiotics, poses one of the most serious and urgent threats to global public health. The limited pipeline of experimental antibiotics in development further exacerbates this looming crisis and new drugs with alternative modes of action are needed to tackle evolving pathogenic adaptation. Transition metal complexes can replenish this diminishing stockpile of drug candidates by providing compounds with unique properties that are not easily accessible using pure organic scaffolds. We spotlight four emerging strategies to harness these unique properties to develop new targeted antibacterial agents.
Collapse
Affiliation(s)
- Cheng Weng
- Department of Chemistry, National University of Singapore, 4 Science Drive 2, Singapore, 117544, Singapore
| | | | - Wayne Gareth Koh
- Department of Chemistry, National University of Singapore, 4 Science Drive 2, Singapore, 117544, Singapore
| | - Wee Han Ang
- Department of Chemistry, National University of Singapore, 4 Science Drive 2, Singapore, 117544, Singapore
- NUS Graduate School of Integrative Sciences and Engineering, 28 Medical Drive, Singapore, 117456, Singapore
| |
Collapse
|
29
|
Pisoni LA, Semple SJ, Liu S, Sykes MJ, Venter H. Combined Structure- and Ligand-Based Approach for the Identification of Inhibitors of AcrAB-TolC in Escherichia coli. ACS Infect Dis 2023; 9:2504-2522. [PMID: 37888944 DOI: 10.1021/acsinfecdis.3c00350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
The inhibition of efflux pumps is a promising approach to combating multidrug-resistant bacteria. We have developed a combined structure- and ligand-based model, using OpenEye software, for the identification of inhibitors of AcrB, the inner membrane protein component of the AcrAB-TolC efflux pump in Escherichia coli. From a database of 1391 FDA-approved drugs, 23 compounds were selected to test for efflux inhibition in E. coli. Seven compounds, including ivacaftor (25), butenafine (19), naftifine (27), pimozide (30), thioridazine (35), trifluoperazine (37), and meloxicam (26), enhanced the activity of at least one antimicrobial substrate and inhibited the efflux pump-mediated removal of the substrate Nile Red from cells. Ivacaftor (25) inhibited efflux dose dependently, had no effect on an E. coli strain with genomic deletion of the gene encoding AcrB, and did not damage the bacterial outer membrane. In the presence of a sub-minimum inhibitory concentration (MIC) of the outer membrane permeabilizer colistin, ivacaftor at 1 μg/mL reduced the MICs of erythromycin and minocycline by 4- to 8-fold. The identification of seven potential AcrB inhibitors shows the merits of a combined structure- and ligand-based approach to virtual screening.
Collapse
Affiliation(s)
- Lily A Pisoni
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, South Australia 5000, Australia
- Quality Use of Medicines and Pharmacy Research Centre, Clinical and Health Sciences, University of South Australia, Adelaide, South Australia 5000, Australia
| | - Susan J Semple
- Quality Use of Medicines and Pharmacy Research Centre, Clinical and Health Sciences, University of South Australia, Adelaide, South Australia 5000, Australia
| | - Sida Liu
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, South Australia 5000, Australia
| | - Matthew J Sykes
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, South Australia 5000, Australia
| | - Henrietta Venter
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, South Australia 5000, Australia
| |
Collapse
|
30
|
Cebrián R, Lucas R, Fernández-Cantos MV, Slot K, Peñalver P, Martínez-García M, Párraga-Leo A, de Paz MV, García F, Kuipers OP, Morales JC. Synthesis and antimicrobial activity of aminoalkyl resveratrol derivatives inspired by cationic peptides. J Enzyme Inhib Med Chem 2023; 38:267-281. [PMID: 36600674 PMCID: PMC9828810 DOI: 10.1080/14756366.2022.2146685] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Antimicrobial resistance is a global concern, far from being resolved. The need of new drugs against new targets is imminent. In this work, we present a family of aminoalkyl resveratrol derivatives with antibacterial activity inspired by the properties of cationic amphipathic antimicrobial peptides. Surprisingly, the newly designed molecules display modest activity against aerobically growing bacteria but show surprisingly good antimicrobial activity against anaerobic bacteria (Gram-negative and Gram-positive) suggesting specificity towards this bacterial group. Preliminary studies into the action mechanism suggest that activity takes place at the membrane level, while no cross-resistance with traditional antibiotics is observed. Actually, some good synergistic relations with existing antibiotics were found against Gram-negative pathogens. However, some cytotoxicity was observed, despite their low haemolytic activity. Our results show the importance of the balance between positively charged moieties and hydrophobicity to improve antimicrobial activity, setting the stage for the design of new drugs based on these molecules.
Collapse
Affiliation(s)
- Rubén Cebrián
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, The Netherlands,Department of Clinical Microbiology, Instituto de Investigación Biosanitaria ibs. GRANADA, University Hospital Clínico San Cecilio, Granada, Spain,CONTACT Rubén Cebrián University Hospital San Cecilio,Clinical Microbiology Department, Av. de la Innovación s/n, 18061, Granada, Spain
| | - Ricardo Lucas
- Department of Organic and Pharmaceutical Chemistry, School of Pharmacy, University of Seville, Seville, Spain
| | - María Victoria Fernández-Cantos
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, The Netherlands
| | - Koen Slot
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, The Netherlands
| | - Pablo Peñalver
- Department of Biochemistry and Molecular Pharmacology, Instituto de Parasitología y Biomedicina López Neyra, CSIC, PTS Granada, Armilla, Granada, Spain
| | - Marta Martínez-García
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, The Netherlands
| | - Antonio Párraga-Leo
- Department of Organic and Pharmaceutical Chemistry, School of Pharmacy, University of Seville, Seville, Spain
| | - María Violante de Paz
- Department of Organic and Pharmaceutical Chemistry, School of Pharmacy, University of Seville, Seville, Spain
| | - Federico García
- Department of Clinical Microbiology, Instituto de Investigación Biosanitaria ibs. GRANADA, University Hospital Clínico San Cecilio, Granada, Spain
| | - Oscar P. Kuipers
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, The Netherlands,Oscar P. Kuipers University of Groningen, Faculty of Science and Engineering, Department of Genetics, Nijenborgh 7, 9747AG, Groningen, The Netherlands
| | - Juan Carlos Morales
- Department of Biochemistry and Molecular Pharmacology, Instituto de Parasitología y Biomedicina López Neyra, CSIC, PTS Granada, Armilla, Granada, Spain,Juan Carlos Morales Instituto de Parasitología y Biomedicina López Neyra, CSIC, PTS Granada, Avda. del Conocimiento 17, Armilla, 18016Granada, Spain
| |
Collapse
|
31
|
Savitskaya A, Masso-Silva J, Haddaoui I, Enany S. Exploring the arsenal of antimicrobial peptides: Mechanisms, diversity, and applications. Biochimie 2023; 214:216-227. [PMID: 37499896 DOI: 10.1016/j.biochi.2023.07.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 07/09/2023] [Accepted: 07/24/2023] [Indexed: 07/29/2023]
Abstract
Antimicrobial peptides (AMPs) are essential for defence against pathogens in all living organisms and possessed activities against bacteria, fungi, viruses, parasites and even cancer cells. AMPs are short peptides containing 12-100 amino acids conferring a net positive charge and an amphiphilic property in most cases. Although, anionic AMPs also exist. AMPs can be classified based on the types of secondary structures, charge, hydrophobicity, amino acid composition, length, etc. Their mechanism of action usually includes a membrane disruption process through pore formation (three different models have been described, barrel-stave, toroidal or carpet model) but AMPs can also penetrate and impair intracellular functions. Besides their activity against pathogens, they have also shown immunomodulatory properties in complex scenarios through many different interactions. The aim of this review to summarize knowledge about AMP's and discuss the potential application of AMPs as therapeutics, the challenges due to their limitations, including their susceptibility to degradation, the potential generation of AMP resistance, cost, etc. We also discuss the current FDA-approved drugs based on AMPs and strategies to circumvent natural AMPs' limitations.
Collapse
Affiliation(s)
- Anna Savitskaya
- Institute of Bioorganic Chemistry of Russian Academy of Science, Moscow, Russian Federation
| | - Jorge Masso-Silva
- Division of Pulmonary, Critical Care, Sleep Medicine and Physiology, University of California San Diego, La Jolla, CA, USA
| | - Imen Haddaoui
- National Research Institute of Rural Engineering, Water and Forestry, University of Carthage, LR Valorization of Unconventional Waters, Ariana, Tunisia
| | - Shymaa Enany
- Microbiology and Immunology Department, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt; Biomedical Research Department, Armed Force College of Medicine, Cairo, Egypt.
| |
Collapse
|
32
|
Magni A, Mattiello S, Beverina L, Mattioli G, Moschetta M, Zucchi A, Paternò GM, Lanzani G. A membrane intercalating metal-free conjugated organic photosensitizer for bacterial photodynamic inactivation. Chem Sci 2023; 14:8196-8205. [PMID: 37538813 PMCID: PMC10395270 DOI: 10.1039/d3sc01168b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 07/05/2023] [Indexed: 08/05/2023] Open
Abstract
Photodynamic inhibition (PDI) of bacteria represents a powerful strategy for dealing with multidrug-resistant pathogens and infections, as it exhibits minimal development of antibiotic resistance. The PDI action stems from the generation of a triplet state in the photosensitizer (PS), which subsequently transfers energy or electrons to molecular oxygen, resulting in the formation of reactive oxygen species (ROS). These ROS are then able to damage cells, eventually causing bacterial eradication. Enhancing the efficacy of PDI includes the introduction of heavy atoms to augment triplet generation in the PS, as well as membrane intercalation to circumvent the problem of the short lifetime of ROS. However, the former approach can pose safety and environmental concerns, while achieving stable membrane partitioning remains challenging due to the complex outer envelope of bacteria. Here, we introduce a novel PS, consisting of a metal-free donor-acceptor thiophene-based conjugate molecule (BV-1). It presents several advantageous features for achieving effective PDI, namely: (i) it exhibits strong light absorption due to the conjugated donor-acceptor moieties; (ii) it exhibits spontaneous and stable membrane partitioning thanks to its amphiphilicity, accompanied by a strong fluorescence turn-on; (iii) it undergoes metal-free intersystem crossing, which occurs preferentially when the molecule resides in the membrane. All these properties, which we rationalized via optical spectroscopies and calculations, enable the effective eradication of Escherichia coli, with an inhibition concentration that is below that of current state-of-the-art treatments. Our approach holds significant potential for the development of new PS for controlling bacterial infections, particularly those caused by Gram-negative bacteria.
Collapse
Affiliation(s)
- Arianna Magni
- Department of Physics, Politecnico di Milano 20133 Milan Italy
- Center for Nano Science and Technology @PoliMi, Istituto Italiano di Tecnologia 20133 Milan Italy
| | - Sara Mattiello
- Department of Materials Science, University of Milano-Bicocca 20125 Milan Italy
| | - Luca Beverina
- Department of Materials Science, University of Milano-Bicocca 20125 Milan Italy
| | - Giuseppe Mattioli
- CNR - Istituto di Struttura della Materia I-00015 Monterotondo Scalo Italy
| | - Matteo Moschetta
- Center for Nano Science and Technology @PoliMi, Istituto Italiano di Tecnologia 20133 Milan Italy
| | - Anita Zucchi
- Department of Materials Science, University of Milano-Bicocca 20125 Milan Italy
| | - Giuseppe Maria Paternò
- Department of Physics, Politecnico di Milano 20133 Milan Italy
- Center for Nano Science and Technology @PoliMi, Istituto Italiano di Tecnologia 20133 Milan Italy
| | - Guglielmo Lanzani
- Department of Physics, Politecnico di Milano 20133 Milan Italy
- Center for Nano Science and Technology @PoliMi, Istituto Italiano di Tecnologia 20133 Milan Italy
| |
Collapse
|
33
|
Butler MS, Henderson IR, Capon RJ, Blaskovich MAT. Antibiotics in the clinical pipeline as of December 2022. J Antibiot (Tokyo) 2023; 76:431-473. [PMID: 37291465 PMCID: PMC10248350 DOI: 10.1038/s41429-023-00629-8] [Citation(s) in RCA: 76] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 04/20/2023] [Accepted: 04/25/2023] [Indexed: 06/10/2023]
Abstract
The need for new antibacterial drugs to treat the increasing global prevalence of drug-resistant bacterial infections has clearly attracted global attention, with a range of existing and upcoming funding, policy, and legislative initiatives designed to revive antibacterial R&D. It is essential to assess whether these programs are having any real-world impact and this review continues our systematic analyses that began in 2011. Direct-acting antibacterials (47), non-traditional small molecule antibacterials (5), and β-lactam/β-lactamase inhibitor combinations (10) under clinical development as of December 2022 are described, as are the three antibacterial drugs launched since 2020. Encouragingly, the increased number of early-stage clinical candidates observed in the 2019 review increased in 2022, although the number of first-time drug approvals from 2020 to 2022 was disappointingly low. It will be critical to monitor how many Phase-I and -II candidates move into Phase-III and beyond in the next few years. There was also an enhanced presence of novel antibacterial pharmacophores in early-stage trials, and at least 18 of the 26 phase-I candidates were targeted to treat Gram-negative bacteria infections. Despite the promising early-stage antibacterial pipeline, it is essential to maintain funding for antibacterial R&D and to ensure that plans to address late-stage pipeline issues succeed.
Collapse
Affiliation(s)
- Mark S Butler
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, 4072, Australia.
| | - Ian R Henderson
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, 4072, Australia
| | - Robert J Capon
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, 4072, Australia
| | - Mark A T Blaskovich
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, 4072, Australia.
| |
Collapse
|
34
|
Kastner S, Dietel AK, Seier F, Ghosh S, Weiß D, Makarewicz O, Csáki A, Fritzsche W. LSPR-Based Biosensing Enables the Detection of Antimicrobial Resistance Genes. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2207953. [PMID: 37093195 DOI: 10.1002/smll.202207953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 03/30/2023] [Indexed: 05/03/2023]
Abstract
The development of rapid, simple, and accurate bioassays for the detection of nucleic acids has received increasing demand in recent years. Here, localized surface plasmon resonance (LSPR) spectroscopy for the detection of an antimicrobial resistance gene, sulfhydryl variable β-lactamase (blaSHV), which confers resistance against a broad spectrum of β-lactam antibiotics is used. By performing limit of detection experiments, a 23 nucleotide (nt) long deoxyribonucleic acid (DNA) sequence down to 25 nm was detected, whereby the signal intensity is inversely correlated with sequence length (23, 43, 63, and 100 nt). In addition to endpoint measurements of hybridization events, the setup also allowed to monitor the hybridization events in real-time, and consequently enabled to extract kinetic parameters of the studied binding reaction. Performing LSPR measurements using single nucleotide polymorphism (SNP) variants of blaSHV revealed that these sequences can be distinguished from the fully complementary sequence. The possibility to distinguish such sequences is of utmost importance in clinical environments, as it allows to identify mutations essential for enzyme function and thus, is crucial for the correct treatment with antibiotics. Taken together, this system provides a robust, label-free, and cost-efficient analytical tool for the detection of nucleic acids and will enable the surveillance of antimicrobial resistance determinants.
Collapse
Affiliation(s)
- Stephan Kastner
- Molecular Plasmonics work group, Department of Nanobiophotonics, Leibniz Institute of Photonic Technology, Albert-Einstein-Strasse 9, 07745, Jena, Germany
- Leibniz Institute of Photonic Technology, Member of Leibniz Research Alliance Health Technologies and Member of the Leibniz Centre for Photonics in Infection Research (LPI), Albert-Einstein-Strasse 9, 07745, Jena, Germany
| | - Anne-Kathrin Dietel
- Molecular Plasmonics work group, Department of Nanobiophotonics, Leibniz Institute of Photonic Technology, Albert-Einstein-Strasse 9, 07745, Jena, Germany
- Leibniz Institute of Photonic Technology, Member of Leibniz Research Alliance Health Technologies and Member of the Leibniz Centre for Photonics in Infection Research (LPI), Albert-Einstein-Strasse 9, 07745, Jena, Germany
| | - Florian Seier
- Molecular Plasmonics work group, Department of Nanobiophotonics, Leibniz Institute of Photonic Technology, Albert-Einstein-Strasse 9, 07745, Jena, Germany
- Leibniz Institute of Photonic Technology, Member of Leibniz Research Alliance Health Technologies and Member of the Leibniz Centre for Photonics in Infection Research (LPI), Albert-Einstein-Strasse 9, 07745, Jena, Germany
| | - Shaunak Ghosh
- Molecular Plasmonics work group, Department of Nanobiophotonics, Leibniz Institute of Photonic Technology, Albert-Einstein-Strasse 9, 07745, Jena, Germany
- Leibniz Institute of Photonic Technology, Member of Leibniz Research Alliance Health Technologies and Member of the Leibniz Centre for Photonics in Infection Research (LPI), Albert-Einstein-Strasse 9, 07745, Jena, Germany
| | - Daniel Weiß
- Institute for Infectious Diseases and Infection Control, Jena University Hospital, Am Klinikum 1, 07747, Jena, Germany
- Leibniz Institute of Photonic Technology e.V., Member of the Leibniz Centre for Photonics in Infection Research (LPI), Albert-Einstein-Strasse 9, 07745, Jena, Germany
| | - Oliwia Makarewicz
- Institute for Infectious Diseases and Infection Control, Jena University Hospital, Am Klinikum 1, 07747, Jena, Germany
- Leibniz Institute of Photonic Technology e.V., Member of the Leibniz Centre for Photonics in Infection Research (LPI), Albert-Einstein-Strasse 9, 07745, Jena, Germany
| | - Andrea Csáki
- Molecular Plasmonics work group, Department of Nanobiophotonics, Leibniz Institute of Photonic Technology, Albert-Einstein-Strasse 9, 07745, Jena, Germany
- Leibniz Institute of Photonic Technology, Member of Leibniz Research Alliance Health Technologies and Member of the Leibniz Centre for Photonics in Infection Research (LPI), Albert-Einstein-Strasse 9, 07745, Jena, Germany
| | - Wolfgang Fritzsche
- Molecular Plasmonics work group, Department of Nanobiophotonics, Leibniz Institute of Photonic Technology, Albert-Einstein-Strasse 9, 07745, Jena, Germany
- Leibniz Institute of Photonic Technology, Member of Leibniz Research Alliance Health Technologies and Member of the Leibniz Centre for Photonics in Infection Research (LPI), Albert-Einstein-Strasse 9, 07745, Jena, Germany
| |
Collapse
|
35
|
Cardoso FS, Kadam AL, Nelson RC, Tomlin JW, Dahal D, Kuehner CS, Gudvangen G, Arduengo AJ, Burns JM, Aleshire SL, Snead DR, Qu F, Belmore K, Ahmad S, Agrawal T, Sieber JD, Donsbach KO. Practical and Scalable Two-Step Process for 6-(2-Fluoro-4-nitrophenyl)-2-oxa-6-azaspiro[3.3]heptane: A Key Intermediate of the Potent Antibiotic Drug Candidate TBI-223. Org Process Res Dev 2023; 27:1390-1399. [PMID: 37496954 PMCID: PMC10367134 DOI: 10.1021/acs.oprd.3c00148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Indexed: 07/28/2023]
Abstract
A low-cost, protecting group-free route to 6-(2-fluoro-4-nitrophenyl)-2-oxa-6-azaspiro[3.3]heptane (1), the starting material for the in-development tuberculosis treatment TBI-223, is described. The key bond forming step in this route is the creation of the azetidine ring through a hydroxide-facilitated alkylation of 2-fluoro-4-nitroaniline (2) with 3,3-bis(bromomethyl)oxetane (BBMO, 3). After optimization, this ring formation reaction was demonstrated at 100 g scale with isolated yield of 87% and final product purity of >99%. The alkylating agent 3 was synthesized using an optimized procedure that starts from tribromoneopentyl alcohol (TBNPA, 4), a commercially available flame retardant. Treatment of 4 with sodium hydroxide under Schotten-Baumann conditions closed the oxetane ring, and after distillation, 3 was recovered in 72% yield and >95% purity. This new approach to compound 1 avoids the previous drawbacks associated with the synthesis of 2-oxa-6-azaspiro[3,3]heptane (5), the major cost driver used in previous routes to TBI-223. The optimization and multigram scale-up results for this new route are reported herein.
Collapse
Affiliation(s)
- Flavio S.P. Cardoso
- Medicines
for All Institute, Virginia Commonwealth
University, 737 N. 5th St., Box 980100, Richmond, Virginia 23298, United States
| | - Appasaheb L. Kadam
- Medicines
for All Institute, Virginia Commonwealth
University, 737 N. 5th St., Box 980100, Richmond, Virginia 23298, United States
| | - Ryan C. Nelson
- Medicines
for All Institute, Virginia Commonwealth
University, 737 N. 5th St., Box 980100, Richmond, Virginia 23298, United States
| | - John W. Tomlin
- Medicines
for All Institute, Virginia Commonwealth
University, 737 N. 5th St., Box 980100, Richmond, Virginia 23298, United States
| | - Dipendra Dahal
- School
of Chemistry and Biochemistry, Georgia Institute
of Technology, Atlanta, Georgia 30332-0400, United States
| | - Christopher S. Kuehner
- School
of Chemistry and Biochemistry, Georgia Institute
of Technology, Atlanta, Georgia 30332-0400, United States
| | - Gard Gudvangen
- School
of Chemistry and Biochemistry, Georgia Institute
of Technology, Atlanta, Georgia 30332-0400, United States
| | - Anthony J. Arduengo
- School
of Chemistry and Biochemistry, Georgia Institute
of Technology, Atlanta, Georgia 30332-0400, United States
| | - Justina M. Burns
- Medicines
for All Institute, Virginia Commonwealth
University, 737 N. 5th St., Box 980100, Richmond, Virginia 23298, United States
| | - Sarah L. Aleshire
- Medicines
for All Institute, Virginia Commonwealth
University, 737 N. 5th St., Box 980100, Richmond, Virginia 23298, United States
| | - David R. Snead
- Medicines
for All Institute, Virginia Commonwealth
University, 737 N. 5th St., Box 980100, Richmond, Virginia 23298, United States
| | - Fengrui Qu
- Department
of Chemistry and Biochemistry, The University
of Alabama, Tuscaloosa, Alabama 35487-0336, United States
| | - Ken Belmore
- Department
of Chemistry and Biochemistry, The University
of Alabama, Tuscaloosa, Alabama 35487-0336, United States
| | - Saeed Ahmad
- Medicines
for All Institute, Virginia Commonwealth
University, 737 N. 5th St., Box 980100, Richmond, Virginia 23298, United States
| | - Toolika Agrawal
- Department
of Chemistry, Virginia Commonwealth University, 1001 West Main Street, Richmond, Virginia 23284-3208, United States
| | - Joshua D. Sieber
- Department
of Chemistry, Virginia Commonwealth University, 1001 West Main Street, Richmond, Virginia 23284-3208, United States
| | - Kai Oliver Donsbach
- Medicines
for All Institute, Virginia Commonwealth
University, 737 N. 5th St., Box 980100, Richmond, Virginia 23298, United States
| |
Collapse
|
36
|
Sterle M, Durcik M, Stevenson CEM, Henderson SR, Szili PE, Czikkely M, Lawson DM, Maxwell A, Cahard D, Kikelj D, Zidar N, Pal C, Mašič LP, Ilaš J, Tomašič T, Cotman AE, Zega A. Exploring the 5-Substituted 2-Aminobenzothiazole-Based DNA Gyrase B Inhibitors Active against ESKAPE Pathogens. ACS OMEGA 2023; 8:24387-24395. [PMID: 37457471 PMCID: PMC10339456 DOI: 10.1021/acsomega.3c01930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 06/16/2023] [Indexed: 07/18/2023]
Abstract
We present a new series of 2-aminobenzothiazole-based DNA gyrase B inhibitors with promising activity against ESKAPE bacterial pathogens. Based on the binding information extracted from the cocrystal structure of DNA gyrase B inhibitor A, in complex with Escherichia coli GyrB24, we expanded the chemical space of the benzothiazole-based series to the C5 position of the benzothiazole ring. In particular, compound E showed low nanomolar inhibition of DNA gyrase (IC50 < 10 nM) and broad-spectrum antibacterial activity against pathogens belonging to the ESKAPE group, with the minimum inhibitory concentration < 0.03 μg/mL for most Gram-positive strains and 4-16 μg/mL against Gram-negative E. coli, Acinetobacter baumannii, Pseudomonas aeruginosa, and Klebsiella pneumoniae. To understand the binding mode of the synthesized inhibitors, a combination of docking calculations, molecular dynamics (MD) simulations, and MD-derived structure-based pharmacophore modeling was performed. The computational analysis has revealed that the substitution at position C5 can be used to modify the physicochemical properties and antibacterial spectrum and enhance the inhibitory potency of the compounds. Additionally, a discussion of challenges associated with the synthesis of 5-substituted 2-aminobenzothiazoles is presented.
Collapse
Affiliation(s)
- Maša Sterle
- Faculty
of Pharmacy, University of Ljubljana, Aškerčeva Cesta 7, Ljubljana 1000, Slovenia
| | - Martina Durcik
- Faculty
of Pharmacy, University of Ljubljana, Aškerčeva Cesta 7, Ljubljana 1000, Slovenia
| | - Clare E. M. Stevenson
- Department
of Biochemistry and Metabolism, John Innes
Centre, Norwich Research Park, Norwich NR4 7UH, U.K.
| | - Sara R. Henderson
- Institute
of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, U.K.
| | - Petra Eva Szili
- Synthetic
and Systems Biology Unit, Biological Research Centre, Institute of Biochemistry, Szeged H-6726, Hungary
| | - Marton Czikkely
- Synthetic
and Systems Biology Unit, Biological Research Centre, Institute of Biochemistry, Szeged H-6726, Hungary
| | - David M. Lawson
- Department
of Biochemistry and Metabolism, John Innes
Centre, Norwich Research Park, Norwich NR4 7UH, U.K.
| | - Anthony Maxwell
- Department
of Biochemistry and Metabolism, John Innes
Centre, Norwich Research Park, Norwich NR4 7UH, U.K.
| | - Dominique Cahard
- CNRS
UMR 6014 COBRA, Normandie Université, Mont Saint Aignan 76821, France
| | - Danijel Kikelj
- Faculty
of Pharmacy, University of Ljubljana, Aškerčeva Cesta 7, Ljubljana 1000, Slovenia
| | - Nace Zidar
- Faculty
of Pharmacy, University of Ljubljana, Aškerčeva Cesta 7, Ljubljana 1000, Slovenia
| | - Csaba Pal
- Synthetic
and Systems Biology Unit, Biological Research Centre, Institute of Biochemistry, Szeged H-6726, Hungary
| | - Lucija Peterlin Mašič
- Faculty
of Pharmacy, University of Ljubljana, Aškerčeva Cesta 7, Ljubljana 1000, Slovenia
| | - Janez Ilaš
- Faculty
of Pharmacy, University of Ljubljana, Aškerčeva Cesta 7, Ljubljana 1000, Slovenia
| | - Tihomir Tomašič
- Faculty
of Pharmacy, University of Ljubljana, Aškerčeva Cesta 7, Ljubljana 1000, Slovenia
| | - Andrej Emanuel Cotman
- Faculty
of Pharmacy, University of Ljubljana, Aškerčeva Cesta 7, Ljubljana 1000, Slovenia
| | - Anamarija Zega
- Faculty
of Pharmacy, University of Ljubljana, Aškerčeva Cesta 7, Ljubljana 1000, Slovenia
| |
Collapse
|
37
|
Zhan W, Gao G, Liu Z, Liu X, Xu L, Wang M, Xu HD, Tang R, Cao J, Sun X, Liang G. Enzymatic Self-Assembly of Adamantane-Peptide Conjugate for Combating Staphylococcus aureus Infection. Adv Healthc Mater 2023; 12:e2203283. [PMID: 36880480 DOI: 10.1002/adhm.202203283] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 02/28/2023] [Indexed: 03/08/2023]
Abstract
Staphylococcus aureus (S. aureus) remains a leading cause of bacterial infections. However, eradication of S. aureus infections with common antibiotics is increasingly difficult due to outbreaks of drug resistance. Therefore, new antibiotic classes and antibacterial strategies are urgently in demand. Herein, it is shown that an adamantane-peptide conjugate, upon dephosphorylation by alkaline phosphatase (ALP) constitutively expressed on S. aureus, generates fibrous assemblies in situ to combat S. aureus infection. By attaching adamantane to a phosphorylated tetrapeptide Nap-Phe-Phe-Lys-Tyr(H2 PO3 )-OH, the rationally designed adamantane-peptide conjugate Nap-Phe-Phe-Lys(Ada)-Tyr(H2 PO3 )-OH (Nap-FYp-Ada) is obtained. Upon bacterial ALP activation, Nap-FYp-Ada is dephosphorylated and self-assembles into nanofibers on the surface of S. aureus. As revealed by cell assays, the assemblies of adamantane-peptide conjugates interact with cell lipid membrane and thereby disrupt membrane integrity to kill S. aureus. Animal experiments further demonstrate the excellent potential of Nap-FYp-Ada in the treatment of S. aureus infection in vivo. This work provides an alternative approach to design antimicrobial agents.
Collapse
Affiliation(s)
- Wenjun Zhan
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou, Nanjing, 210096, P. R. China
| | - Ge Gao
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou, Nanjing, 210096, P. R. China
| | - Zhiyu Liu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou, Nanjing, 210096, P. R. China
| | - Xiaoyang Liu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou, Nanjing, 210096, P. R. China
| | - Lingling Xu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou, Nanjing, 210096, P. R. China
| | - Manli Wang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou, Nanjing, 210096, P. R. China
| | - Hai-Dong Xu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou, Nanjing, 210096, P. R. China
| | - Runqun Tang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou, Nanjing, 210096, P. R. China
| | - Jingyuan Cao
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou, Nanjing, 210096, P. R. China
| | - Xianbao Sun
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou, Nanjing, 210096, P. R. China
| | - Gaolin Liang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou, Nanjing, 210096, P. R. China
| |
Collapse
|
38
|
Douglas EJA, Wulandari SW, Lovell SD, Laabei M. Novel antimicrobial strategies to treat multi-drug resistant Staphylococcus aureus infections. Microb Biotechnol 2023; 16:1456-1474. [PMID: 37178319 PMCID: PMC10281381 DOI: 10.1111/1751-7915.14268] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/12/2023] [Accepted: 04/18/2023] [Indexed: 05/15/2023] Open
Abstract
Antimicrobial resistance is a major obstacle for the treatment of infectious diseases and currently represents one of the most significant threats to global health. Staphylococcus aureus remains a formidable human pathogen with high mortality rates associated with severe systemic infections. S. aureus has become notorious as a multidrug resistant bacterium, which when combined with its extensive arsenal of virulence factors that exacerbate disease, culminates in an incredibly challenging pathogen to treat clinically. Compounding this major health issue is the lack of antibiotic discovery and development, with only two new classes of antibiotics approved for clinical use in the last 20 years. Combined efforts from the scientific community have reacted to the threat of dwindling treatment options to combat S. aureus disease in several innovative and exciting developments. This review describes current and future antimicrobial strategies aimed at treating staphylococcal colonization and/or disease, examining therapies that show significant promise at the preclinical development stage to approaches that are currently being investigated in clinical trials.
Collapse
|
39
|
Silva V, Silva A, Ribeiro J, Aires A, Carvalho R, Amaral JS, Barros L, Igrejas G, Poeta P. Screening of Chemical Composition, Antimicrobial and Antioxidant Activities in Pomegranate, Quince, and Persimmon Leaf, Peel, and Seed: Valorization of Autumn Fruits By-Products for a One Health Perspective. Antibiotics (Basel) 2023; 12:1086. [PMID: 37508182 PMCID: PMC10376090 DOI: 10.3390/antibiotics12071086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/15/2023] [Accepted: 06/16/2023] [Indexed: 07/30/2023] Open
Abstract
Antimicrobial resistance is increasing globally and is now one of the major public health problems. Therefore, there is a need to search for new antimicrobial agents. The food industry generates large amounts of by-products that are rich in bioactive compounds, such as phenolic compounds, which are known to have several health benefits, including antioxidant and antimicrobial properties. Thus, we aimed to characterize the phenolic compounds present in pomegranate, quince, and persimmon by-products, as well as their antioxidant and antimicrobial activities. Phenolic compounds were extracted from pomegranate, quince, and persimmon leaves, seeds, and peels using a mixture of ethanol/water (80/20). The polyphenol profile of the extracts was determined by high-performance liquid chromatography. The antioxidant activity of the extracts was determined by the 2,2-diphenyl-1-picrylhydrazyl (DPPH), ferric reducing antioxidant power (FRAP), and cupric reducing antioxidant capacity (CUPRAC) methods. Antimicrobial susceptibility was evaluated using the Kirby-Bauer disk diffusion method. In general, leaves showed higher concentrations of phenolics than the peel and seeds of fruits. In total, 23 phenolic compounds were identified and quantified, with sanguiin and apigenin-3-O-galactoside being present in the highest concentrations. Leaf extracts of pomegranate showed higher antioxidant activities than the other components in all methods used. In general, all extracts had a greater antimicrobial activity against Gram-positive bacteria. Persimmon leaf and seed extracts inhibited a greater number of bacteria, both Gram-positive and -negative. The lowest minimum inhibitory concentration (MIC) detected among Gram-positive and -negative bacteria was 10 mg/mL for pomegranate peel and leaf extracts against Staphylococcus aureus and S. pseudintermedius and for pomegranate leaf extract against Escherichia coli. Our results reinforce the need to value food industry by-products that could be used as food preservatives and antibiotic adjuvants against multiresistant bacteria.
Collapse
Affiliation(s)
- Vanessa Silva
- Microbiology and Antibiotic Resistance Team (MicroART), Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal
- Associated Laboratory for Green Chemistry (LAQV-REQUIMTE), University NOVA of Lisbon, 2829-516 Caparica, Portugal
- Department of Genetics and Biotechnology, University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal
- Functional Genomics and Proteomics Unit, University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal
| | - Adriana Silva
- Microbiology and Antibiotic Resistance Team (MicroART), Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal
- Associated Laboratory for Green Chemistry (LAQV-REQUIMTE), University NOVA of Lisbon, 2829-516 Caparica, Portugal
- Department of Genetics and Biotechnology, University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal
- Functional Genomics and Proteomics Unit, University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal
| | - Jessica Ribeiro
- Microbiology and Antibiotic Resistance Team (MicroART), Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal
- Associated Laboratory for Green Chemistry (LAQV-REQUIMTE), University NOVA of Lisbon, 2829-516 Caparica, Portugal
- Centro de Investigação de Montanha (CIMO), Instituto Politécnico de Bragança, Campus Santa Apolónia, 5300-253 Bragança, Portugal
| | - Alfredo Aires
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences, CITAB, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal
| | - Rosa Carvalho
- Department of Agronomy, School of Agrarian and Veterinary Sciences, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal
| | - Joana S Amaral
- Centro de Investigação de Montanha (CIMO), Instituto Politécnico de Bragança, Campus Santa Apolónia, 5300-253 Bragança, Portugal
- Laboratório Associado para a Sustentabilidade e Tecnologia em Regiões de Montanha (SusTEC), Instituto Politécnico de Bragança, Campus de Santa Apolónia, 5300-253 Bragança, Portugal
| | - Lillian Barros
- Centro de Investigação de Montanha (CIMO), Instituto Politécnico de Bragança, Campus Santa Apolónia, 5300-253 Bragança, Portugal
- Laboratório Associado para a Sustentabilidade e Tecnologia em Regiões de Montanha (SusTEC), Instituto Politécnico de Bragança, Campus de Santa Apolónia, 5300-253 Bragança, Portugal
| | - Gilberto Igrejas
- Associated Laboratory for Green Chemistry (LAQV-REQUIMTE), University NOVA of Lisbon, 2829-516 Caparica, Portugal
- Department of Genetics and Biotechnology, University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal
- Functional Genomics and Proteomics Unit, University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal
| | - Patrícia Poeta
- Microbiology and Antibiotic Resistance Team (MicroART), Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal
- Associated Laboratory for Green Chemistry (LAQV-REQUIMTE), University NOVA of Lisbon, 2829-516 Caparica, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal
- Veterinary and Animal Research Centre (CECAV), University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal
| |
Collapse
|
40
|
Gulyás D, Kamotsay K, Szabó D, Kocsis B. Investigation of Delafloxacin Resistance in Multidrug-Resistant Escherichia coli Strains and the Detection of E. coli ST43 International High-Risk Clone. Microorganisms 2023; 11:1602. [PMID: 37375104 DOI: 10.3390/microorganisms11061602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/13/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
Delafloxacin is a novel fluoroquinolone agent that is approved for clinical application. In this study, we analyzed the antibacterial efficacy of delafloxacin in a collection of 47 Escherichia coli strains. Antimicrobial susceptibility testing was performed by the broth microdilution method and minimum inhibitory concentration (MIC) values were determined for delafloxacin, ciprofloxacin, levofloxacin, moxifloxacin, ceftazidime, cefotaxime, and imipenem. Two multidrug-resistant E. coli strains, which exhibited delafloxacin and ciprofloxacin resistance as well as extended-spectrum beta-lactamase (ESBL) phenotype, were selected for whole-genome sequencing (WGS). In our study, delafloxacin and ciprofloxacin resistance rates were 47% (22/47) and 51% (24/47), respectively. In the strain collection, 46 E. coli were associated with ESBL production. The MIC50 value for delafloxacin was 0.125 mg/L, while all other fluoroquinolones had an MIC50 value of 0.25 mg/L in our collection. Delafloxacin susceptibility was detected in 20 ESBL positive and ciprofloxacin resistant E. coli strains; by contrast, E. coli strains that exhibited a ciprofloxacin MIC value above 1 mg/L were delafloxacin-resistant. WGS analysis on the two selected E. coli strains (920/1 and 951/2) demonstrated that delafloxacin resistance is mediated by multiple chromosomal mutations, namely, five mutations in E. coli 920/1 (gyrA S83L, D87N, parC S80I, E84V, and parE I529L) and four mutations in E. coli 951/2 (gyrA S83L, D87N, parC S80I, and E84V). Both strains carried an ESBL gene, blaCTX-M-1 in E. coli 920/1 and blaCTX-M-15 in E. coli 951/2. Based on multilocus sequence typing, both strains belong to the E. coli sequence type 43 (ST43). In this paper, we report a remarkable high rate (47%) of delafloxacin resistance among multidrug-resistant E. coli as well as the E. coli ST43 international high-risk clone in Hungary.
Collapse
Affiliation(s)
- Dániel Gulyás
- Institute of Medical Microbiology, Semmelweis University, 1089 Budapest, Hungary
| | - Katalin Kamotsay
- Central Microbiology Laboratory, National Institute of Hematology and Infectious Disease, Central Hospital of Southern-Pest, 1097 Budapest, Hungary
| | - Dóra Szabó
- Institute of Medical Microbiology, Semmelweis University, 1089 Budapest, Hungary
- Human Microbiota Study Group, Semmelweis University-Eötvös Lóránd Research Network, 1089 Budapest, Hungary
| | - Béla Kocsis
- Institute of Medical Microbiology, Semmelweis University, 1089 Budapest, Hungary
| |
Collapse
|
41
|
Koh AJJ, Thombare V, Hussein M, Rao GG, Li J, Velkov T. Bifunctional antibiotic hybrids: A review of clinical candidates. Front Pharmacol 2023; 14:1158152. [PMID: 37397488 PMCID: PMC10313405 DOI: 10.3389/fphar.2023.1158152] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 06/02/2023] [Indexed: 07/04/2023] Open
Abstract
Antibiotic resistance is a top threat to human health and a priority across the globe. This problematic issue is accompanied by the decline of new antibiotics in the pipeline over the past 30 years. In this context, an urgent need to develop new strategies to combat antimicrobial resistance is in great demand. Lately, among the possible approaches used to deal with antimicrobial resistance is the covalent ligation of two antibiotic pharmacophores that target the bacterial cells through a dissimilar mode of action into a single hybrid molecule, namely hybrid antibiotics. This strategy exhibits several advantages, including better antibacterial activity, overcoming the existing resistance towards individual antibiotics, and may ultimately delay the onset of bacterial resistance. This review sheds light on the latest development of the dual antibiotic hybrids pipeline, their potential mechanisms of action, and challenges in their use.
Collapse
Affiliation(s)
- Augustine Jing Jie Koh
- Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIP, Australia
| | - Varsha Thombare
- Monash Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Parkville, VIP, Australia
| | - Maytham Hussein
- Monash Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Parkville, VIP, Australia
| | - Gauri G. Rao
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, United States
| | - Jian Li
- Monash Biomedicine Discovery Institute, Department of Microbiology, Monash University, Parkville, VIP, Australia
| | - Tony Velkov
- Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIP, Australia
- Monash Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Parkville, VIP, Australia
| |
Collapse
|
42
|
Mutalik C, Saukani M, Khafid M, Krisnawati DI, Darmayanti R, Puspitasari B, Cheng TM, Kuo TR. Gold-Based Nanostructures for Antibacterial Application. Int J Mol Sci 2023; 24:10006. [PMID: 37373154 DOI: 10.3390/ijms241210006] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/04/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Bacterial infections have become a fatal threat because of the abuse of antibiotics in the world. Various gold (Au)-based nanostructures have been extensively explored as antibacterial agents to combat bacterial infections based on their remarkable chemical and physical characteristics. Many Au-based nanostructures have been designed and their antibacterial activities and mechanisms have been further examined and demonstrated. In this review, we collected and summarized current developments of antibacterial agents of Au-based nanostructures, including Au nanoparticles (AuNPs), Au nanoclusters (AuNCs), Au nanorods (AuNRs), Au nanobipyramids (AuNBPs), and Au nanostars (AuNSs) according to their shapes, sizes, and surface modifications. The rational designs and antibacterial mechanisms of these Au-based nanostructures are further discussed. With the developments of Au-based nanostructures as novel antibacterial agents, we also provide perspectives, challenges, and opportunities for future practical clinical applications.
Collapse
Affiliation(s)
- Chinmaya Mutalik
- Graduate Institute of Nanomedicine and Medical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Muhammad Saukani
- International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
- Department of Mechanical Engineering, Faculty of Engineering, Universitas Islam Kalimantan MAB, Banjarmasin 70124, Kalimantan Selatan, Indonesia
| | - Muhamad Khafid
- Department of Nursing, Faculty of Nursing and Midwifery, Universitas Nahdlatul Ulama Surabaya, Surabaya 60237, East Java, Indonesia
| | | | - Rofik Darmayanti
- Dharma Husada Nursing Academy, Kediri 64117, East Java, Indonesia
| | | | - Tsai-Mu Cheng
- Graduate Institute for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- Taipei Heart Institute, Taipei Medical University, Taipei 11031, Taiwan
- Cardiovascular Research Center, Taipei Medical University Hospital, Taipei Medical University, Taipei 11031, Taiwan
| | - Tsung-Rong Kuo
- Graduate Institute of Nanomedicine and Medical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
- Stanford Byers Center for Biodesign, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
43
|
Díaz-Puertas R, Adamek M, Mallavia R, Falco A. Fish Skin Mucus Extracts: An Underexplored Source of Antimicrobial Agents. Mar Drugs 2023; 21:350. [PMID: 37367675 DOI: 10.3390/md21060350] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/01/2023] [Accepted: 06/05/2023] [Indexed: 06/28/2023] Open
Abstract
The slow discovery of new antibiotics combined with the alarming emergence of antibiotic-resistant bacteria underscores the need for alternative treatments. In this regard, fish skin mucus has been demonstrated to contain a diverse array of bioactive molecules with antimicrobial properties, including peptides, proteins, and other metabolites. This review aims to provide an overview of the antimicrobial molecules found in fish skin mucus and its reported in vitro antimicrobial capacity against bacteria, fungi, and viruses. Additionally, the different methods of mucus extraction, which can be grouped as aqueous, organic, and acidic extractions, are presented. Finally, omic techniques (genomics, transcriptomics, proteomics, metabolomics, and multiomics) are described as key tools for the identification and isolation of new antimicrobial compounds. Overall, this study provides valuable insight into the potential of fish skin mucus as a promising source for the discovery of new antimicrobial agents.
Collapse
Affiliation(s)
- Rocío Díaz-Puertas
- Institute of Research, Development and Innovation in Healthcare Biotechnology in Elche (IDiBE), Miguel Hernández University, 03202 Elche, Spain
| | - Mikolaj Adamek
- Fish Disease Research Unit, Institute for Parasitology, University of Veterinary Medicine, 30559 Hannover, Germany
| | - Ricardo Mallavia
- Institute of Research, Development and Innovation in Healthcare Biotechnology in Elche (IDiBE), Miguel Hernández University, 03202 Elche, Spain
| | - Alberto Falco
- Institute of Research, Development and Innovation in Healthcare Biotechnology in Elche (IDiBE), Miguel Hernández University, 03202 Elche, Spain
| |
Collapse
|
44
|
Maghrebi S, Thomas N, Prestidge CA, Joyce P. Inulin-lipid hybrid (ILH) microparticles promote pH-triggered release of rifampicin within infected macrophages. Drug Deliv Transl Res 2023; 13:1716-1729. [PMID: 36630076 PMCID: PMC10126022 DOI: 10.1007/s13346-022-01287-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/2022] [Indexed: 01/12/2023]
Abstract
Intracellular bacteria serve as a problematic source of infection due to their ability to evade biological immune responses and the inability for conventional antibiotics to efficiently penetrate cellular membranes. Subsequently, new treatment approaches are urgently required to effectively eradicate intracellular pathogens residing within immune cells (e.g. macrophages). In this study, the poorly soluble and poorly permeable antibiotic, rifampicin, was re-purposed via micro-encapsulation within inulin-lipid hybrid (ILH) particles for the treatment of macrophages infected with small colony variants of Staphylococcus aureus (SCV S. aureus). Rifampicin-encapsulated ILH (Rif-ILH) microparticles were synthesized by spray drying a lipid nano-emulsion, with inulin dissolved throughout the aqueous phase and rifampicin pre-loaded within the lipid phase. Rif-ILH were strategically designed and engineered with pH-responsive properties to promote lysosomal drug release upon cellular internalization, while preventing premature rifampicin release in plasma-simulating media. The pH-responsiveness of Rif-ILH was controlled by the acid-mediated hydrolysis of the inulin coating, where exposure to acidic media simulating the lysosomal environment of macrophages triggered hydrolysis of the oligofructose chain and the subsequent diffusion of rifampicin from Rif-ILH. This pH-provoked release mechanism, as well as the ability for ILH microparticles to be more readily internalized by macrophages, was found to be influential in triggering a 2.9-fold increase in intracellular rifampicin concentration within infected macrophages, compared to the pure drug. The subsequent increase in exposure of intracellular pathogens to rifampicin leads to a ~ 2-log improvement in antibacterial activity for Rif-ILH, at a rifampicin dose of 2.5 µg/mL. Thus, the reduction in viability of intracellular SCV S. aureus, in the absence of cellular toxicity, is indicative of ILH microparticles serving as a unique approach for the safe and efficacious delivery of antibiotics to phagocytic cells for the treatment of intracellular infections.
Collapse
Affiliation(s)
- Sajedeh Maghrebi
- UniSA Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, 5000, Australia
| | - Nicky Thomas
- UniSA Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, 5000, Australia
| | - Clive A Prestidge
- UniSA Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, 5000, Australia.
| | - Paul Joyce
- UniSA Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, 5000, Australia.
| |
Collapse
|
45
|
Rayner B, Verderosa AD, Ferro V, Blaskovich MAT. Siderophore conjugates to combat antibiotic-resistant bacteria. RSC Med Chem 2023; 14:800-822. [PMID: 37252105 PMCID: PMC10211321 DOI: 10.1039/d2md00465h] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 02/21/2023] [Indexed: 10/31/2023] Open
Abstract
Antimicrobial resistance (AMR) is a global threat to society due to the increasing emergence of multi-drug resistant bacteria that are not susceptible to our last line of defence antibiotics. Exacerbating this issue is a severe gap in antibiotic development, with no new clinically relevant classes of antibiotics developed in the last two decades. The combination of the rapidly increasing emergence of resistance and scarcity of new antibiotics in the clinical pipeline means there is an urgent need for new efficacious treatment strategies. One promising solution, known as the 'Trojan horse' approach, hijacks the iron transport system of bacteria to deliver antibiotics directly into cells - effectively tricking bacteria into killing themselves. This transport system uses natively produced siderophores, which are small molecules with a high affinity for iron. By linking antibiotics to siderophores, to make siderophore antibiotic conjugates, the activity of existing antibiotics can potentially be reinvigorated. The success of this strategy was recently exemplified with the clinical release of cefiderocol, a cephalosporin-siderophore conjugate with potent antibacterial activity against carbapenem-resistant and multi-drug resistant Gram-negative bacilli. This review discusses the recent advancements in siderophore antibiotic conjugates and the challenges associated with the design of these compounds that need to be overcome to deliver more efficacious therapeutics. Potential strategies have also been suggested for new generations of siderophore-antibiotics with enhanced activity.
Collapse
Affiliation(s)
- Beth Rayner
- Centre for Superbug Solutions, Institute for Molecular Bioscience, University of Queensland Brisbane Queensland Australia
- Australian Infectious Disease Research Centre, The University of Queensland Brisbane Queensland Australia
| | - Anthony D Verderosa
- Centre for Superbug Solutions, Institute for Molecular Bioscience, University of Queensland Brisbane Queensland Australia
- Australian Infectious Disease Research Centre, The University of Queensland Brisbane Queensland Australia
| | - Vito Ferro
- Australian Infectious Disease Research Centre, The University of Queensland Brisbane Queensland Australia
- School of Chemistry and Molecular Biosciences, The University of Queensland Australia
| | - Mark A T Blaskovich
- Centre for Superbug Solutions, Institute for Molecular Bioscience, University of Queensland Brisbane Queensland Australia
- Australian Infectious Disease Research Centre, The University of Queensland Brisbane Queensland Australia
- School of Chemistry and Molecular Biosciences, The University of Queensland Australia
| |
Collapse
|
46
|
Kapp E, Calitz H, Streicher EM, Dippenaar A, Egieyeh S, Jordaan A, Warner DF, Joubert J, Malan SF, Sampson SL. Discovery and biological evaluation of an adamantyl-amide derivative with likely MmpL3 inhibitory activity. Tuberculosis (Edinb) 2023; 141:102350. [PMID: 37244249 DOI: 10.1016/j.tube.2023.102350] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 05/02/2023] [Accepted: 05/07/2023] [Indexed: 05/29/2023]
Abstract
A series of molecules containing bulky lipophilic scaffolds was screened for activity against Mycobacterium tuberculosis and a number of compounds with antimycobacterial activity were identified. The most active compound, (2E)-N-(adamantan-1-yl)-3-phenylprop-2-enamide (C1), has a low micromolar minimum inhibitory concentration, low cytotoxicity (therapeutic index = 32.26), low mutation frequency and is active against intracellular Mycobacterium tuberculosis. Whole genome sequencing of mutants resistant to C1 showed a mutation in mmpL3 which may point to the involvement of MmpL3 in the antimycobacterial activity of the compound. In silico mutagenesis and molecular modelling studies were performed to better understand the binding of C1 within MmpL3 and the role that the specific mutation may play in the interaction at protein level. These analyses revealed that the mutation increases the energy required for binding of C1 within the protein translocation channel of MmpL3. The mutation also decreases the solvation energy of the protein, suggesting that the mutant protein might be more solvent-accessible, thereby restricting its interaction with other molecules. The results reported here describe a new molecule that may interact with the MmpL3 protein, providing insights into the effect of mutations on protein-ligand interactions and enhancing our understanding of this essential protein as a priority drug target.
Collapse
Affiliation(s)
- Erika Kapp
- School of Pharmacy, Faculty of Natural Sciences, University of the Western Cape, Cape Town, South Africa; University of the Western Cape, Private Bag x17, Bellville, 7535, South Africa.
| | - Hanri Calitz
- DSI/NRF Centre of Excellence for Biomedical Tuberculosis Research/South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, P.O. Box 241, Cape Town, 8000, South Africa.
| | - Elizabeth M Streicher
- DSI/NRF Centre of Excellence for Biomedical Tuberculosis Research/South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, P.O. Box 241, Cape Town, 8000, South Africa.
| | - Anzaan Dippenaar
- DSI/NRF Centre of Excellence for Biomedical Tuberculosis Research/South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, P.O. Box 241, Cape Town, 8000, South Africa; Global Health Institute, Department of Family Medicine and Population Health, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium; Gouverneur Kinsbergencentrum, Doornstraat 331, 2610, Wilrijk, Belgium.
| | - Samuel Egieyeh
- School of Pharmacy, Faculty of Natural Sciences, University of the Western Cape, Cape Town, South Africa; University of the Western Cape, Private Bag x17, Bellville, 7535, South Africa.
| | - Audrey Jordaan
- Molecular Mycobacteriology Research Unit, DSI/NRF Centre of Excellence for Biomedical Tuberculosis Research, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Private Bag X3, Rondebosch, 7701, Cape Town, South Africa.
| | - Digby F Warner
- Molecular Mycobacteriology Research Unit, DSI/NRF Centre of Excellence for Biomedical Tuberculosis Research, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Private Bag X3, Rondebosch, 7701, Cape Town, South Africa.
| | - Jacques Joubert
- School of Pharmacy, Faculty of Natural Sciences, University of the Western Cape, Cape Town, South Africa; University of the Western Cape, Private Bag x17, Bellville, 7535, South Africa.
| | - Sarel F Malan
- School of Pharmacy, Faculty of Natural Sciences, University of the Western Cape, Cape Town, South Africa; University of the Western Cape, Private Bag x17, Bellville, 7535, South Africa.
| | - Samantha L Sampson
- DSI/NRF Centre of Excellence for Biomedical Tuberculosis Research/South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, P.O. Box 241, Cape Town, 8000, South Africa.
| |
Collapse
|
47
|
Galani I, Papoutsaki V, Karaiskos I, Moustakas N, Galani L, Maraki S, Mavromanolaki VE, Legga O, Fountoulis K, Platsouka ED, Giannopoulou P, Papadogeorgaki H, Damala M, Chinou E, Pasxali A, Deliolanis I, Vagiakou H, Petinaki E, Chli A, Vagdatli E, Kazila P, Papaioannou V, Kontopoulou K, Ferke AN, Moraitou E, Antoniadou A, Giamarellou H. In vitro activities of omadacycline, eravacycline, cefiderocol, apramycin, and comparator antibiotics against Acinetobacter baumannii causing bloodstream infections in Greece, 2020-2021: a multicenter study. Eur J Clin Microbiol Infect Dis 2023:10.1007/s10096-023-04616-7. [PMID: 37133639 PMCID: PMC10155660 DOI: 10.1007/s10096-023-04616-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 04/26/2023] [Indexed: 05/04/2023]
Abstract
Resistance of Acinetobacter baumannii to multiple clinically important antimicrobials has increased to very high rates in Greece, rendering most of them obsolete. The aim of this study was to determine the molecular epidemiology and susceptibilities of A. baumannii isolates collected from different hospitals across Greece. Single-patient A. baumannii strains isolated from blood cultures (n = 271), from 19 hospitals, in a 6-month period (November 2020-April 2021) were subjected to minimum inhibitory concentration determination and molecular testing for carbapenemase, 16S rRNA methyltransferase and mcr gene detection and epidemiological evaluation. 98.9% of all isolates produced carbapenemase OXA-23. The vast majority (91.8%) of OXA-23 producers harbored the armA and were assigned mainly (94.3%) to sequence group G1, corresponding to IC II. Apramycin (EBL-1003) was the most active agent inhibiting 100% of the isolates at ≤16 mg/L, followed by cefiderocol which was active against at least 86% of them. Minocycline, colistin and ampicillin-sulbactam exhibited only sparse activity (S <19%), while eravacycline was 8- and 2-fold more active than minocycline and tigecycline respectively, by comparison of their MIC50/90 values. OXA-23-ArmA producing A. baumannii of international clone II appears to be the prevailing epidemiological type of this organism in Greece. Cefiderocol could provide a useful alternative for difficult to treat Gram-negative infections, while apramycin (EBL-1003), the structurally unique aminoglycoside currently in clinical development, may represent a highly promising agent against multi-drug resistant A. baumanni infections, due to its high susceptibility rates and low toxicity.
Collapse
Affiliation(s)
- Irene Galani
- Infectious Diseases Laboratory, 4th Department of Internal Medicine, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece.
| | | | - Ilias Karaiskos
- 1st Department of Internal Medicine-Infectious Diseases, Hygeia General Hospital, Athens, Greece
| | - Nikolaos Moustakas
- Infectious Diseases Laboratory, 4th Department of Internal Medicine, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Lamprini Galani
- 1st Department of Internal Medicine-Infectious Diseases, Hygeia General Hospital, Athens, Greece
| | - Sofia Maraki
- Department of Clinical Bacteriology, Parasitology, Zoonoses and Geographical Medicine, University Hospital of Heraklion, Heraklion, Greece
| | - Viktoria Eirini Mavromanolaki
- Department of Clinical Bacteriology, Parasitology, Zoonoses and Geographical Medicine, University Hospital of Heraklion, Heraklion, Greece
| | - Olga Legga
- Department of Microbiology, General Hospital of Lamia, Lamia, Greece
| | - Kimon Fountoulis
- Department of Clinical Microbiology, Evangelismos General Hospital, Athens, Greece
| | - Evangelia D Platsouka
- Department of Microbiology, General Hospital of Nea Ionia, "Konstantopouleio-Patission", Athens, Greece
| | | | | | - Maria Damala
- Microbiology Department, "Alexandra" General Hospital of Athens, Athens, Greece
| | - Efrosini Chinou
- Department of Microbiology, St Savvas Cancer Hospital, Athens, Greece
| | - Aggeliki Pasxali
- Microbiology Laboratory, General Hospital of Corfu, Corfu, Greece
| | | | - Helen Vagiakou
- Microbiology Laboratory, General Hospital of Athens "G. Gennimatas", Athens, Greece
| | - Efthymia Petinaki
- Department of Microbiology, University Hospital of Larissa, Larissa, Greece
| | - Anastasia Chli
- Microbiology Laboratory, General Hospital of Kavala, Kavala, Greece
| | - Eleni Vagdatli
- Microbiology Department, Hippokration General Hospital, Thessaloniki, Greece
| | - Polyzo Kazila
- Department of Clinical Chemistry, "THEAGENEIO" Cancer Hospital, Thessaloniki, Greece
| | | | - Konstantina Kontopoulou
- Department of Microbiology, General Hospital of Thessaloniki "G. Gennimatas", Thessaloniki, Greece
| | | | - Eleni Moraitou
- Department of Clinical Microbiology, Sotiria General Hospital of Chest Diseases, Athens, Greece
| | - Anastasia Antoniadou
- Infectious Diseases Laboratory, 4th Department of Internal Medicine, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Helen Giamarellou
- 1st Department of Internal Medicine-Infectious Diseases, Hygeia General Hospital, Athens, Greece
| |
Collapse
|
48
|
Ottonello A, Wyllie JA, Yahiaoui O, Sun S, Koelln RA, Homer JA, Johnson RM, Murray E, Williams P, Bolla JR, Robinson CV, Fallon T, Soares da Costa TP, Moses JE. Shapeshifting bullvalene-linked vancomycin dimers as effective antibiotics against multidrug-resistant gram-positive bacteria. Proc Natl Acad Sci U S A 2023; 120:e2208737120. [PMID: 37011186 PMCID: PMC10104512 DOI: 10.1073/pnas.2208737120] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 02/24/2023] [Indexed: 04/05/2023] Open
Abstract
The alarming rise in superbugs that are resistant to drugs of last resort, including vancomycin-resistant enterococci and staphylococci, has become a significant global health hazard. Here, we report the click chemistry synthesis of an unprecedented class of shapeshifting vancomycin dimers (SVDs) that display potent activity against bacteria that are resistant to the parent drug, including the ESKAPE pathogens, vancomycin-resistant Enterococcus (VRE), methicillin-resistant Staphylococcus aureus (MRSA), as well as vancomycin-resistant S. aureus (VRSA). The shapeshifting modality of the dimers is powered by a triazole-linked bullvalene core, exploiting the dynamic covalent rearrangements of the fluxional carbon cage and creating ligands with the capacity to inhibit bacterial cell wall biosynthesis. The new shapeshifting antibiotics are not disadvantaged by the common mechanism of vancomycin resistance resulting from the alteration of the C-terminal dipeptide with the corresponding d-Ala-d-Lac depsipeptide. Further, evidence suggests that the shapeshifting ligands destabilize the complex formed between the flippase MurJ and lipid II, implying the potential for a new mode of action for polyvalent glycopeptides. The SVDs show little propensity for acquired resistance by enterococci, suggesting that this new class of shapeshifting antibiotic will display durable antimicrobial activity not prone to rapidly acquired clinical resistance.
Collapse
Affiliation(s)
- Alessandra Ottonello
- La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC3086, Australia
| | - Jessica A. Wyllie
- La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC3086, Australia
| | - Oussama Yahiaoui
- Department of Chemistry, School of Physical Sciences, The University of Adelaide, Adelaide, SA5005, Australia
| | - Shoujun Sun
- Cancer Center, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY11724
| | - Rebecca A. Koelln
- Cancer Center, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY11724
| | - Joshua A. Homer
- Cancer Center, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY11724
| | - Robert M. Johnson
- Cancer Center, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY11724
| | - Ewan Murray
- National Biofilms Innovation Centre, Biodiscovery Institute and School of Life Sciences, University of Nottingham, University Park, NottinghamNG7 2RD, U.K.
| | - Paul Williams
- National Biofilms Innovation Centre, Biodiscovery Institute and School of Life Sciences, University of Nottingham, University Park, NottinghamNG7 2RD, U.K.
| | - Jani R. Bolla
- Department of Biology, University of Oxford, OxfordOX1 3RB, U.K.
- The Kavli Institute for Nanoscience Discovery, University of Oxford, OxfordOX1 3QU, U.K.
| | - Carol V. Robinson
- The Kavli Institute for Nanoscience Discovery, University of Oxford, OxfordOX1 3QU, U.K.
- Physical and Theoretical Chemistry Laboratory, University of Oxford, OxfordOX1 3QZ, U.K.
| | - Thomas Fallon
- Department of Chemistry, School of Physical Sciences, The University of Adelaide, Adelaide, SA5005, Australia
| | | | - John E. Moses
- Cancer Center, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY11724
| |
Collapse
|
49
|
Shanbhag C, Saraogi I. Bacterial GTPases as druggable targets to tackle antimicrobial resistance. Bioorg Med Chem Lett 2023; 87:129276. [PMID: 37030567 DOI: 10.1016/j.bmcl.2023.129276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 04/03/2023] [Accepted: 04/04/2023] [Indexed: 04/08/2023]
Abstract
Small molecules as antibacterial agents have contributed immensely to the growth of modern medicine over the last several decades. However, the emergence of drug resistance among bacterial pathogens has undermined the effectiveness of the existing antibiotics. Thus, there is an exigency to address the antibiotic crisis by developing new antibacterial agents and identifying novel drug targets in bacteria. In this review, we summarize the importance of guanosine triphosphate hydrolyzing proteins (GTPases) as key agents for bacterial survival. We also discuss representative examples of small molecules that target bacterial GTPases as novel antibacterial agents, and highlight areas that are ripe for exploration. Given their vital roles in cell viability, virulence, and antibiotic resistance, bacterial GTPases are highly attractive antibacterial targets that will likely play a vital role in the fight against antimicrobial resistance.
Collapse
|
50
|
Gao P, Nasution AK, Yang S, Chen Z, Ono N, Kanaya S, Altaf-Ul-Amin MD. On Finding Natural Antibiotics based on TCM Formulae. Methods 2023; 214:35-45. [PMID: 37019293 DOI: 10.1016/j.ymeth.2023.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 02/12/2023] [Accepted: 04/01/2023] [Indexed: 04/05/2023] Open
Abstract
CONTEXT Novel kinds of antibiotics are needed to combat the emergence of antibacterial resistance. Natural products (NPs) have shown potential as antibiotic candidates. Current experimental methods are not yet capable of exploring the massive, redundant, and noise-involved chemical space of NPs. In silico approaches are needed to select NPs as antibiotic candidates. OBJECTIVE This study screens out NPs with antibacterial efficacy guided by both TCM and modern medicine and constructed a dataset aiming to serve the new antibiotic design. METHOD A knowledge-based network is proposed in this study involving NPs, herbs, the concepts of TCM, and the treatment protocols (or etiologies) of infectious in modern medicine. Using this network, the NPs candidates are screened out and compose the dataset. Feature selection of machine learning approaches is conducted to evaluate the constructed dataset and statistically validate the im- portance of all NPs candidates for different antibiotics by a classification task. RESULTS The extensive experiments prove the constructed dataset reaches a convincing classification performance with a 0.9421 weighted accuracy, 0.9324 recall, and 0.9409 precision. The further visu- alizations of sample importance prove the comprehensive evaluation for model interpretation based on medical value considerations.
Collapse
Affiliation(s)
- Pei Gao
- Nara Institute of Science and Technology (NAIST), Ikoma, Nara 630-0101, Japan
| | | | - Shuo Yang
- Nara Institute of Science and Technology (NAIST), Ikoma, Nara 630-0101, Japan
| | - Zheng Chen
- Osaka University, Suita, Osaka 567-0047, Japan
| | - Naoaki Ono
- Nara Institute of Science and Technology (NAIST), Ikoma, Nara 630-0101, Japan
| | - Shigehiko Kanaya
- Nara Institute of Science and Technology (NAIST), Ikoma, Nara 630-0101, Japan
| | - M D Altaf-Ul-Amin
- Nara Institute of Science and Technology (NAIST), Ikoma, Nara 630-0101, Japan.
| |
Collapse
|