1
|
Liu Q, Liu C, Wang S, Zhang L, Sun H, Liao X. Differing envelope composition of Gram-negative and Gram-positive bacteria controls the adhesion and bactericidal performance of nanoscale zero-valent iron. JOURNAL OF HAZARDOUS MATERIALS 2025; 489:137663. [PMID: 39987735 DOI: 10.1016/j.jhazmat.2025.137663] [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: 11/11/2024] [Revised: 01/18/2025] [Accepted: 02/17/2025] [Indexed: 02/25/2025]
Abstract
Zero-valent-iron (nZVI) is a candidate antimicrobial agent, and previous works revealed its varying inactivation performance on Gram-negative (G-) and Gram-positive (G+) bacteria, but the underlying mechanism remains ambiguous. Herein, we reported the easier inactivation of Escherichia coli (G-, E. coli) than Staphylococcus aureus (G+, S. aureus) by nZVI, and revealed the key role of cell-nZVI adsorption. nZVI adhered more massively on E. coli surface than on S. aureus, and subsequently led to more pronounced membrane damage of E. coli. Investigations of pH, zeta potential, and ionic strength ruled out the essential contribution of nZVI-bacteria electrostatic interaction due to the different surface charges of E. coli and S. aureus. Three-dimensional excitation emission matrix suggested that the extracellular polymeric substances of E. coli suffered more severe damage by nZVI and lead to greater exposure of membrane. Infrared spectra indicated that nZVI strongly bound with the membrane proteins of E. coli and destroyed the membrane components. By contrast, the bonding between nZVI and S. aureus was minimal because of the dominant multi-layered peptidoglycan. The enhanced nZVI adsorption and membrane disruption would result in magnified reactive oxygen species (ROS) generation and oxidative stress of E. coli. Moreover, the catalase activity normalized by ROS concentration of S. aureus was 14.9-fold higher than that of E. coli after nZVI treatment, suggesting the stronger antioxidative capability of S. aureus. Our findings highlight that the different envelope compositions and antioxidant capacities between G- and G+ bacteria were responsible for their varying susceptibility to nZVI.
Collapse
Affiliation(s)
- Qianhui Liu
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, PR China; Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, Institute of Environmental & Applied Chemistry, College of Chemistry, Central China Normal University, Wuhan 430079, PR China
| | - Congcong Liu
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, PR China
| | - Shaohui Wang
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, PR China
| | - Lizhi Zhang
- School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Hongwei Sun
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, Institute of Environmental & Applied Chemistry, College of Chemistry, Central China Normal University, Wuhan 430079, PR China.
| | - Xiaomei Liao
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, PR China.
| |
Collapse
|
2
|
Cui J, Sun Y, Wang L, Ji Y, Zhao H, Sun M, Guo Z, Dong F. Quaternary ammonium salts of chitosan containing aromatic ring: Synthesis, characterization, antimicrobial, antioxidant and cytotoxicity. Carbohydr Res 2025; 552:109431. [PMID: 40010275 DOI: 10.1016/j.carres.2025.109431] [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: 10/18/2024] [Revised: 02/17/2025] [Accepted: 02/18/2025] [Indexed: 02/28/2025]
Abstract
Quaternary ammonium salts of chitosan have been widely used in the development of anti-microbial biomaterials for their important role in inhibiting the growth of microorganisms. However, it is important to modify its structure to obtain more efficient and biologically active derivatives. Herein, a series of chitosan derivatives with antibacterial, antifungal, antioxidant, and non-cytotoxic properties was synthesized. These four quaternary ammonium salts of chitosan were prepared by incorporating 2-thiophenecarboxaldehyde, 2-furancarboxaldehyde, 2-pyridinecarboxaldehyde and benzaldehyde to form Schiff bases, followed by a reductive amination to obtain the chitosan N-derivatives, and quaternized by iodomethane. The inhibition rate of thiophenecarboxaldehyde chitosan trimethyl ammonium iodide (TpTMC) to B. cinerea could reach 93.78 % and the inhibition ability of pyridinecarboxaldehyde chitosan trimethyl ammonium iodide (PyTMC) to F. graminearum could reach 98.08 % at the concentration of 1.0 mg/mL. Furthermore, furancarboxaldehyde chitosan trimethyl ammonium iodide (FrTMC) exhibited a strong ability to scavenge hydroxyl radicals and DPPH radicals, especially the DPPH radicals scavenging ability was comparable to that of l-ascorbic acid at the concentration of 1.6 mg/mL. The B3LYP/6-311++G (d,p) basis set was employed to determine electronic properties, including HOMO-LUMO energies, and to analyze the chemical reactivity of the compounds. L929 cells were used to evaluate the cytotoxicity of the compounds at different concentrations (1-1000 μg/mL). The results demonstrated the diverse applications of aromatic ring-modified quaternary ammonium salts of chitosan, along with their significant antioxidant and antifungal effects against plant fungi. Therefore, these compounds have potential applications in the preparation of agricultural, biological, and medical materials.
Collapse
Affiliation(s)
- Jingmin Cui
- Key Laboratory of Coastal Biology and Bioresource Utilization, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, 264003, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yan Sun
- Key Laboratory of Coastal Biology and Bioresource Utilization, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, 264003, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Linqing Wang
- Key Laboratory of Coastal Biology and Bioresource Utilization, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, 264003, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yuting Ji
- Key Laboratory of Coastal Biology and Bioresource Utilization, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, 264003, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Huishuang Zhao
- School of Chemistry and Chemical Engineering, Yantai University, Yantai, 264003, China
| | - Mingzhi Sun
- Yantai Fuhui Agricultural Technology Co., Ltd, Yantai, 264000, China
| | - Zhanyong Guo
- Key Laboratory of Coastal Biology and Bioresource Utilization, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, 264003, China; College of Marine Life and Fisheries, Jiangsu Ocean University, Lianyungang, 222005, China.
| | - Fang Dong
- Key Laboratory of Coastal Biology and Bioresource Utilization, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, 264003, China; University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
3
|
Nabila TT, Damayanti E, Widada J. Exploration of antibacterial compounds from Bacillus velezensis BP1 against foodborne pathogens Staphylococcus aureus and Salmonella enterica Typhimurium using metabolomic and genomic approaches. J Food Prot 2025:100546. [PMID: 40403971 DOI: 10.1016/j.jfp.2025.100546] [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: 10/07/2024] [Revised: 05/12/2025] [Accepted: 05/14/2025] [Indexed: 05/24/2025]
Abstract
Food contamination by pathogenic microorganisms has become a significant issue. This study investigated the antibacterial compounds of a Bacillus isolate from stingless bee bread, Bacillus velezensis BP1, against foodborne pathogens Staphylococcus aureus ATCC 25923 and Salmonella enterica Typhimurium ATCC 14028 using the one strain many compounds (OSMAC), metabolomic, and genomic approaches. The culture media used were Tryptic Soy Broth (TSB), TSB with Chitosan (TSB-Chi), several synthetic broths composed of Mineral Salts with Glucose (MG) and Glucose-Fructose (MGF), Carboxymethyl Cellulose (CMC), and Starch Nitrate (SNB), to analyze diverse antibacterial compounds. Twelve extracts from the supernatant (S) and pellet (P) were screened using the microdilution method. P-TSB and S-TSB-Chi demonstrated the highest antibacterial effects, with inhibitory concentration (IC50) values of 253.5 ppm and 740.28 ppm, respectively. Fourier transform infrared spectroscopy (FTIR) analysis identified an amide I group contributing to extract clustering. Untargeted liquid chromatography-high resolution mass spectrometry (LC-HRMS) revealed six compounds significantly contributing to extract clustering. Enrichment analysis showed that chitosan was associated with the metabolic processes of pyrimidine and nucleotide metabolisms. Growth curve assay and scanning electron microscopy (SEM) confirmed the extracts' efficacy. The Bacillus isolate showed an average nucleotide identity (ANI) of 98.08% to Bacillus velezensis NRRL B-41580. Genome mining revealed twelve biosynthetic gene clusters, six 100% similar to known clusters. Molecular docking demonstrated that genome mining-derived bacillibactin and LC-HRMS-derived bis(4-ethylbenzylidene)sorbitol and cyclo(phenylalanyl-prolyl) exhibited the strongest binding affinities against four pathogens associated proteins, outperforming ampicillin. This study highlights Bacillus velezensis BP1's potential as a source of diverse antibacterial compounds.
Collapse
Affiliation(s)
- Tsania Taskia Nabila
- Study Program of Biotechnology, Graduate School of Universitas Gadjah Mada (UGM), Jl. Teknika Utara, Pogung, Mlati, Sleman, Yogyakarta 55281, Indonesia; Research Assistant of Food Microbiology Research Group, Research Center for Food Processing and Technology, National Research and Innovation Agency (BRIN), Jl. Jogja Wonosari KM 31.5, Gading, Playen, Gunungkidul 55861, Indonesia
| | - Ema Damayanti
- Research Center for Food Processing and Technology, BRIN, Jl. Jogja Wonosari KM 31.5, Gading, Playen, Gunungkidul 55861, Indonesia
| | - Jaka Widada
- Department of Agricultural Microbiology, Faculty of Agriculture, UGM Jl. Flora No 1, Bulaksumur, Yogyakarta 55281, Indonesia.
| |
Collapse
|
4
|
Tinajero-Trejo M, Aindow M, Pasquina-Lemonche L, Lafage L, Adedeji-Olulana AF, Sutton JAF, Wacnik K, Jia Y, Bilyk B, Yu W, Hobbs JK, Foster SJ. Control of morphogenesis during the Staphylococcus aureus cell cycle. SCIENCE ADVANCES 2025; 11:eadr5011. [PMID: 40215301 PMCID: PMC11988411 DOI: 10.1126/sciadv.adr5011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 03/06/2025] [Indexed: 04/14/2025]
Abstract
Bacterial cell division is a complex, multistage process requiring septum development while maintaining cell wall integrity. A dynamic, macromolecular protein complex, the divisome, tightly controls morphogenesis both spatially and temporally, but the mechanisms that tune septal progression are largely unknown. By studying conditional mutants of genes encoding DivIB, DivIC, and FtsL, an essential trimeric complex central to cell division in bacteria, we demonstrate that FtsL and DivIB play independent, hierarchical roles coordinating peptidoglycan synthesis across specific septal developmental checkpoints. They are required for the localization of downstream divisome components and the redistribution of peptidoglycan synthesis from the cell periphery to the septum. This is achieved by positive regulation of septum production and negative regulation of peripheral cell wall synthesis. Our analysis has led to a model for the coordination of cell division in Staphylococcus aureus, forming a framework for understanding how protein localization and function are integrated with cell wall structural dynamics across the bacteria.
Collapse
Affiliation(s)
- Mariana Tinajero-Trejo
- School of Biosciences, University of Sheffield, Sheffield, UK
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, Sheffield, UK
| | - Matthew Aindow
- School of Biosciences, University of Sheffield, Sheffield, UK
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, Sheffield, UK
| | - Laia Pasquina-Lemonche
- School of Biosciences, University of Sheffield, Sheffield, UK
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, Sheffield, UK
| | - Lucia Lafage
- School of Biosciences, University of Sheffield, Sheffield, UK
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, Sheffield, UK
| | - Abimbola Feyisara Adedeji-Olulana
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, Sheffield, UK
- Department of Physics and Astronomy, University of Sheffield, Sheffield, UK
| | - Joshua A. F. Sutton
- School of Biosciences, University of Sheffield, Sheffield, UK
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, Sheffield, UK
| | - Katarzyna Wacnik
- School of Biosciences, University of Sheffield, Sheffield, UK
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, Sheffield, UK
| | - Yaosheng Jia
- Department of Molecular Biosciences, University of South Florida, Tampa, FL, USA
| | - Bohdan Bilyk
- School of Biosciences, University of Sheffield, Sheffield, UK
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, Sheffield, UK
| | - Wenqi Yu
- Department of Molecular Biosciences, University of South Florida, Tampa, FL, USA
| | - Jamie K. Hobbs
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, Sheffield, UK
- Department of Physics and Astronomy, University of Sheffield, Sheffield, UK
| | - Simon J. Foster
- School of Biosciences, University of Sheffield, Sheffield, UK
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, Sheffield, UK
| |
Collapse
|
5
|
Vásquez A, Leidy C, Manrique-Moreno M. Lysyl-Phosphatidylglycerol: A Lipid Involved in the Resistance of Staphylococcus aureus to Antimicrobial Peptide Activity. Antibiotics (Basel) 2025; 14:349. [PMID: 40298503 PMCID: PMC12024345 DOI: 10.3390/antibiotics14040349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 03/25/2025] [Accepted: 03/26/2025] [Indexed: 04/30/2025] Open
Abstract
Lysyl-phosphatidylglycerol (lysyl-PG) is one of the major lipids found in bacterial membranes; it is synthesized by attaching lysine to the headgroup of phosphatidylglycerol. First identified in Staphylococcus aureus in 1964, lysyl-PG is now recognized as a virulence factor that protects Staphylococcus aureus from antimicrobial agents, such as cationic antimicrobial peptides and phospholipase A2 type IIA. Under normal growth conditions, Staphylococcus aureus membranes are negatively charged due to a high proportion of anionic lipids, such as phosphatidylglycerol and cardiolipin. This intrinsic anionic charge helps attract positively charged antimicrobial agents to the membrane surface, increasing their disruptive activity. The presence of lysyl-PG reduces electrostatic interactions, making the membrane less susceptible to cationic agents. The biosynthesis of lysyl-PG is mediated by the multiple peptide resistance factor (MprF) enzyme, which catalyzes the modification of phosphatidylglycerol and translocation of lysyl-PG to the outer membrane in the presence of antimicrobial agents. However, several studies indicate that lysyl-PG not only responds to the presence of antimicrobial agents but can fluctuate based on environmental factors such as oxygen availability and nutrient composition. Acidic conditions and nutrient-rich media often result in increased lysyl-PG production, suggesting that bacterial membranes can be resistant to cationic antimicrobial agents even in their native state. Recent studies propose that targeting MprF to inhibit lysyl-PG biosynthesis could be a promising strategy to counter antimicrobial resistance. This review highlights the role of lysyl-PG in modulating membrane charge and its influence on antimicrobial agent efficacy and discusses a possible strategy for treatment by targeting lysyl-PG synthesis.
Collapse
Affiliation(s)
- Andrea Vásquez
- Chemistry Institute, Faculty of Exact and Natural Sciences, University of Antioquia, A.A. 1226, Medellin 050010, Colombia;
| | - Chad Leidy
- Biophysics Group, Physics Department, Universidad de los Andes, Bogotá 111711, Colombia
| | - Marcela Manrique-Moreno
- Chemistry Institute, Faculty of Exact and Natural Sciences, University of Antioquia, A.A. 1226, Medellin 050010, Colombia;
| |
Collapse
|
6
|
Bîrcă AC, Minculescu MA, Niculescu AG, Hudiță A, Holban AM, Alberts A, Grumezescu AM. Nanoparticle-Enhanced Collagen Hydrogels for Chronic Wound Management. J Funct Biomater 2025; 16:91. [PMID: 40137370 PMCID: PMC11943201 DOI: 10.3390/jfb16030091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/26/2025] [Accepted: 02/28/2025] [Indexed: 03/27/2025] Open
Abstract
Chronic wound infections present a persistent medical challenge; however, advancements in wound dressings and antimicrobial nanomaterials offer promising solutions for improving healing outcomes. This study introduces a hydrothermal synthesis approach for producing zinc oxide (ZnO) and copper oxide (CuO) nanoparticles, subsequently incorporated into PLGA microspheres and embedded within collagen hydrogels. The nanoparticles' physicochemical properties were characterized using X-ray diffraction (XRD) to confirm crystalline structure, scanning electron microscopy (SEM) for surface morphology, and Fourier-transform infrared spectroscopy (FT-IR) to verify functional groups and successful hydrogel integration. The hydrogels were tested for antimicrobial activity against Staphylococcus aureus, Pseudomonas aeruginosa, and Candida albicans, which are key pathogens in chronic wounds. Biocompatibility was assessed using the human HaCat keratinocyte cell line. Both ZnO- and CuO-loaded hydrogels exhibited broad-spectrum antimicrobial efficacy. Cytocompatibility tests demonstrated that both ZnO- and CuO-loaded hydrogels sustain cell viability and proliferation, highlighting their biocompatibility and suitability for chronic wound healing applications, with superior biological performance of ZnO-loaded hydrogels. Furthermore, the distinct antimicrobial profiles of ZnO and CuO hydrogels suggest their tailored use based on wound microbial composition, with CuO hydrogels excelling in antibacterial applications and ZnO hydrogels showing potential for antifungal treatments. These results underscore the potential of nanoparticle-based collagen hydrogels as innovative therapeutic tools for managing chronic wounds.
Collapse
Affiliation(s)
- Alexandra Cătălina Bîrcă
- Department of Science and Engineering of Oxide Materials and Nanomaterials, National University of Science and Technology Politehnica Bucharest, 011061 Bucharest, Romania; (A.C.B.); (M.A.M.); (A.-G.N.); (A.M.G.)
| | - Mihai Alexandru Minculescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, National University of Science and Technology Politehnica Bucharest, 011061 Bucharest, Romania; (A.C.B.); (M.A.M.); (A.-G.N.); (A.M.G.)
| | - Adelina-Gabriela Niculescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, National University of Science and Technology Politehnica Bucharest, 011061 Bucharest, Romania; (A.C.B.); (M.A.M.); (A.-G.N.); (A.M.G.)
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 050657 Bucharest, Romania; (A.H.); (A.M.H.)
| | - Ariana Hudiță
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 050657 Bucharest, Romania; (A.H.); (A.M.H.)
- Faculty of Biology, University of Bucharest, 030018 Bucharest, Romania
| | - Alina Maria Holban
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 050657 Bucharest, Romania; (A.H.); (A.M.H.)
- Faculty of Biology, University of Bucharest, 030018 Bucharest, Romania
| | - Adina Alberts
- Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Alexandru Mihai Grumezescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, National University of Science and Technology Politehnica Bucharest, 011061 Bucharest, Romania; (A.C.B.); (M.A.M.); (A.-G.N.); (A.M.G.)
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 050657 Bucharest, Romania; (A.H.); (A.M.H.)
| |
Collapse
|
7
|
Nagaiah HP, Samsudeen MB, Augustus AR, Shunmugiah KP. In vitro evaluation of silver-zinc oxide-eugenol nanocomposite for enhanced antimicrobial and wound healing applications in diabetic conditions. DISCOVER NANO 2025; 20:14. [PMID: 39847138 PMCID: PMC11757845 DOI: 10.1186/s11671-025-04183-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 01/08/2025] [Indexed: 01/24/2025]
Abstract
Diabetic wounds with chronic infections present a significant challenge, exacerbated by the growing issue of antimicrobial resistance, which often leads to delayed healing and increased morbidity. This study introduces a novel silver-zinc oxide-eugenol (Ag+ZnO+EU) nanocomposite, specifically designed to enhance antimicrobial activity and promote wound healing. The nanocomposite was thoroughly characterized using advanced analytical techniques, confirming its nanoscale structure, stability and chemical composition. The Ag+ZnO+EU nanocomposite demonstrated potent antimicrobial efficacy against a range of wound associated pathogens, including standard and clinical isolates of Staphylococcus aureus, Pseudomonas aeruginosa and Candida albicans. Minimum inhibitory concentrations of Ag+ZnO+EU for standard and clinical isolates were significantly lower than those of the individual components, highlighting the synergistic effect of the nanocomposite. Time-kill assays revealed rapid microbial eradication, achieving complete sterility within 240-min. Importantly, the nanocomposite effectively eliminated persister-like cells, which are typically resistant to conventional treatments, suggesting a potential solution for persistent infections. In vitro scratch assays using human keratinocyte cells demonstrated that the Ag+ZnO+EU nanocomposite significantly accelerated wound closure, with near-complete healing observed within 24-h, indicating enhanced cell migration and tissue regeneration. Additionally, the nanocomposite showed potential antidiabetic effects by increasing glucose uptake up to 97.21% in an in vitro assay using 2-[N-(7-nitrobenz-2-oxa-1,3-diazol-4-yl) amino]-2-deoxy-D-glucose, a fluorescent glucose analog, suggesting potential applications beyond wound healing. These findings highlight the Ag+ZnO+EU nanocomposite as a promising candidate for addressing both antimicrobial resistance and impaired wound healing in diabetic contexts.
Collapse
|
8
|
Soliman MM, Elwahy AHM, Sayed AM, Ibrahim M, Dawoud MA, Ali SHM, Nady MTS, Hassan NA, Saad W, Abdelhamid IA. Synthesis and antimicrobial evaluation of a new hybrid bis-cyanoacrylamide-based-piperazine containing sulphamethoxazole moiety against rheumatoid arthritis-associated pathogens. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-024-03780-7. [PMID: 39831977 DOI: 10.1007/s00210-024-03780-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Accepted: 12/30/2024] [Indexed: 01/22/2025]
Abstract
Piperazine-based compounds have garnered significant attention due to their notable biological and pharmacological activities, making them essential in fine chemical and pharmaceutical applications. In this study, we managed to synthesize a novel hybrid bis-cyanoacrylamide bearing the piperazine core via phenoxymethyl linker and incorporating sulphamethoxazole moiety. The novel compound was fully characterized using different spectral data including 1H-NMR, 13C-NMR, and FTIR spectroscopy. Piperazine-based compounds were screened for in silico studies to understand the antimicrobial activity against infections that may contribute to rheumatoid arthritis symptoms. The tested piperazine compound was also evaluated for its antimicrobial activity against Aspergillus niger, Candida albicans, Enterococcus faecalis ATCC 29212, Staphylococcus aureus ATCC 29213, Pseudomonas aeuroginosa ATCC 27853, Escherichia coli ATCC 25922, and Klebsiella pneumoniae ATCC 700603. S. aureus showed the highest inhibition, with a zone diameter of 16.0 ± 1.0 mm at a concentration of 0.8 mg/ml. The minimal inhibitory concentration (MIC) for all bacterial species ranged from 5 to 40 mg/ml. In contrast, fungal species were the most resistant to the tested compound. Molecular docking studies were conducted to elucidate the interaction mechanisms, binding energies, and hydrogen bonding interactions within protein-ligand complexes. Molecular docking studies were performed against five bacterial proteins and two fungal proteins, including DNA gyrase subunit B (UniProt ID: Q839Z1), protein RecA of (UniProt ID: P0A7G6), cyclic AMP-AMP-AMP synthase (UniProt ID: P0DTF7), UDP-N-acetylglucosamine 1-carboxyvinyl transferase (UniProt ID: A0A1S5RKE3), and clumping factor A (UniProt ID: Q53653). The tested compound achieved the highest binding score of ∆G = - 10.9 kcal/mol at the cyclic AMP synthase active site (UniProt ID: P0DTF7), forming 26 interactions. The results demonstrated that the synthesized piperazine compound exhibits promising antibacterial and antifungal activities, highlighting its potential as a candidate for antimicrobial development.
Collapse
Affiliation(s)
- Mona M Soliman
- Department of Botany and Microbiology, Faculty of Science, Cairo University, Giza, 12613, Egypt
| | - Ahmed H M Elwahy
- Department of Chemistry, Faculty of Science, Cairo University, Giza, 12613, Egypt.
| | - Ahmed M Sayed
- Department of Botany and Microbiology, Faculty of Science, Cairo University, Giza, 12613, Egypt
| | - Mahmoud Ibrahim
- Department of Botany and Microbiology, Faculty of Science, Cairo University, Giza, 12613, Egypt
| | - Mohamed A Dawoud
- Department of Botany and Microbiology, Faculty of Science, Cairo University, Giza, 12613, Egypt
| | | | - Menna Tallah S Nady
- Department of Biotechnology, Faculty of Agriculture, Cairo University, Giza, 12613, Egypt
| | - Nada A Hassan
- Department of Zoology, Faculty of Science, Cairo University, Giza, 12613, Egypt
| | - Wessam Saad
- Department of Biotechnology, Faculty of Agriculture, Cairo University, Giza, 12613, Egypt
| | - Ismail A Abdelhamid
- Department of Chemistry, Faculty of Science, Cairo University, Giza, 12613, Egypt.
| |
Collapse
|
9
|
Olaru I, Stefanache A, Gutu C, Lungu II, Mihai C, Grierosu C, Calin G, Marcu C, Ciuhodaru T. Combating Bacterial Resistance by Polymers and Antibiotic Composites. Polymers (Basel) 2024; 16:3247. [PMID: 39683992 DOI: 10.3390/polym16233247] [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: 10/01/2024] [Revised: 11/09/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
(1) Background: Since the discovery of antibiotics in the first half of the 20th century, humans have abused this privilege, giving rise to antibiotic-resistant pathogens. Recent research has brought to light the use of antimicrobial peptides in polymers, hydrogels, and nanoparticles (NPs) as a newer and safer alternative to traditional antibiotics. (2) Methods: This review article is a synthesis of the scientific works published in the last 15 years, focusing on the synthesis of polymers with proven antimicrobial properties. (3) Results: After a critical review of the literature was made, information and data about the synthesis and antimicrobial activity of antibacterial polymers and NPs functionalized with antibiotics were extracted. Fluorinated surfactants such as the Quaterfluo® series presented significant antimicrobial effects and could be modulated to contain thioesters to boost this characteristic. Biopolymers like chitosan and starch were also doped with iodine and used as iodophors to deliver iodine atoms directly to pathogens, as well as being antimicrobial on their own. Quaternary phosphonium salts are known for their increased antimicrobial activity compared to ammonium-containing polymers and are more thermally stable. (4) Conclusions: In summary, polymers and polymeric NPs seem like future alternatives to traditional antibiotics. Future research is needed to determine functional doses for clinical use and their toxicity.
Collapse
Affiliation(s)
- Iulia Olaru
- Faculty of Medicine and Pharmacy, University "Dunarea de Jos", 47 Domneasca Str., 800008 Galati, Romania
| | - Alina Stefanache
- Faculty of Pharmacy, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Cristian Gutu
- Faculty of Medicine and Pharmacy, University "Dunarea de Jos", 47 Domneasca Str., 800008 Galati, Romania
| | - Ionut Iulian Lungu
- Faculty of Pharmacy, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Cozmin Mihai
- Faculty of Dental Medicine, "Apollonia" University of Iasi, 11 Pacurari Street, 700115 Iasi, Romania
| | - Carmen Grierosu
- Faculty of Dental Medicine, "Apollonia" University of Iasi, 11 Pacurari Street, 700115 Iasi, Romania
| | - Gabriela Calin
- Faculty of Dental Medicine, "Apollonia" University of Iasi, 11 Pacurari Street, 700115 Iasi, Romania
| | - Constantin Marcu
- Faculty of Medicine and Pharmacy, University "Dunarea de Jos", 47 Domneasca Str., 800008 Galati, Romania
| | - Tudor Ciuhodaru
- Faculty of Dental Medicine, "Apollonia" University of Iasi, 11 Pacurari Street, 700115 Iasi, Romania
| |
Collapse
|
10
|
Wang H, Kim SJ, Lei Y, Wang S, Wang H, Huang H, Zhang H, Tsung A. Neutrophil extracellular traps in homeostasis and disease. Signal Transduct Target Ther 2024; 9:235. [PMID: 39300084 PMCID: PMC11415080 DOI: 10.1038/s41392-024-01933-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 06/25/2024] [Accepted: 07/16/2024] [Indexed: 09/22/2024] Open
Abstract
Neutrophil extracellular traps (NETs), crucial in immune defense mechanisms, are renowned for their propensity to expel decondensed chromatin embedded with inflammatory proteins. Our comprehension of NETs in pathogen clearance, immune regulation and disease pathogenesis, has grown significantly in recent years. NETs are not only pivotal in the context of infections but also exhibit significant involvement in sterile inflammation. Evidence suggests that excessive accumulation of NETs can result in vessel occlusion, tissue damage, and prolonged inflammatory responses, thereby contributing to the progression and exacerbation of various pathological states. Nevertheless, NETs exhibit dual functionalities in certain pathological contexts. While NETs may act as autoantigens, aggregated NET complexes can function as inflammatory mediators by degrading proinflammatory cytokines and chemokines. The delineation of molecules and signaling pathways governing NET formation aids in refining our appreciation of NETs' role in immune homeostasis, inflammation, autoimmune diseases, metabolic dysregulation, and cancer. In this comprehensive review, we delve into the multifaceted roles of NETs in both homeostasis and disease, whilst discussing their potential as therapeutic targets. Our aim is to enhance the understanding of the intricate functions of NETs across the spectrum from physiology to pathology.
Collapse
Affiliation(s)
- Han Wang
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Susan J Kim
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Yu Lei
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shuhui Wang
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hui Wang
- Department of Medical Genetics, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hai Huang
- Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Hongji Zhang
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, USA.
| | - Allan Tsung
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
11
|
Xu J, Zhao Y, Zhou Y, Dai S, Zhu N, Meng Q, Fan S, Zhao W, Yuan X. Fungal Extracellular Vesicle Proteins with Potential in Biological Interaction. Molecules 2024; 29:4012. [PMID: 39274860 PMCID: PMC11396447 DOI: 10.3390/molecules29174012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/20/2024] [Accepted: 08/22/2024] [Indexed: 09/16/2024] Open
Abstract
Extracellular vesicles (EVs) are vesicle-like structures composed of lipid bilayers, which can be divided into apoptotic bodies, microbubbles and exosomes. They are nanoparticles used for the exchange of information between cells. EVs contains many substances, including protein. With the development of proteomics, we know more about the types and functions of protein in vesicles. The potential functions of proteins in the envelope are mainly discussed, including cell wall construction, fungal virulence transmission, signal transmission and redox reactions, which provides a new perspective for studying the interaction mechanism between fungi and other organisms. The fungal protein markers of EVs are also summarized, which provided an exploration tool for studying the mechanism of vesicles. In addition, the possible role of immune protein in the EVs in the treatment of human diseases is also discussed, which provides new ideas for vaccine development.
Collapse
Affiliation(s)
- Jingyan Xu
- College of Life Science, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, China
| | - Yujin Zhao
- College of Life Science, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, China
| | - Yanguang Zhou
- College of Life Science, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, China
| | - Shijie Dai
- College of Life Science, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, China
| | - Na Zhu
- College of Life Science, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, China
| | - Qingling Meng
- College of Life Science, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, China
| | - Sen Fan
- College of Life Science, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, China
| | - Weichun Zhao
- College of Life Science, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, China
| | - Xiaofeng Yuan
- College of Life Science, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, China
| |
Collapse
|
12
|
Bollinger KW, Müh U, Ocius KL, Apostolos AJ, Pires MM, Helm RF, Popham DL, Weiss DS, Ellermeier CD. Identification of a family of peptidoglycan transpeptidases reveals that Clostridioides difficile requires noncanonical cross-links for viability. Proc Natl Acad Sci U S A 2024; 121:e2408540121. [PMID: 39150786 PMCID: PMC11348318 DOI: 10.1073/pnas.2408540121] [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: 04/30/2024] [Accepted: 07/12/2024] [Indexed: 08/18/2024] Open
Abstract
Most bacteria are surrounded by a cell wall that contains peptidoglycan (PG), a large polymer composed of glycan strands held together by short peptide cross-links. There are two major types of cross-links, termed 4-3 and 3-3 based on the amino acids involved. 4-3 cross-links are created by penicillin-binding proteins, while 3-3 cross-links are created by L,D-transpeptidases (LDTs). In most bacteria, the predominant mode of cross-linking is 4-3, and these cross-links are essential for viability, while 3-3 cross-links comprise only a minor fraction and are not essential. However, in the opportunistic intestinal pathogen Clostridioides difficile, about 70% of the cross-links are 3-3. We show here that 3-3 cross-links and LDTs are essential for viability in C. difficile. We also show that C. difficile has five LDTs, three with a YkuD catalytic domain as in all previously known LDTs and two with a VanW catalytic domain, whose function was until now unknown. The five LDTs exhibit extensive functional redundancy. VanW domain proteins are found in many gram-positive bacteria but scarce in other lineages. We tested seven non-C. difficile VanW domain proteins and confirmed LDT activity in three cases. In summary, our findings uncover a previously unrecognized family of PG cross-linking enzymes, assign a catalytic function to VanW domains, and demonstrate that 3-3 cross-linking is essential for viability in C. difficile, the first time this has been shown in any bacterial species. The essentiality of LDTs in C. difficile makes them potential targets for antibiotics that kill C. difficile selectively.
Collapse
Affiliation(s)
- Kevin W. Bollinger
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA52242
| | - Ute Müh
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA52242
| | - Karl L. Ocius
- Department of Chemistry, University of Virginia, Charlottesville, VA22904
| | | | - Marcos M. Pires
- Department of Chemistry, University of Virginia, Charlottesville, VA22904
| | - Richard F. Helm
- Department of Biochemistry, Virginia Tech, Blacksburg, VA24061
| | - David L. Popham
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA24061
| | - David S. Weiss
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA52242
- Graduate Program in Genetics, University of Iowa, Iowa City, IA52242
| | - Craig D. Ellermeier
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA52242
- Graduate Program in Genetics, University of Iowa, Iowa City, IA52242
| |
Collapse
|
13
|
Ledger EVK, Edwards AM. Host-induced cell wall remodeling impairs opsonophagocytosis of Staphylococcus aureus by neutrophils. mBio 2024; 15:e0164324. [PMID: 39041819 PMCID: PMC11323798 DOI: 10.1128/mbio.01643-24] [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: 05/29/2024] [Accepted: 07/02/2024] [Indexed: 07/24/2024] Open
Abstract
The bacterial pathogen Staphylococcus aureus responds to the host environment by increasing the thickness of its cell wall. However, the impact of cell wall thickening on susceptibility to host defenses is unclear. Using bacteria incubated in human serum, we show that host-induced increases in cell wall thickness led to a reduction in the exposure of bound antibody and complement and a corresponding reduction in phagocytosis and killing by neutrophils. The exposure of opsonins bound to protein antigens or lipoteichoic acid (LTA) was most significantly reduced, while opsonization by IgG against wall teichoic acid or peptidoglycan was largely unaffected. Partial digestion of accumulated cell wall using the enzyme lysostaphin restored opsonin exposure and promoted phagocytosis and killing. Concordantly, the antibiotic fosfomycin inhibited cell wall remodeling and maintained the full susceptibility of S. aureus to opsonophagocytic killing by neutrophils. These findings reveal that host-induced changes to the S. aureus cell wall reduce the ability of the immune system to detect and kill this pathogen through reduced exposure of protein- and LTA-bound opsonins. IMPORTANCE Understanding how bacteria adapt to the host environment is critical in determining fundamental mechanisms of immune evasion, pathogenesis, and the identification of targets for new therapeutic approaches. Previous work demonstrated that Staphylococcus aureus remodels its cell envelope in response to host factors and we hypothesized that this may affect recognition by antibodies and thus killing by immune cells. As expected, incubation of S. aureus in human serum resulted in rapid binding of antibodies. However, as bacteria adapted to the serum, the increase in cell wall thickness resulted in a significant reduction in exposure of bound antibodies. This reduced antibody exposure, in turn, led to reduced killing by human neutrophils. Importantly, while antibodies bound to some cell surface structures became obscured, this was not the case for those bound to wall teichoic acid, which may have important implications for vaccine design.
Collapse
Affiliation(s)
- Elizabeth V. K. Ledger
- Centre for Bacterial Resistance Biology, Imperial College London, London, United Kingdom
| | - Andrew M. Edwards
- Centre for Bacterial Resistance Biology, Imperial College London, London, United Kingdom
| |
Collapse
|
14
|
Xu Z, Li Y, Xu A, Soteyome T, Yuan L, Ma Q, Seneviratne G, Li X, Liu J. Cell-wall-anchored proteins affect invasive host colonization and biofilm formation in Staphylococcus aureus. Microbiol Res 2024; 285:127782. [PMID: 38833832 DOI: 10.1016/j.micres.2024.127782] [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: 12/16/2023] [Revised: 05/21/2024] [Accepted: 05/21/2024] [Indexed: 06/06/2024]
Abstract
As a major human and animal pathogen, Staphylococcus aureus can attach to medical implants (abiotic surface) or host tissues (biotic surface), and further establish robust biofilms which enhances resistance and persistence to host immune system and antibiotics. Cell-wall-anchored proteins (CWAPs) covalently link to peptidoglycan, and largely facilitate the colonization of S. aureus on various surfaces (including adhesion and biofilm formation) and invasion into host cells (including adhesion, immune evasion, iron acquisition and biofilm formation). During biofilm formation, CWAPs function in adhesion, aggregation, collagen-like fiber network formation, and consortia formation. In this review, we firstly focus on the structural features of CWAPs, including their intracellular function and interactions with host cells, as well as the functions and ligand binding of CWAPs in different stages of S. aureus biofilm formation. Then, the roles of CWAPs in different biofilm processes with regards in development of therapeutic approaches are clarified, followed by the association between CWAPs genes and clonal lineages. By touching upon these aspects, we hope to provide comprehensive knowledge and clearer understanding on the CWAPs of S. aureus and their roles in biofilm formation, which may further aid in prevention and treatment infection and vaccine development.
Collapse
Affiliation(s)
- Zhenbo Xu
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Vegetable Protein Processing Ministry of Education, South China University of Technology, Guangzhou 510640, China; Department of Laboratory Medicine, the Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China.
| | - Yaqin Li
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Vegetable Protein Processing Ministry of Education, South China University of Technology, Guangzhou 510640, China
| | - Aijuan Xu
- Guangzhou Hybribio Medical Laboratory, Guangzhou 510730, China
| | - Thanapop Soteyome
- Home Economics Technology, Rajamangala University of Technology Phra Nakhon, Bangkok, Thailand
| | - Lei Yuan
- School of Food Science and Engineering, Yangzhou University, Yangzhou, Jiangsu 225127, China
| | - Qin Ma
- Sericultural & Agri-Food Research Institute Guangdong Academy of Agricultural Sciences/Key Laboratory of Functional Foods, Ministry of Agriculture /Guangdong Key Laboratory of Agricultural Products Processing, Guangzhou 510610, China
| | - Gamini Seneviratne
- National Institute of Fundamental Studies, Hantana road, Kandy, Sri Lanka
| | - Xuejie Li
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Vegetable Protein Processing Ministry of Education, South China University of Technology, Guangzhou 510640, China.
| | - Junyan Liu
- College of Light Industry and Food Science, Guangdong Provincial Key Laboratory of Lingnan Specialty Food Science and Technology, Academy of Contemporary Agricultural Engineering Innovations, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China; Key Laboratory of Green Processing and Intelligent Manufacturing of Lingnan Specialty Food, Ministry of Agriculture, Guangzhou 510225, China.
| |
Collapse
|
15
|
Long DR, Holmes EA, Lo HY, Penewit K, Almazan J, Hodgson T, Berger NF, Bishop ZH, Lewis JD, Waalkes A, Wolter DJ, Salipante SJ. Clinical and in vitro models identify distinct adaptations enhancing Staphylococcus aureus pathogenesis in human macrophages. PLoS Pathog 2024; 20:e1012394. [PMID: 38991026 PMCID: PMC11265673 DOI: 10.1371/journal.ppat.1012394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 07/23/2024] [Accepted: 07/04/2024] [Indexed: 07/13/2024] Open
Abstract
Staphylococcus aureus is a facultative intracellular pathogen of human macrophages, which facilitates chronic infection. The genotypes, pathways, and mutations influencing that phenotype remain incompletely explored. Here, we used two distinct strategies to ascertain S. aureus gene mutations affecting pathogenesis in macrophages. First, we analyzed isolates collected serially from chronic cystic fibrosis (CF) respiratory infections. We found that S. aureus strains evolved greater macrophage invasion capacity during chronic human infection. Bacterial genome-wide association studies (GWAS) identified 127 candidate genes for which mutation was significantly associated with macrophage pathogenesis in vivo. In parallel, we passaged laboratory S. aureus strains in vitro to select for increased infection of human THP-1 derived macrophages, which identified 15 candidate genes by whole-genome sequencing. Functional validation of candidate genes using isogenic transposon mutant knockouts and CRISPR interference (CRISPRi) knockdowns confirmed virulence contributions from 37 of 39 tested genes (95%) implicated by in vivo studies and 7 of 10 genes (70%) ascertained from in vitro selection, with one gene in common to the two strategies. Validated genes included 17 known virulence factors (39%) and 27 newly identified by our study (61%), some encoding functions not previously associated with macrophage pathogenesis. Most genes (80%) positively impacted macrophage invasion when disrupted, consistent with the phenotype readily arising from loss-of-function mutations in vivo. This work reveals genes and mechanisms that contribute to S. aureus infection of macrophages, highlights differences in mutations underlying convergent phenotypes arising from in vivo and in vitro systems, and supports the relevance of S. aureus macrophage pathogenesis during chronic respiratory infection in CF. Additional studies will be needed to illuminate the exact mechanisms by which implicated mutations affect their phenotypes.
Collapse
Affiliation(s)
- Dustin R. Long
- Division of Critical Care Medicine, Department of Anesthesiology and Pain Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Elizabeth A. Holmes
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Hsin-Yu Lo
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Kelsi Penewit
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Jared Almazan
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Taylor Hodgson
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Nova F. Berger
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Zoe H. Bishop
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Janessa D. Lewis
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Adam Waalkes
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Daniel J. Wolter
- Department of Pediatrics, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Stephen J. Salipante
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, United States of America
| |
Collapse
|
16
|
Wei PW, Wang X, Wang C, Chen M, Liu MZ, Liu WX, He YL, Xu GB, Zheng XH, Zhang H, Liu HM, Wang B. Ginkgo biloba L. exocarp petroleum ether extract inhibits methicillin-resistant Staphylococcus aureus by modulating ion transport, virulence, and biofilm formation in vitro and in vivo. JOURNAL OF ETHNOPHARMACOLOGY 2024; 328:117957. [PMID: 38493904 DOI: 10.1016/j.jep.2024.117957] [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: 01/04/2024] [Revised: 02/10/2024] [Accepted: 02/19/2024] [Indexed: 03/19/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE As reported in the Ancient Chinese Medicinal Books, Ginkgo biloba L. fruit has been used as a traditional Chinese medicine for the treatment asthma and cough or as a disinfectant. Our previous study demonstrated that G. biloba exocarp extract (GBEE), an extract of a traditional Chinese herb, inhibits the formation of methicillin-resistant Staphylococcus aureus (MRSA) biofilms. However, GBEE is a crude extract that contains many components, and the underlying mechanisms of purified GBEE fractions extracted with solvents of different polarities are unknown. AIM OF THE STUDY This study aimed to investigate the different components in GBEE fractions extracted with solvents of different polarities and their antibacterial effects and mechanisms against MRSA and Staphylococcus haemolyticus biofilms both in vitro and in vivo. METHODS The components in different fractions were detected by high-performance liquid chromatography-high resolution mass spectrometry (HPLC-HRMS). Microbroth dilution assays and time growth curves were used to determine the antibacterial effects of the fractions on 15 clinical bacterial isolates. Crystal violet staining, scanning electron microscopy (SEM) and transmission electron microscopy (TEM) were utilized to identify the fractions that affected bacterial biofilm formation. The potential MRSA targets of the GBEE fraction obtained with petroleum ether (PE), denoted GBEE-PE, were screened by transcriptome sequencing, and the gene expression profile was verified by quantitative polymerase chain reaction (qPCR). RESULTS HPLC-HRMS analysis revealed that the four GBEE fractions (extracted with petroleum ether, ethyl acetate, n-butanol, and water) contained different ginkgo components, and the antibacterial effects decreased as the polarity of the extraction solvent increased. The antibacterial activity of GBEE-PE was greater than that of the GBEE fraction extracted with ethyl acetate (EA). GBEE-PE improved H. illucens survival and reduced MRSA colonization in model mouse organs. Crystal violet staining and SEM and TEM analyses revealed that GBEE-PE inhibited MRSA and S. haemolyticus biofilm formation. Transcriptional analysis revealed that GBEE-PE inhibits MRSA biofilms by altering ion transport, cell wall metabolism and virulence-related gene expression. In addition, the LO2 cell viability and H. illucens toxicity assay data showed that GBEE-PE at 20 mg/kg was nontoxic. CONCLUSION The GBEE fractions contained different components, and their antibacterial effects decreased with increases in the polarity of the extraction solvent. GBEE-PE limited MRSA growth and biofilm formation by affecting ion transport, cell wall synthesis, and virulence-related pathways. This research provides a more detailed overview of the mechanism by which GBEE-PE inhibits MRSA both in vitro and in vivo and suggests that GBEE-PE is a new prospective antimicrobial with the potential to be used in MRSA therapeutics in the future.
Collapse
Affiliation(s)
- Peng-Wei Wei
- Engineering Research Center of Health Medicine Biotechnology of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering (Modern Industry College of Health Medicine), Guizhou Medical University, Guiyang, 561113, Guizhou, China
| | - Xu Wang
- Engineering Research Center of Health Medicine Biotechnology of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering (Modern Industry College of Health Medicine), Guizhou Medical University, Guiyang, 561113, Guizhou, China
| | - Cong Wang
- The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Formulation (R&D) Department, Guiyang, 550001, China
| | - Ming Chen
- Engineering Research Center of Health Medicine Biotechnology of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering (Modern Industry College of Health Medicine), Guizhou Medical University, Guiyang, 561113, Guizhou, China; Key Laboratory of Environmental Pollution Monitoring and Disease Control, China Ministry of Education (School of Public Health, Guizhou Medical University), Guiyang, 561113, Guizhou, China
| | - Meng-Zhu Liu
- Engineering Research Center of Health Medicine Biotechnology of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering (Modern Industry College of Health Medicine), Guizhou Medical University, Guiyang, 561113, Guizhou, China; Key Laboratory of Environmental Pollution Monitoring and Disease Control, China Ministry of Education (School of Public Health, Guizhou Medical University), Guiyang, 561113, Guizhou, China
| | - Wen-Xia Liu
- Engineering Research Center of Health Medicine Biotechnology of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering (Modern Industry College of Health Medicine), Guizhou Medical University, Guiyang, 561113, Guizhou, China; Key Laboratory of Microbiology and Parasitology of Education Department of Guizhou, School of Basic Medical Science, Guizhou Medical University, Guiyang, 561113, Guizhou, China
| | - Yan-Ling He
- Zhejiang Hisun Pharmaceutical Co., Ltd., Taizhou, 318000, Zhejiang, China
| | - Guo-Bo Xu
- State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmacy, Guizhou Medical University, Guian New Area, 561113, Guizhou, China.
| | - Xiao-He Zheng
- Zhejiang Hisun Pharmaceutical Co., Ltd., Taizhou, 318000, Zhejiang, China
| | - Hua Zhang
- Department of Laboratory Medicine, Guizhou Provincial People's Hospital, Affiliated Hospital of Guizhou University, Guiyang, 550002, Guizhou, China.
| | - Hong-Mei Liu
- Engineering Research Center of Health Medicine Biotechnology of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering (Modern Industry College of Health Medicine), Guizhou Medical University, Guiyang, 561113, Guizhou, China.
| | - Bing Wang
- Engineering Research Center of Health Medicine Biotechnology of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering (Modern Industry College of Health Medicine), Guizhou Medical University, Guiyang, 561113, Guizhou, China; Key Laboratory of Environmental Pollution Monitoring and Disease Control, China Ministry of Education (School of Public Health, Guizhou Medical University), Guiyang, 561113, Guizhou, China; Key Laboratory of Microbiology and Parasitology of Education Department of Guizhou, School of Basic Medical Science, Guizhou Medical University, Guiyang, 561113, Guizhou, China.
| |
Collapse
|
17
|
Mediati DG, Dan W, Lalaouna D, Dinh H, Pokhrel A, Rowell KN, Michie KA, Stinear TP, Cain AK, Tree JJ. The 3' UTR of vigR is required for virulence in Staphylococcus aureus and has expanded through STAR sequence repeat insertions. Cell Rep 2024; 43:114082. [PMID: 38583155 DOI: 10.1016/j.celrep.2024.114082] [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: 05/22/2023] [Revised: 01/17/2024] [Accepted: 03/25/2024] [Indexed: 04/09/2024] Open
Abstract
Infections caused by methicillin-resistant Staphylococcus aureus (MRSA) are alarmingly common, and treatment is confined to last-line antibiotics. Vancomycin is the treatment of choice for MRSA bacteremia, and treatment failure is often associated with vancomycin-intermediate S. aureus isolates. The regulatory 3' UTR of the vigR mRNA contributes to vancomycin tolerance and upregulates the autolysin IsaA. Using MS2-affinity purification coupled with RNA sequencing, we find that the vigR 3' UTR also regulates dapE, a succinyl-diaminopimelate desuccinylase required for lysine and peptidoglycan synthesis, suggesting a broader role in controlling cell wall metabolism and vancomycin tolerance. Deletion of the 3' UTR increased virulence, while the isaA mutant is completely attenuated in a wax moth larvae model. Sequence and structural analyses of vigR indicated that the 3' UTR has expanded through the acquisition of Staphylococcus aureus repeat insertions that contribute sequence for the isaA interaction seed and may functionalize the 3' UTR.
Collapse
Affiliation(s)
- Daniel G Mediati
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia; Australian Institute for Microbiology and Infection, University of Technology Sydney, Ultimo, NSW, Australia.
| | - William Dan
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia
| | - David Lalaouna
- Université de Strasbourg, CNRS, ARN UPR 9002, Strasbourg, France
| | - Hue Dinh
- School of Natural Sciences, ARC Centre of Excellence in Synthetic Biology, Macquarie University, Sydney, NSW, Australia
| | - Alaska Pokhrel
- Australian Institute for Microbiology and Infection, University of Technology Sydney, Ultimo, NSW, Australia; School of Natural Sciences, ARC Centre of Excellence in Synthetic Biology, Macquarie University, Sydney, NSW, Australia
| | - Keiran N Rowell
- Structural Biology Facility, University of New South Wales, Sydney, NSW, Australia
| | - Katharine A Michie
- Structural Biology Facility, University of New South Wales, Sydney, NSW, Australia
| | - Timothy P Stinear
- Department of Microbiology and Immunology, Peter Doherty Institute, University of Melbourne, Melbourne, VIC, Australia
| | - Amy K Cain
- School of Natural Sciences, ARC Centre of Excellence in Synthetic Biology, Macquarie University, Sydney, NSW, Australia
| | - Jai J Tree
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia.
| |
Collapse
|
18
|
Yang Y, Yao Z, Zhang J, Shao W, Li B, Wu H, Tang W, Zhang J. Inhibiting Peptidoglycan Hydrolase Alleviates MRSA Pneumonia Through Autolysin-Mediated MDP-NOD2 Pathway. Infect Drug Resist 2024; 17:1231-1242. [PMID: 38560705 PMCID: PMC10981453 DOI: 10.2147/idr.s455339] [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: 12/17/2023] [Accepted: 03/15/2024] [Indexed: 04/04/2024] Open
Abstract
Background Methicillin-resistant Staphylococcus aureus (MRSA) is a cause of staph infection that is difficult to treat because of resistance to some antibiotics. A recent study indicated that diarylurea ZJ-2 is a novel antibacterial agent against multi-drug resistant Enterococcus faecium. In this work, we refined the bactericidal mechanism of ZJ-2 as a peptidoglycan (PG) hydrolase by affecting AtlA-mediated PG homeostasis. Methods A wild-type strain (WT) and a mutant strain (ΔatlA) were used to investigate the effects of ZJ-2 on the cell wall, PG, and autolysin regulatory system by antimicrobial susceptibility testing, hemolytic toxin assay, microanalysis, autolysis assay, qRT-PCR, ELISA and mouse model of pneumonia. Results The results revealed that ZJ-2 down-regulated the expression of genes related to peptidoglycan hydrolase (PGH) (sprX, walR, atlA, and lytM), and reduced the levels of PG, muramyl dipeptide (MDP), cytokines, and hemolytic toxin, while ΔatlA interfered with the genes regulation and PG homeostasis. In the mouse MRSA pneumonia model, the same trend was observed in the nucleotide oligomerization domain protein 2 (NOD2) and relative proinflammatory factors. Conclusion ZJ-2 may act as a novel inhibitor of PG hydrolyse, disrupting autolysin-mediated PG homeostasis, and reducing inflammation by down-regulating the MDP-NOD2 pathway.
Collapse
Affiliation(s)
- Yang Yang
- School of Medicine, Anhui University of Science and Technology, Huainan, People’s Republic of China
- Anhui Province Key Laboratory of Occupational Health, Anhui No.2 Provincial People’s Hospital, Hefei, People’s Republic of China
| | - Zongze Yao
- School of Medicine, Anhui University of Science and Technology, Huainan, People’s Republic of China
| | - Jiazhen Zhang
- School of Medicine, Anhui University of Science and Technology, Huainan, People’s Republic of China
| | - Wei Shao
- School of Pharmacy, Anhui Medical University, Hefei, People’s Republic of China
| | - Bo Li
- Anhui Province Key Laboratory of Occupational Health, Anhui No.2 Provincial People’s Hospital, Hefei, People’s Republic of China
| | - Huihui Wu
- Anhui Province Key Laboratory of Occupational Health, Anhui No.2 Provincial People’s Hospital, Hefei, People’s Republic of China
| | - Wenjian Tang
- School of Pharmacy, Anhui Medical University, Hefei, People’s Republic of China
| | - Jing Zhang
- Anhui Province Key Laboratory of Occupational Health, Anhui No.2 Provincial People’s Hospital, Hefei, People’s Republic of China
| |
Collapse
|
19
|
Pottie I, Vázquez Fernández R, Van de Wiele T, Briers Y. Phage lysins for intestinal microbiome modulation: current challenges and enabling techniques. Gut Microbes 2024; 16:2387144. [PMID: 39106212 PMCID: PMC11305034 DOI: 10.1080/19490976.2024.2387144] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 07/05/2024] [Accepted: 07/26/2024] [Indexed: 08/09/2024] Open
Abstract
The importance of the microbiota in the intestinal tract for human health has been increasingly recognized. In this perspective, microbiome modulation, a targeted alteration of the microbial composition, has gained interest. Phage lysins, peptidoglycan-degrading enzymes encoded by bacteriophages, are a promising new class of antibiotics currently under clinical development for treating bacterial infections. Due to their high specificity, lysins are considered microbiome-friendly. This review explores the opportunities and challenges of using lysins as microbiome modulators. First, the high specificity of endolysins, which can be further modulated using protein engineering or targeted delivery methods, is discussed. Next, obstacles and possible solutions to assess the microbiome-friendliness of lysins are considered. Finally, lysin delivery to the intestinal tract is discussed, including possible delivery methods such as particle-based and probiotic vehicles. Mapping the hurdles to developing lysins as microbiome modulators and identifying possible ways to overcome these hurdles can help in their development. In this way, the application of these innovative antimicrobial agents can be expanded, thereby taking full advantage of their characteristics.
Collapse
Affiliation(s)
- Iris Pottie
- Laboratory of Applied Biotechnology, Department of Biotechnology, Ghent University, Gent, Belgium
- Center for Microbial Ecology and Technology (CMET), Faculty of Bioscience Engineering, Ghent University, Gent, Belgium
| | - Roberto Vázquez Fernández
- Laboratory of Applied Biotechnology, Department of Biotechnology, Ghent University, Gent, Belgium
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Tom Van de Wiele
- Center for Microbial Ecology and Technology (CMET), Faculty of Bioscience Engineering, Ghent University, Gent, Belgium
| | - Yves Briers
- Laboratory of Applied Biotechnology, Department of Biotechnology, Ghent University, Gent, Belgium
| |
Collapse
|
20
|
Valdivieso González D, Jara J, Almendro-Vedia VG, Orgaz B, López-Montero I. Expansion microscopy applied to mono- and dual-species biofilms. NPJ Biofilms Microbiomes 2023; 9:92. [PMID: 38049404 PMCID: PMC10696089 DOI: 10.1038/s41522-023-00460-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Accepted: 11/20/2023] [Indexed: 12/06/2023] Open
Abstract
Expansion microscopy (ExM) is a new super-resolution technique based on embedding the biological sample within a hydrogel and its physical expansion after swelling. This allows increasing its size by several times while preserving its structural details. Applied to prokaryotic cells, ExM requires digestion steps for efficient expansion as bacteria are surrounded by a rigid cell wall. Furthermore, bacteria can live in social groups forming biofilms, where cells are protected from environmental stresses by a self-produced matrix. The extracellular matrix represents an additional impenetrable barrier for ExM. Here we optimize the current protocols of ExM and apply them to mono- and dual-species biofilms formed by clinical isolates of Limosilactobacillus reuteri, Enterococcus faecalis, Serratia marcescens and Staphylococcus aureus. Using scanning electron microscopy for comparison, our results demonstrate that embedded bacteria expanded 3-fold. Moreover, ExM allowed visualizing the three-dimensional architecture of the biofilm and identifying the distribution of different microbial species and their interactions. We also detected the presence of the extracellular matrix after expansion with a specific stain of the polysaccharide component. The potential applications of ExM in biofilms will improve our understanding of these complex communities and have far-reaching implications for industrial and clinical research.
Collapse
Affiliation(s)
- David Valdivieso González
- Dto. Química Física, Universidad Complutense de Madrid, Avda. Complutense s/n, 28040, Madrid, Spain
- Instituto Pluridisciplinar, Universidad Complutense de Madrid, Ps. Juan XXIII 1, 28040, Madrid, Spain
- Instituto de Investigación Biomédica Hospital Doce de Octubre (Imas12), Avda. de Córdoba s/n, 28041, Madrid, Spain
| | - Josué Jara
- Sección Departamental de Nutrición y Ciencia de los Alimentos, Facultad de Veterinaria, Universidad Complutense de Madrid, 28040, Madrid, Spain
| | - Víctor G Almendro-Vedia
- Instituto Pluridisciplinar, Universidad Complutense de Madrid, Ps. Juan XXIII 1, 28040, Madrid, Spain
- Sección Departamental de Farmacia Galénica y Tecnología Alimentaria, Facultad de Veterinaria, Universidad Complutense de Madrid, Avda. Complutense s/n, 28040, Madrid, Spain
- Instituto de Investigación Biomédica Hospital Doce de Octubre (Imas12), Avda. de Córdoba s/n, 28041, Madrid, Spain
| | - Belén Orgaz
- Sección Departamental de Farmacia Galénica y Tecnología Alimentaria, Facultad de Veterinaria, Universidad Complutense de Madrid, Avda. Complutense s/n, 28040, Madrid, Spain
| | - Iván López-Montero
- Dto. Química Física, Universidad Complutense de Madrid, Avda. Complutense s/n, 28040, Madrid, Spain.
- Instituto Pluridisciplinar, Universidad Complutense de Madrid, Ps. Juan XXIII 1, 28040, Madrid, Spain.
- Instituto de Investigación Biomédica Hospital Doce de Octubre (Imas12), Avda. de Córdoba s/n, 28041, Madrid, Spain.
| |
Collapse
|
21
|
Chavarria D, Borges A, Benfeito S, Sequeira L, Ribeiro M, Oliveira C, Borges F, Simões M, Cagide F. Phytochemicals and quaternary phosphonium ionic liquids: Connecting the dots to develop a new class of antimicrobial agents. J Adv Res 2023; 54:251-269. [PMID: 36822390 DOI: 10.1016/j.jare.2023.02.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 02/03/2023] [Accepted: 02/08/2023] [Indexed: 02/24/2023] Open
Abstract
INTRODUCTION The infections by multidrug-resistant bacteria are a growing threat to human health, and the efficacy of the available antibiotics is gradually decreasing. As such, new antibiotic classes are urgently needed. OBJECTIVES This study aims to evaluate the antimicrobial activity, safety and mechanism of action of phytochemical-based triphenylphosphonium (TPP+) conjugates. METHODS A library of phytochemical-based TPP+ conjugates was repositioned and extended, and its antimicrobial activity was evaluated against a panel of Gram-positive (methicillin-resistant Staphylococcus aureus - MRSA) and Gram-negative bacteria (Escherichia coli, Klebsiella pneumoniae, Pseudomonas aeruginosa, Acinetobacter baumannii) and fungi (Candida albicans, Cryptococcus neoformans var. grubii). The compounds' cytotoxicity and haemolytic profile were also evaluated. To unravel the mechanism of action of the best compounds, the alterations in the surface charge, bacterial membrane integrity, and cytoplasmic leakage were assessed. RESULTS Structure-activity-toxicity data revealed the contributions of the different structural components (phenolic ring, carbon-based spacers, carboxamide group, alkyl linker) to the compounds' bioactivity and safety. Dihydrocinnamic derivatives 5 m and 5n stood out as safe, potent and selective antibacterial agents against S. aureus (MIC < 0.25 µg/mL; CC50 > 32 µg/mL; HC10 > 32 µg/mL). Mechanistic studies suggest that the antibacterial activity of compounds 5 m and 5n may result from interactions with the bacterial cell wall and membrane. CONCLUSIONS Collectively, these studies demonstrate the potential of phytochemical-based TPP+ conjugates as a new class of antibiotics.
Collapse
Affiliation(s)
- Daniel Chavarria
- CIQUP-IMS/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Rua do Campo Alegre s/n, 4169-007 Porto, Portugal
| | - Anabela Borges
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal; ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Sofia Benfeito
- CIQUP-IMS/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Rua do Campo Alegre s/n, 4169-007 Porto, Portugal
| | - Lisa Sequeira
- CIQUP-IMS/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Rua do Campo Alegre s/n, 4169-007 Porto, Portugal
| | - Marta Ribeiro
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Catarina Oliveira
- CIQUP-IMS/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Rua do Campo Alegre s/n, 4169-007 Porto, Portugal
| | - Fernanda Borges
- CIQUP-IMS/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Rua do Campo Alegre s/n, 4169-007 Porto, Portugal
| | - Manuel Simões
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal; ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal.
| | - Fernando Cagide
- CIQUP-IMS/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Rua do Campo Alegre s/n, 4169-007 Porto, Portugal.
| |
Collapse
|
22
|
Senevirathne SWMAI, Mathew A, Toh YC, Yarlagadda PKDV. Preferential adhesion of bacterial cells onto top- and bottom-mounted nanostructured surfaces under flow conditions. NANOSCALE ADVANCES 2023; 5:6458-6472. [PMID: 38024307 PMCID: PMC10662052 DOI: 10.1039/d3na00581j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 10/07/2023] [Indexed: 12/01/2023]
Abstract
The bactericidal effect of biomimetic nanostructured surfaces has been known for a long time, with recent data suggesting an enhanced efficiency of the nanostructured surfaces under fluid shear. While some of the influential factors on the bactericidal effect of nanostructured surfaces under fluid shear are understood, there are numerous important factors yet to be studied, which is essential for the successful implementation of this technology in industrial applications. Among those influential factors, the orientation of the nanostructured surface can play an important role in bacterial cell adhesion onto surfaces. Gravitational effects can become dominant under low flow velocities, making the diffusive transport of bacterial cells more prominent than the advective transport. However, the role of nanostructure orientation in determining its bactericidal efficiency under flow conditions is still not clear. In this study, we analysed the effect of surface orientation of nanostructured surfaces, along with bacterial cell concentration, fluid flow rate, and the duration of time which the surface is exposed to flow, on bacterial adhesion and viability on these surfaces. Two surface orientations, with one on the top and the other on the bottom of a flow channel, were studied. Under flow conditions, the bactericidal efficacy of the nanostructured surface is both orientation and bacterial species dependent. The effects of cell concentration, fluid flow rate, and exposure time on cell adhesion are independent of the nanostructured surface orientation. Fluid shear showed a species-dependent effect on bacterial adhesion, while the effects of concentration and exposure time on bacterial cell adhesion are independent of the bacterial species. Moreover, bacterial cells demonstrate preferential adhesion onto surfaces based on the surface orientation, and these effects are species dependent. These results outline the capabilities and limitations of nanostructures under flow conditions. This provides valuable insights into the applications of nanostructures in medical or industrial sectors, which are associated with overlaying fluid flow.
Collapse
Affiliation(s)
- S W M A Ishantha Senevirathne
- Queensland University of Technology, Faculty of Engineering, School of Mechanical, Medical, and Process Engineering Brisbane QLD 4000 Australia
- Queensland University of Technology, Centre for Biomedical Technologies Brisbane QLD 4000 Australia
| | - Asha Mathew
- Queensland University of Technology, Faculty of Engineering, School of Mechanical, Medical, and Process Engineering Brisbane QLD 4000 Australia
- Queensland University of Technology, Centre for Biomedical Technologies Brisbane QLD 4000 Australia
| | - Yi-Chin Toh
- Queensland University of Technology, Faculty of Engineering, School of Mechanical, Medical, and Process Engineering Brisbane QLD 4000 Australia
- Queensland University of Technology, Centre for Biomedical Technologies Brisbane QLD 4000 Australia
| | - Prasad K D V Yarlagadda
- School of Engineering, University of Southern Queensland, Springfield Campus Springfield Central QLD 4300 Australia
| |
Collapse
|
23
|
Sutton JAF, Cooke M, Tinajero-Trejo M, Wacnik K, Salamaga B, Portman-Ross C, Lund VA, Hobbs JK, Foster SJ. The roles of GpsB and DivIVA in Staphylococcus aureus growth and division. Front Microbiol 2023; 14:1241249. [PMID: 37711690 PMCID: PMC10498921 DOI: 10.3389/fmicb.2023.1241249] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 08/04/2023] [Indexed: 09/16/2023] Open
Abstract
The spheroid bacterium Staphylococcus aureus is often used as a model of morphogenesis due to its apparently simple cell cycle. S. aureus has many cell division proteins that are conserved across bacteria alluding to common functions. However, despite intensive study, we still do not know the roles of many of these components. Here, we have examined the functions of the paralogues DivIVA and GpsB in the S. aureus cell cycle. Cells lacking gpsB display a more spherical phenotype than the wild-type cells, which is associated with a decrease in peripheral cell wall peptidoglycan synthesis. This correlates with increased localization of penicillin-binding proteins at the developing septum, notably PBPs 2 and 3. Our results highlight the role of GpsB as an apparent regulator of cell morphogenesis in S. aureus.
Collapse
Affiliation(s)
- Joshua A. F. Sutton
- School of Biosciences, University of Sheffield, Sheffield, United Kingdom
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, Sheffield, United Kingdom
| | - Mark Cooke
- School of Biosciences, University of Sheffield, Sheffield, United Kingdom
| | - Mariana Tinajero-Trejo
- School of Biosciences, University of Sheffield, Sheffield, United Kingdom
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, Sheffield, United Kingdom
| | - Katarzyna Wacnik
- School of Biosciences, University of Sheffield, Sheffield, United Kingdom
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, Sheffield, United Kingdom
| | - Bartłomiej Salamaga
- School of Biosciences, University of Sheffield, Sheffield, United Kingdom
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, Sheffield, United Kingdom
| | - Callum Portman-Ross
- School of Biosciences, University of Sheffield, Sheffield, United Kingdom
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, Sheffield, United Kingdom
| | - Victoria A. Lund
- School of Biosciences, University of Sheffield, Sheffield, United Kingdom
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, Sheffield, United Kingdom
| | - Jamie K. Hobbs
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, Sheffield, United Kingdom
- Department of Physics and Astronomy, University of Sheffield, Sheffield, United Kingdom
| | - Simon J. Foster
- School of Biosciences, University of Sheffield, Sheffield, United Kingdom
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
24
|
Szepe CK, Kafle A, Bhattarai S, Handy ST, Farone MB. Evaluation of the Antibacterial Effect of Aurone-Derived Triazoles on Staphylococcus aureus. Antibiotics (Basel) 2023; 12:1370. [PMID: 37760667 PMCID: PMC10525585 DOI: 10.3390/antibiotics12091370] [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: 08/01/2023] [Revised: 08/22/2023] [Accepted: 08/24/2023] [Indexed: 09/29/2023] Open
Abstract
Infections caused by antibiotic-resistant bacteria continue to pose a significant public health threat despite their overall decreasing numbers in the last two decades. One group of compounds fundamental to the search for new agents is low-cost natural products. In this study, we explored a group of newly synthesized novel aurone-derived triazole compounds to identify those with pharmaceutical potential as inhibitors of antibiotic-resistant Staphylococcus aureus. Using the broth microdilution method, antibacterial activities against methicillin-resistant S. aureus ATCC 43300 (MRSA) and methicillin-sensitive S. aureus ATCC 29213 (MSSA) were identified for four aurone-derived triazole compounds, AT106, AT116, AT125, and AT137, using the half-maximal inhibitory concentrations for the bacteria (IC50) and mammalian cell lines (CC50). Compounds AT125 and AT137 were identified to have pharmaceutical potential as the IC50 values against MRSA were 5.412 µM and 3.870 µM, whereas the CC50 values measured on HepG2 cells were 50.57 µM and 39.81 µM, respectively, resulting in selectivity indexes (SI) > 10. Compounds AT106 and AT116 were also selected for further study. IC50 values for these compounds were 5.439 µM and 3.178 µM, and the CC50 values were 60.33 µM and 50.87 µM, respectively; however, SI values > 10 were for MSSA only. Furthermore, none of the selected compounds showed significant hemolytic activity for human erythrocytes. We also tested the four compounds against S. aureus biofilms. Although AT116 and AT125 successfully disrupted MSSA biofilms, there was no measurable potency against MRSA biofilms. Checkerboard antibiotic assays to identify inhibitory mechanisms for these compounds indicated activity against bacterial cell membranes and cell walls, supporting the pharmaceutical potential for aurone-derived triazoles against antibiotic-resistant bacteria. Examining structure-activity relationships between the four compounds in this study and other aurone-derived triazoles in our library suggest that substitution with a halogen on either the salicyl ring or triazole aryl group along with triazoles having nitrile groups improves anti-Staphylococcal activity with the location of the functionality being very important.
Collapse
Affiliation(s)
- Csilla Klara Szepe
- Department of Biology, Middle Tennessee State University, Murfreesboro, TN 37132, USA;
| | - Arjun Kafle
- Department of Chemistry, Middle Tennessee State University, Murfreesboro, TN 37132, USA (S.T.H.)
| | - Shrijana Bhattarai
- Department of Chemistry, Middle Tennessee State University, Murfreesboro, TN 37132, USA (S.T.H.)
| | - Scott T. Handy
- Department of Chemistry, Middle Tennessee State University, Murfreesboro, TN 37132, USA (S.T.H.)
| | - Mary B. Farone
- Department of Biology, Middle Tennessee State University, Murfreesboro, TN 37132, USA;
| |
Collapse
|
25
|
Volokhov DV, Zagorodnyaya TA, Furtak VA, Nattanmai G, Randall L, Jose S, Gao Y, Eisenberg T, Delmonte P, Blom J, Mitchell KK. Streptococcus sciuri sp. nov., Staphylococcus marylandisciuri sp. nov. and Staphylococcus americanisciuri sp. nov., isolated from faeces of eastern grey squirrel ( Sciurus carolinensis). Int J Syst Evol Microbiol 2023; 73. [PMID: 37578899 DOI: 10.1099/ijsem.0.006015] [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: 08/16/2023] Open
Abstract
One novel Streptococcus strain (SQ9-PEAT) and two novel Staphylococcus strains (SQ8-PEAT and GRT3T) were isolated from faeces of a wild eastern grey squirrel. The strains were non-spore-forming, non-motile Gram-positive cocci, facultative anaerobes. The genomes for these strains were sequenced. The 16S rRNA gene and core-genome-based phylogenetic analyses showed that strain SQ9-PEAT was closely related to Streptococcus hyointestinalis, strain SQ8-PEAT to Staphylococcus pettenkoferi and Staphylococcus argensis, and strain GRT3T to Staphylococcus rostri, Staphylococcus muscae and Staphylococcus microti. Average nucleotide identity and pairwise digital DNA-DNA hybridization values calculated for these novel strains compared to type strain genomes of phylogenetically related species within the genera Streptococcus and Staphylococcus clearly revealed that strain SQ9-PEAT represents a novel species of the genus Streptococcus and strains SQ8-PEAT and GRT3T represent two novel species of the genus Staphylococcus. Phenotypical features of these novel type strains differed from the features of the type strains of other phylogenetically related species. MALDI-TOF mass spectrometry supported identification of these novel species. Based on these data, we propose one novel species of the genus Streptococcus, for which the name Streptococcus sciuri sp. nov. with the type strain SQ9-PEAT (=DSM 114656T=CCUG 76426T=NCTC 14727T) is proposed, and two novel species of the genus Staphylococcus, for which the names Staphylococcus marylandisciuri sp. nov. with the type strain SQ8-PEAT (=DSM 114685T=CCUG 76423T=NCTC 14723T) and Staphylococcus americanisciuri sp. nov. with the type strain GRT3T (=DSM 114696T=CCUG 76427T=NCTC 14722T) are proposed. The genome G+C contents are 38.29, 36.49 and 37.26 mol% and complete draft genome sizes are 1 692 266, 2 371 088 and 2 237 001 bp for strains SQ9-PEAT, SQ8-PEAT and GRT3T, respectively.
Collapse
Affiliation(s)
- Dmitriy V Volokhov
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD 20993, USA
| | - Tatiana A Zagorodnyaya
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD 20993, USA
| | - Vyacheslav A Furtak
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD 20993, USA
| | - Geetha Nattanmai
- Bacteriology Laboratory, Wadsworth Center, the New York State Department of Health, 120 New Scotland Ave, Albany, NY 12208, USA
| | - Linnell Randall
- Bacteriology Laboratory, Wadsworth Center, the New York State Department of Health, 120 New Scotland Ave, Albany, NY 12208, USA
| | - Sherly Jose
- Bacteriology Laboratory, Wadsworth Center, the New York State Department of Health, 120 New Scotland Ave, Albany, NY 12208, USA
| | - Yamei Gao
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD 20993, USA
| | - Tobias Eisenberg
- Hessian State Laboratory (LHL), Department of Veterinary Medicine, Schubertstrasse 60, 35392 Giessen, Germany
| | - Pierluigi Delmonte
- Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, 5001 Campus Drive, College Park, MD 20740, USA
| | - Jochen Blom
- Bioinformatics and Systems Biology, Justus-Liebig-University Giessen, Heinrich Buff Ring 58, 35392 Giessen, Germany
| | - Kara K Mitchell
- Bacteriology Laboratory, Wadsworth Center, the New York State Department of Health, 120 New Scotland Ave, Albany, NY 12208, USA
| |
Collapse
|
26
|
Tătaru AM, Canciu A, Tertiș M, Cristea C, Cernat A. Staphylococcus aureus - Review on potential targets for sensors development. Bioelectrochemistry 2023; 153:108492. [PMID: 37413820 DOI: 10.1016/j.bioelechem.2023.108492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/14/2023] [Accepted: 06/19/2023] [Indexed: 07/08/2023]
Abstract
Staphylococcus aureus (S. aureus) is accountable for a wide variety of clinical disease with a high rate of morbidity and mortality around the globe. It has a leading place into the ESKAPE group that includes six pathogens and exhibit multidrug resistance and are the major cause of healthcare associated infections: Enterococcus faecium, S. aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter spp. A critical overview regarding the development of sensors for both S. aureus and his, more dangerous alter ego, Methicillin-resistant S. aureus (MRSA) was presented focusing on the bacteria targets starting with the detection of the whole cell, up to specific wall components, toxins or other virulence factors. The literature data was systematically assessed having in sight the design of the sensing platforms, the analytical performances, and possible courses of action to be implemented in real practice as point-of-care (POC) devices. Moreover, a distinct section was dedicated to commercially available devices and out of the box approaches, namely the use of bacteriophages as an alternative to antimicrobial therapy and as sensors modifiers. The reviewed sensors and devices were discussed in terms of their suitability for different biosensing applications, in early screening of contamination regarding food analysis, environmental monitoring and in clinical diagnosis.
Collapse
Affiliation(s)
- Ana-Maria Tătaru
- Analytical Chemistry Department, Faculty of Pharmacy, Iuliu Haţieganu University of Medicine and Pharmacy, 4 Louis Pasteur St., 400349 Cluj-Napoca, Romania
| | - Alexandra Canciu
- Analytical Chemistry Department, Faculty of Pharmacy, Iuliu Haţieganu University of Medicine and Pharmacy, 4 Louis Pasteur St., 400349 Cluj-Napoca, Romania
| | - Mihaela Tertiș
- Analytical Chemistry Department, Faculty of Pharmacy, Iuliu Haţieganu University of Medicine and Pharmacy, 4 Louis Pasteur St., 400349 Cluj-Napoca, Romania
| | - Cecilia Cristea
- Analytical Chemistry Department, Faculty of Pharmacy, Iuliu Haţieganu University of Medicine and Pharmacy, 4 Louis Pasteur St., 400349 Cluj-Napoca, Romania.
| | - Andreea Cernat
- Analytical Chemistry Department, Faculty of Pharmacy, Iuliu Haţieganu University of Medicine and Pharmacy, 4 Louis Pasteur St., 400349 Cluj-Napoca, Romania
| |
Collapse
|
27
|
Lu Q, Regan DP, Barlow DE, Fears KP. Antimicrobial efficacy of cyclic α- and β-peptides incorporated in polyurethane coatings. Biointerphases 2023; 18:031008. [PMID: 37289032 DOI: 10.1116/6.0002515] [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: 01/23/2023] [Accepted: 05/12/2023] [Indexed: 06/09/2023] Open
Abstract
Microbial growth on surfaces poses health concerns and can accelerate the biodegradation of engineered materials and coatings. Cyclic peptides are promising agents to combat biofouling because they are more resistant to enzymatic degradation than their linear counterparts. They can also be designed to interact with extracellular targets and intracellular targets and/or self-assemble into transmembrane pores. Here, we determine the antimicrobial efficacy of two pore-forming cyclic peptides, α-K3W3 and β-K3W3, against bacterial and fungal liquid cultures and their capacity to inhibit biofilm formation on coated surfaces. These peptides display identical sequences, but the additional methylene group in the peptide backbone of β-amino acids results in a larger diameter and an enhancement in the dipole moment. In liquid cultures, β-K3W3 exhibited lower minimum inhibitory concentration values and greater microbicidal power in reducing the number of colony forming units (CFUs) when exposed to a gram-positive bacterium, Staphylococcus aureus, and two fungal strains, Naganishia albida and Papiliotrema laurentii. To evaluate the efficacy against the formation of fungal biofilms on painted surfaces, cyclic peptides were incorporated into polyester-based thermoplastic polyurethane. The formation of N. albida and P. laurentii microcolonies (105 per inoculation) for cells extracted from coatings containing either peptide could not be detected after a 7-day exposure. Moreover, very few CFUs (∼5) formed after 35 days of repeated depositions of freshly cultured P. laurentii every 7 days. In contrast, the number of CFUs for cells extracted from the coating without cyclic peptides was >8 log CFU.
Collapse
Affiliation(s)
- Qin Lu
- Chemistry Division, U.S. Naval Research Laboratory, 4555 Overlook Ave SW, Washington, DC 20375
| | - Daniel P Regan
- Chemistry Division, U.S. Naval Research Laboratory, 4555 Overlook Ave SW, Washington, DC 20375
| | - Daniel E Barlow
- Chemistry Division, U.S. Naval Research Laboratory, 4555 Overlook Ave SW, Washington, DC 20375
| | - Kenan P Fears
- Chemistry Division, U.S. Naval Research Laboratory, 4555 Overlook Ave SW, Washington, DC 20375
| |
Collapse
|
28
|
Salamaga B, Turner RD, Elsarmane F, Galley NF, Kulakauskas S, Mesnage S. A moonlighting role for LysM peptidoglycan binding domains underpins Enterococcus faecalis daughter cell separation. Commun Biol 2023; 6:428. [PMID: 37072531 PMCID: PMC10113225 DOI: 10.1038/s42003-023-04808-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 04/04/2023] [Indexed: 04/20/2023] Open
Abstract
Control of cell size and morphology is of paramount importance for bacterial fitness. In the opportunistic pathogen Enterococcus faecalis, the formation of diplococci and short cell chains facilitates innate immune evasion and dissemination in the host. Minimisation of cell chain size relies on the activity of a peptidoglycan hydrolase called AtlA, dedicated to septum cleavage. To prevent autolysis, AtlA activity is tightly controlled, both temporally and spatially. Here, we show that the restricted localization of AtlA at the septum occurs via an unexpected mechanism. We demonstrate that the C-terminal LysM domain that allows the enzyme to bind peptidoglycan is essential to target this enzyme to the septum inside the cell before its translocation across the membrane. We identify a membrane-bound cytoplasmic protein partner (called AdmA) involved in the recruitment of AtlA via its LysM domains. This work reveals a moonlighting role for LysM domains, and a mechanism evolved to restrict the subcellular localization of a potentially lethal autolysin to its site of action.
Collapse
Affiliation(s)
| | - Robert D Turner
- School of Biosciences, University of Sheffield, Sheffield, UK
| | - Fathe Elsarmane
- School of Biosciences, University of Sheffield, Sheffield, UK
| | - Nicola F Galley
- School of Biosciences, University of Sheffield, Sheffield, UK
| | - Saulius Kulakauskas
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, 78350, Jouy-en-Josas, France
| | | |
Collapse
|
29
|
Lu Y, Chen F, Zhao Q, Cao Q, Chen R, Pan H, Wang Y, Huang H, Huang R, Liu Q, Li M, Bae T, Liang H, Lan L. Modulation of MRSA virulence gene expression by the wall teichoic acid enzyme TarO. Nat Commun 2023; 14:1594. [PMID: 36949052 PMCID: PMC10032271 DOI: 10.1038/s41467-023-37310-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 03/10/2023] [Indexed: 03/24/2023] Open
Abstract
Phenol-soluble modulins (PSMs) and Staphylococcal protein A (SpA) are key virulence determinants for community-associated methicillin-resistant Staphylococcus aureus (CA-MRSA), an important human pathogen that causes a wide range of diseases. Here, using chemical and genetic approaches, we show that inhibition of TarO, the first enzyme in the wall teichoic acid (WTA) biosynthetic pathway, decreases the expression of genes encoding PSMs and SpA in the prototypical CA-MRSA strain USA300 LAC. Mechanistically, these effects are linked to the activation of VraRS two-component system that directly represses the expression of accessory gene regulator (agr) locus and spa. The activation of VraRS was due in part to the loss of the functional integrity of penicillin-binding protein 2 (PBP2) in a PBP2a-dependent manner. TarO inhibition can also activate VraRS in a manner independent of PBP2a. We provide multiple lines of evidence that accumulation of lipid-linked peptidoglycan precursors is a trigger for the activation of VraRS. In sum, our results reveal that WTA biosynthesis plays an important role in the regulation of virulence gene expression in CA-MRSA, underlining TarO as an attractive target for anti-virulence therapy. Our data also suggest that acquisition of PBP2a-encoding mecA gene can impart an additional regulatory layer for the modulation of key signaling pathways in S. aureus.
Collapse
Affiliation(s)
- Yunfu Lu
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, China
| | - Feifei Chen
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- College of Life Science, Northwest University, Xi'an, 710127, China
| | - Qingmin Zhao
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, China
| | - Qiao Cao
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- College of Life Science, Northwest University, Xi'an, 710127, China
| | - Rongrong Chen
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, China
| | - Huiwen Pan
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, China
| | - Yanhui Wang
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, China
| | - Haixin Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Ruimin Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, China
| | - Qian Liu
- Department of Laboratory Medicine, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Min Li
- Department of Laboratory Medicine, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Taeok Bae
- Department of Microbiology and Immunology, Indiana University School of Medicine-Northwest, Gary, IN, 46408, USA
| | - Haihua Liang
- College of Life Science, Northwest University, Xi'an, 710127, China.
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, China.
| | - Lefu Lan
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China.
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, China.
- College of Life Science, Northwest University, Xi'an, 710127, China.
| |
Collapse
|
30
|
Abramov VM, Kosarev IV, Machulin AV, Priputnevich TV, Deryusheva EI, Nemashkalova EL, Chikileva IO, Abashina TN, Panin AN, Melnikov VG, Suzina NE, Nikonov IN, Selina MV, Khlebnikov VS, Sakulin VK, Samoilenko VA, Gordeev AB, Sukhikh GT, Uversky VN, Karlyshev AV. Limosilactobacillus fermentum 3872 That Produces Class III Bacteriocin Forms Co-Aggregates with the Antibiotic-Resistant Staphylococcus aureus Strains and Induces Their Lethal Damage. Antibiotics (Basel) 2023; 12:antibiotics12030471. [PMID: 36978338 PMCID: PMC10044573 DOI: 10.3390/antibiotics12030471] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 02/19/2023] [Accepted: 02/23/2023] [Indexed: 03/03/2023] Open
Abstract
LF3872 was isolated from the milk of a healthy lactating and breastfeeding woman. Earlier, the genome of LF3872 was sequenced, and a gene encoding unique bacteriocin was discovered. We have shown here that the LF3872 strain produces a novel thermolabile class III bacteriolysin (BLF3872), exhibiting antimicrobial activity against antibiotic-resistant Staphylococcus aureus strains. Sequence analysis revealed the two-domain structural (lysozyme-like domain and peptidase M23 domain) organization of BLF3872. At least 25% residues of this protein are expected to be intrinsically disordered. Furthermore, BLF3872 is predicted to have a very high liquid-liquid phase separation. According to the electron microscopy data, the bacterial cells of LF3872 strain form co-aggregates with the S. aureus 8325-4 bacterial cells. LF3872 produced bacteriolysin BLF3872 that lyses the cells of the S. aureus 8325-4 mastitis-inducing strain. The sensitivity of the antibiotic-resistant S. aureus collection strains and freshly isolated antibiotic-resistant strains was tested using samples from women with lactation mastitis; the human nasopharynx and oral cavity; the oropharynx of pigs; and the cows with a diagnosis of clinical mastitis sensitive to the lytic action of the LF3872 strain producing BLF3872. The co-cultivation of LF3872 strain with various antibiotic-resistant S. aureus strains for 24 h reduced the level of living cells of these pathogens by six log. The LF3872 strain was found to be able to co-aggregate with all studied S. aureus strains. The cell-free culture supernatant of LF3872 (CSLF3872) induced S. aureus cell damage and ATP leakage. The effectiveness of the bacteriolytic action of LF3872 strain did not depend on the origin of the S. aureus strains. The results reported here are important for the creation of new effective drugs against antibiotic-resistant strains of S. aureus circulating in humans and animals.
Collapse
Affiliation(s)
- Vyacheslav M. Abramov
- Federal Service for Veterinary and Phytosanitary Surveillance (Rosselkhoznadzor) Federal State Budgetary Institution “The Russian State Center for Animal Feed and Drug Standardization and Quality” (FGBU VGNKI), 123022 Moscow, Russia
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Health, 117997 Moscow, Russia
- Correspondence:
| | - Igor V. Kosarev
- Federal Service for Veterinary and Phytosanitary Surveillance (Rosselkhoznadzor) Federal State Budgetary Institution “The Russian State Center for Animal Feed and Drug Standardization and Quality” (FGBU VGNKI), 123022 Moscow, Russia
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Health, 117997 Moscow, Russia
| | - Andrey V. Machulin
- Skryabin Institute of Biochemistry and Physiology of Microorganisms, Federal Research Center “Pushchino Scientific Center for Biological Research of Russian Academy of Science”, Russian Academy of Science, 142290 Pushchino, Russia
| | - Tatiana V. Priputnevich
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Health, 117997 Moscow, Russia
| | - Evgenia I. Deryusheva
- Institute for Biological Instrumentation, Federal Research Center “Pushchino Scientific Center for Biological Research of Russian Academy of Science”, Russian Academy of Science, 142290 Pushchino, Russia
| | - Ekaterina L. Nemashkalova
- Institute for Biological Instrumentation, Federal Research Center “Pushchino Scientific Center for Biological Research of Russian Academy of Science”, Russian Academy of Science, 142290 Pushchino, Russia
| | - Irina O. Chikileva
- Laboratory of Cell Immunity, Blokhin National Research Center of Oncology, Ministry of Health RF, 115478 Moscow, Russia
| | - Tatiana N. Abashina
- Skryabin Institute of Biochemistry and Physiology of Microorganisms, Federal Research Center “Pushchino Scientific Center for Biological Research of Russian Academy of Science”, Russian Academy of Science, 142290 Pushchino, Russia
| | - Alexander N. Panin
- Federal Service for Veterinary and Phytosanitary Surveillance (Rosselkhoznadzor) Federal State Budgetary Institution “The Russian State Center for Animal Feed and Drug Standardization and Quality” (FGBU VGNKI), 123022 Moscow, Russia
| | - Vyacheslav G. Melnikov
- Gabrichevsky Research Institute for Epidemiology and Microbiology, 125212 Moscow, Russia
| | - Nataliya E. Suzina
- Skryabin Institute of Biochemistry and Physiology of Microorganisms, Federal Research Center “Pushchino Scientific Center for Biological Research of Russian Academy of Science”, Russian Academy of Science, 142290 Pushchino, Russia
| | - Ilia N. Nikonov
- Federal State Educational Institution of Higher Professional Education, Moscow State Academy of Veterinary Medicine and Biotechnology named after K.I. Skryabin, 109472 Moscow, Russia
| | - Marina V. Selina
- Federal State Educational Institution of Higher Professional Education, Moscow State Academy of Veterinary Medicine and Biotechnology named after K.I. Skryabin, 109472 Moscow, Russia
| | | | - Vadim K. Sakulin
- Institute of Immunological Engineering, 142380 Lyubuchany, Russia
| | - Vladimir A. Samoilenko
- Skryabin Institute of Biochemistry and Physiology of Microorganisms, Federal Research Center “Pushchino Scientific Center for Biological Research of Russian Academy of Science”, Russian Academy of Science, 142290 Pushchino, Russia
| | - Alexey B. Gordeev
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Health, 117997 Moscow, Russia
| | - Gennady T. Sukhikh
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Health, 117997 Moscow, Russia
| | - Vladimir N. Uversky
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Andrey V. Karlyshev
- Department of Biomolecular Sciences, Faculty of Health, Science, Social Care and Education, Kingston University London, Kingston upon Thames KT1 2EE, UK
| |
Collapse
|
31
|
Resko ZJ, Anderson CM, Federle MJ, Alonzo F. A Staphylococcal Glucosaminidase Drives Inflammatory Responses by Processing Peptidoglycan Chains to Physiological Lengths. Infect Immun 2023; 91:e0050022. [PMID: 36715551 PMCID: PMC9933629 DOI: 10.1128/iai.00500-22] [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: 11/07/2022] [Accepted: 01/10/2023] [Indexed: 01/31/2023] Open
Abstract
The peptidoglycan of Staphylococcus aureus is a critical cell envelope constituent and virulence factor that subverts host immune defenses and provides protection against environmental stressors. Peptidoglycan chains of the S. aureus cell wall are processed to characteristically short lengths by the glucosaminidase SagB. It is well established that peptidoglycan is an important pathogen-associated molecular pattern (PAMP) that is recognized by the host innate immune system and promotes production of proinflammatory cytokines, including interleukin-1β (IL-1β). However, how bacterial processing of peptidoglycan drives IL-1β production is comparatively unexplored. Here, we tested the involvement of staphylococcal glucosaminidases in shaping innate immune responses and identified SagB as a mediator of IL-1β production. A ΔsagB mutant fails to promote IL-1β production by macrophages and dendritic cells, and processing of peptidoglycan by SagB is essential for this response. SagB-dependent IL-1β production by macrophages is independent of canonical pattern recognition receptor engagement and NLRP3 inflammasome-mediated caspase activity. Instead, treatment of macrophages with heat-killed cells from a ΔsagB mutant leads to reduced caspase-independent cleavage of pro-IL-1β, resulting in accumulation of the pro form in the macrophage cytosol. Furthermore, SagB is required for virulence in systemic infection and promotes IL-1β production in a skin and soft tissue infection model. Taken together, our results suggest that the length of S. aureus cell wall glycan chains can drive IL-1β production by innate immune cells through a previously undescribed mechanism related to IL-1β maturation.
Collapse
Affiliation(s)
- Zachary J. Resko
- Department of Microbiology and Immunology, Loyola University Chicago—Stritch School of Medicine, Maywood, Illinois, USA
| | - Caleb M. Anderson
- Department of Pharmaceutical Sciences, University of Illinois at Chicago—College of Pharmacy, Chicago, Illinois, USA
| | - Michael J. Federle
- Department of Pharmaceutical Sciences, University of Illinois at Chicago—College of Pharmacy, Chicago, Illinois, USA
| | - Francis Alonzo
- Department of Microbiology and Immunology, University of Illinois at Chicago—College of Medicine, Chicago, Illinois, USA
| |
Collapse
|
32
|
Nikolic P, Mudgil P. The Cell Wall, Cell Membrane and Virulence Factors of Staphylococcus aureus and Their Role in Antibiotic Resistance. Microorganisms 2023; 11:microorganisms11020259. [PMID: 36838224 PMCID: PMC9965861 DOI: 10.3390/microorganisms11020259] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 01/13/2023] [Accepted: 01/17/2023] [Indexed: 01/20/2023] Open
Abstract
Antibiotic resistant strains of bacteria are a serious threat to human health. With increasing antibiotic resistance in common human pathogens, fewer antibiotics remain effective against infectious diseases. Staphylococcus aureus is a pathogenic bacterium of particular concern to human health as it has developed resistance to many of the currently used antibiotics leaving very few remaining as effective treatment. Alternatives to conventional antibiotics are needed for treating resistant bacterial infections. A deeper understanding of the cellular characteristics of resistant bacteria beyond well characterized resistance mechanisms can allow for increased ability to properly treat them and to potentially identify targetable changes. This review looks at antibiotic resistance in S aureus in relation to its cellular components, the cell wall, cell membrane and virulence factors. Methicillin resistant S aureus bacteria are resistant to most antibiotics and some strains have even developed resistance to the last resort antibiotics vancomycin and daptomycin. Modifications in cell wall peptidoglycan and teichoic acids are noted in antibiotic resistant bacteria. Alterations in cell membrane lipids affect susceptibility to antibiotics through surface charge, permeability, fluidity, and stability of the bacterial membrane. Virulence factors such as adhesins, toxins and immunomodulators serve versatile pathogenic functions in S aureus. New antimicrobial strategies can target cell membrane lipids and virulence factors including anti-virulence treatment as an adjuvant to traditional antibiotic therapy.
Collapse
|
33
|
Satishkumar N, Lai LY, Mukkayyan N, Vogel BE, Chatterjee SS. A Nonclassical Mechanism of β-Lactam Resistance in Methicillin-Resistant Staphylococcus aureus and Its Effect on Virulence. Microbiol Spectr 2022; 10:e0228422. [PMID: 36314912 PMCID: PMC9769611 DOI: 10.1128/spectrum.02284-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 10/07/2022] [Indexed: 11/07/2022] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is a group of pathogenic bacteria that are infamously resistant to β-lactam antibiotics, a property attributed to the mecA gene. Recent studies have reported that mutations associated with the promoter region of pbp4 demonstrated high levels of β-lactam resistance, suggesting the role of PBP4 as an important non-mecA mediator of β-lactam resistance. The pbp4-promoter-associated mutations have been detected in strains with or without mecA. Our previous studies that were carried out in strains devoid of mecA described that pbp4-promoter-associated mutations lead to PBP4 overexpression and β-lactam resistance. In this study, by introducing various pbp4-promoter-associated mutations in the genome of a MRSA strain, we demonstrate that PBP4 overexpression can supplement mecA-associated resistance in S. aureus and can lead to increased β-lactam resistance. The promoter and regulatory region of pbp4 is shared with a divergently transcribed gene, abcA, which encodes a multidrug exporter. We demonstrate that the promoter mutations caused an upregulation of pbp4 and downregulation of abcA, confirming that the resistant phenotype is associated with PBP4 overexpression. PBP4 has also been associated with staphylococcal pathogenesis, however, its exact role remains unclear. Using a Caenorhabditis elegans model, we demonstrate that strains having increased PBP4 expression are less virulent than wild-type strains, suggesting that β-lactam resistance mediated via PBP4 likely comes at the cost of virulence. IMPORTANCE Our study demonstrates the ability of PBP4 to be an important mediator of β-lactam resistance in not only methicillin-susceptible Staphylococcus aureus (MSSA) background strains as previously demonstrated but also in MRSA strains. When present together, PBP2a and PBP4 overexpression can produce increased levels of β-lactam resistance, causing complications in treatment. Thus, this study suggests the importance of monitoring PBP4-associated resistance in clinical settings, as well as understanding the mechanistic basis of associated resistance, so that treatments targeting PBP4 may be developed. This study also demonstrates that S. aureus strains with increased PBP4 expression are less pathogenic, providing important hints about the role of PBP4 in S. aureus resistance and pathogenesis.
Collapse
Affiliation(s)
- Nidhi Satishkumar
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, Baltimore, Maryland, USA
- Institute of Marine and Environmental Technology (IMET), Baltimore, Maryland, USA
| | - Li-Yin Lai
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, Baltimore, Maryland, USA
- Institute of Marine and Environmental Technology (IMET), Baltimore, Maryland, USA
| | - Nagaraja Mukkayyan
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, Baltimore, Maryland, USA
- Institute of Marine and Environmental Technology (IMET), Baltimore, Maryland, USA
| | - Bruce E. Vogel
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, University of Maryland, Baltimore, Maryland, USA
| | - Som S. Chatterjee
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, Baltimore, Maryland, USA
- Institute of Marine and Environmental Technology (IMET), Baltimore, Maryland, USA
| |
Collapse
|
34
|
The Staphylococcus aureus cell division protein, DivIC, interacts with the cell wall and controls its biosynthesis. Commun Biol 2022; 5:1228. [DOI: 10.1038/s42003-022-04161-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 10/24/2022] [Indexed: 11/13/2022] Open
Abstract
AbstractBacterial cell division is a complex, dynamic process that requires multiple protein components to orchestrate its progression. Many division proteins are highly conserved across bacterial species alluding to a common, basic mechanism. Central to division is a transmembrane trimeric complex involving DivIB, DivIC and FtsL in Gram-positives. Here, we show a distinct, essential role for DivIC in division and survival of Staphylococcus aureus. DivIC spatially regulates peptidoglycan synthesis, and consequently cell wall architecture, by influencing the recruitment to the division septum of the major peptidoglycan synthetases PBP2 and FtsW. Both the function of DivIC and its recruitment to the division site depend on its extracellular domain, which interacts with the cell wall via binding to wall teichoic acids. DivIC facilitates the spatial and temporal coordination of peptidoglycan synthesis with the developing architecture of the septum during cell division. A better understanding of the cell division mechanisms in S. aureus and other pathogenic microorganisms can provide possibilities for the development of new, more effective treatments for bacterial infections.
Collapse
|
35
|
Yang R, Wei Y, Zhao M, Shi M, Zhao Y, Sun P. PBA functionalized single-atom Fe for efficient therapy of multidrug-resistant bacterial infections. Colloids Surf B Biointerfaces 2022; 219:112811. [PMID: 36067683 DOI: 10.1016/j.colsurfb.2022.112811] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/22/2022] [Accepted: 08/25/2022] [Indexed: 10/31/2022]
Abstract
The abuse of antibiotics has led to the emergence of multidrug-resistant bacterial strains worldwide, which greatly threatens human health. In the present work, we developed single-atom catalysts (SACs) with atomically dispersed Fe as catalytic sites (Fe-SACs) to combat multidrug-resistant bacteria by elevating cellular reactive oxygen species (ROS). Our intensive studies confirmed that Fe-SACs were successfully prepared and exhibited excellent catalase (CAT)-, oxidase (OXD)-, and peroxidase (POD)-like activities. To enhance water dispersibility, biosafety and the interactions between the nanodrugs and gram-positive bacteria, phenylboronic acid group-functionalized carboxylated chitosan (CCS-PBA) was coated on the surface of Fe-SACs to yield Fe-SACs@CCS-PBA for in vitro and in vivo studies. The synergistic catalytic activity and photothermal activity of Fe-SACs@CCS-PBA effectively overcame multidrug-resistant bacterial strains (MRSA) in vitro and significantly accelerated wound healing in vivo, suggesting the great potential of SACs to overcome infectious disease caused by multidrug-resistant bacteria.
Collapse
Affiliation(s)
- Ruigeng Yang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, PR China
| | - Yueru Wei
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, PR China
| | - Mengyang Zhao
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, PR China
| | - Mengxiao Shi
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, PR China
| | - Yongxing Zhao
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, PR China; Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou 450001, Henan, PR China; Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, PR China.
| | - Pengchao Sun
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, PR China; Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou 450001, Henan, PR China; Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, PR China.
| |
Collapse
|
36
|
Apostolos AJ, Kelly JJ, Ongwae GM, Pires MM. Structure Activity Relationship of the Stem Peptide in Sortase A Mediated Ligation from Staphylococcus aureus. Chembiochem 2022; 23:e202200412. [PMID: 36018606 PMCID: PMC9632411 DOI: 10.1002/cbic.202200412] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/24/2022] [Indexed: 01/11/2023]
Abstract
The surfaces of most Gram-positive bacterial cells, including that of Staphylococcus aureus (S. aureus), are heavily decorated with proteins that coordinate cellular interactions with the extracellular space. In S. aureus, sortase A is the principal enzyme responsible for covalently anchoring proteins, which display the sorting signal LPXTG, onto the peptidoglycan (PG) matrix. Considerable efforts have been made to understand the role of this signal peptide in the sortase-mediated reaction. In contrast, much less is known about how the primary structure of the other substrate involved in the reaction (PG stem peptide) could impact sortase activity. To assess the sortase activity, a library of synthetic analogs of the stem peptide that mimic naturally existing variations found in the S. aureus PG primary sequence were evaluated. Using a combination of two unique assays, we showed that there is broad tolerability of substrate variations that are effectively processed by sortase A. While some of these stem peptide derivatives are naturally found in mature PG, they are not known to be present in the PG precursor, lipid II. These results suggest that sortase A could process both lipid II and mature PG as acyl-acceptor strands that might reside near the membrane, which has not been previously described.
Collapse
Affiliation(s)
| | - Joey J. Kelly
- Department of ChemistryUniversity of VirginiaCharlottesville, VA22904USA
| | - George M. Ongwae
- Department of ChemistryUniversity of VirginiaCharlottesville, VA22904USA
| | - Marcos M. Pires
- Department of ChemistryUniversity of VirginiaCharlottesville, VA22904USA
| |
Collapse
|
37
|
Uskoković V, Wu VM. Altering Microbiomes with Hydroxyapatite Nanoparticles: A Metagenomic Analysis. MATERIALS (BASEL, SWITZERLAND) 2022; 15:5824. [PMID: 36079205 PMCID: PMC9456825 DOI: 10.3390/ma15175824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 08/18/2022] [Accepted: 08/19/2022] [Indexed: 06/15/2023]
Abstract
Hydroxyapatite (HAp), the most abundant biological material among mammals, has been recently demonstrated to possess moderate antibacterial properties. Metagenomics provides a series of tools for analyzing the simultaneous interaction of materials with larger communities of microbes, which may aid in optimizing the antibacterial activity of a material such as HAp. Here, a microbiome intrinsic to the sample of sandy soil collected from the base of an African Natal plum (Carissa macrocarpa) shrub surrounding the children's sandbox at the Arrowhead Park in Irvine, California was challenged with HAp nanoparticles and analyzed with next-generation sequencing for hypervariable 16S ribosomal DNA base pair homologies. HAp nanoparticles overwhelmingly reduced the presence of Gram-negative phyla, classes, orders, families, genera and species, and consequently elevated the relative presence of their Gram-positive counterparts. Thermodynamic, electrostatic and chemical bonding arguments were combined in a model proposed to explain this selective affinity. The ability of amphiphilic surface protrusions of lipoteichoic acid in Gram-positive bacteria and mycolic acid in mycobacteria to increase the dispersibility of the bacterial cells and assist in their resistance to capture by the solid phase is highlighted. Within the Gram-negative group, the variability of the distal, O-antigen portion of the membrane lipopolysaccharide was shown to be excessive and the variability of its proximal, lipid A portion insufficient to explain the selectivity based on chemical sequence arguments. Instead, flagella-driven motility proves to be a factor favoring the evasion of binding to HAp. HAp displayed a preference toward binding to less pathogenic bacteria than those causative of disease in humans, while taxa having a positive agricultural effect were largely captured by HAp, indicating an evolutionary advantage this may have given it as a biological material. The capacity to selectively sequester Gram-negative microorganisms and correspondingly alter the composition of the microbiome may open up a new avenue in environmental and biomedical applications of HAp.
Collapse
Affiliation(s)
- Vuk Uskoković
- TardigradeNano LLC, Irvine, CA 92604, USA;
- Department of Mechanical Engineering, San Diego State University, San Diego, CA 92182, USA
| | | |
Collapse
|
38
|
Penicillin-Binding Protein 1 (PBP1) of Staphylococcus aureus Has Multiple Essential Functions in Cell Division. mBio 2022; 13:e0066922. [PMID: 35703435 PMCID: PMC9426605 DOI: 10.1128/mbio.00669-22] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Bacterial cell division is a complex process requiring the coordination of multiple components to allow the appropriate spatial and temporal control of septum formation and cell scission. Peptidoglycan (PG) is the major structural component of the septum, and our recent studies in the human pathogen Staphylococcus aureus have revealed a complex, multistage PG architecture that develops during septation. Penicillin-binding proteins (PBPs) are essential for the final steps of PG biosynthesis; their transpeptidase activity links the peptide side chains of nascent glycan strands. PBP1 is required for cell division in S. aureus, and here, we demonstrate that it has multiple essential functions associated with its enzymatic activity and as a regulator of division. Loss of PBP1, or just its C-terminal PASTA domains, results in cessation of division at the point of septal plate formation. The PASTA domains can bind PG and thereby potentially coordinate the cell division process. The transpeptidase activity of PBP1 is also essential, but its loss leads to a strikingly different phenotype of thickened and aberrant septa, which is phenocopied by the morphological effects of adding the PBP1-specific β-lactam, meropenem. Together, these results lead to a model for septal PG synthesis where PBP1 enzyme activity is required for the characteristic architecture of the septum and PBP1 protein molecules enable the formation of the septal plate.
Collapse
|
39
|
Wang M, Buist G, van Dijl JM. Staphylococcus aureus cell wall maintenance - the multifaceted roles of peptidoglycan hydrolases in bacterial growth, fitness, and virulence. FEMS Microbiol Rev 2022; 46:6604383. [PMID: 35675307 PMCID: PMC9616470 DOI: 10.1093/femsre/fuac025] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 04/22/2022] [Accepted: 05/25/2022] [Indexed: 01/07/2023] Open
Abstract
Staphylococcus aureus is an important human and livestock pathogen that is well-protected against environmental insults by a thick cell wall. Accordingly, the wall is a major target of present-day antimicrobial therapy. Unfortunately, S. aureus has mastered the art of antimicrobial resistance, as underscored by the global spread of methicillin-resistant S. aureus (MRSA). The major cell wall component is peptidoglycan. Importantly, the peptidoglycan network is not only vital for cell wall function, but it also represents a bacterial Achilles' heel. In particular, this network is continuously opened by no less than 18 different peptidoglycan hydrolases (PGHs) encoded by the S. aureus core genome, which facilitate bacterial growth and division. This focuses attention on the specific functions executed by these enzymes, their subcellular localization, their control at the transcriptional and post-transcriptional levels, their contributions to staphylococcal virulence and their overall importance in bacterial homeostasis. As highlighted in the present review, our understanding of the different aspects of PGH function in S. aureus has been substantially increased over recent years. This is important because it opens up new possibilities to exploit PGHs as innovative targets for next-generation antimicrobials, passive or active immunization strategies, or even to engineer them into effective antimicrobial agents.
Collapse
Affiliation(s)
- Min Wang
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, PO Box 30001, 9700 RB Groningen, the Netherlands
| | | | - Jan Maarten van Dijl
- Corresponding author: Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P.O. box 30001, HPC EB80, 9700 RB Groningen, the Netherlands, Tel. +31-50-3615187; Fax. +31-50-3619105; E-mail:
| |
Collapse
|
40
|
Ledger EVK, Mesnage S, Edwards AM. Human serum triggers antibiotic tolerance in Staphylococcus aureus. Nat Commun 2022; 13:2041. [PMID: 35440121 PMCID: PMC9018823 DOI: 10.1038/s41467-022-29717-3] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 03/30/2022] [Indexed: 12/13/2022] Open
Abstract
Staphylococcus aureus frequently causes infections that are challenging to treat, leading to high rates of persistent and relapsing infection. Here, to understand how the host environment influences treatment outcomes, we study the impact of human serum on staphylococcal antibiotic susceptibility. We show that serum triggers a high degree of tolerance to the lipopeptide antibiotic daptomycin and several other classes of antibiotic. Serum-induced daptomycin tolerance is due to two independent mechanisms. Firstly, the host defence peptide LL-37 induces tolerance by triggering the staphylococcal GraRS two-component system, leading to increased peptidoglycan accumulation. Secondly, GraRS-independent increases in membrane cardiolipin abundance are required for full tolerance. When both mechanisms are blocked, S. aureus incubated in serum is as susceptible to daptomycin as when grown in laboratory media. Our work demonstrates that host factors can significantly modulate antibiotic susceptibility via diverse mechanisms, and combination therapy may provide a way to mitigate this.
Collapse
Affiliation(s)
- Elizabeth V K Ledger
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, Armstrong Rd, London, SW7 2AZ, UK
| | - Stéphane Mesnage
- School of Biosciences, University of Sheffield, Sheffield, S10 2TN, UK
| | - Andrew M Edwards
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, Armstrong Rd, London, SW7 2AZ, UK.
| |
Collapse
|
41
|
Woroszyło M, Ciecholewska-Juśko D, Junka A, Drozd R, Wardach M, Migdał P, Szymczyk-Ziółkowska P, Styburski D, Fijałkowski K. Rotating Magnetic Field Increases β-Lactam Antibiotic Susceptibility of Methicillin-Resistant Staphylococcus aureus Strains. Int J Mol Sci 2021; 22:ijms222212397. [PMID: 34830278 PMCID: PMC8618647 DOI: 10.3390/ijms222212397] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 11/12/2021] [Accepted: 11/15/2021] [Indexed: 11/16/2022] Open
Abstract
Methicillin-resistant strains of Staphylococcus aureus (MRSA) have developed resistance to most β-lactam antibiotics and have become a global health issue. In this work, we analyzed the impact of a rotating magnetic field (RMF) of well-defined and strictly controlled characteristics coupled with β-lactam antibiotics against a total of 28 methicillin-resistant and sensitive S. aureus strains. The results indicate that the application of RMF combined with β-lactam antibiotics correlated with favorable changes in growth inhibition zones or in minimal inhibitory concentrations of the antibiotics compared to controls unexposed to RMF. Fluorescence microscopy indicated a drop in the relative number of cells with intact cell walls after exposure to RMF. These findings were additionally supported by the use of SEM and TEM microscopy, which revealed morphological alterations of RMF-exposed cells manifested by change of shape, drop in cell wall density and cytoplasm condensation. The obtained results indicate that the originally limited impact of β-lactam antibiotics in MRSA is boosted by the disturbances caused by RMF in the bacterial cell walls. Taking into account the high clinical need for new therapeutic options, effective against MRSA, the data presented in this study have high developmental potential and could serve as a basis for new treatment options for MRSA infections.
Collapse
Affiliation(s)
- Marta Woroszyło
- Department of Microbiology and Biotechnology, Faculty of Biotechnology and Animal Husbandry, West Pomeranian University of Technology in Szczecin, Piastów 45, 70-311 Szczecin, Poland; (M.W.); (D.C.-J.); (R.D.)
| | - Daria Ciecholewska-Juśko
- Department of Microbiology and Biotechnology, Faculty of Biotechnology and Animal Husbandry, West Pomeranian University of Technology in Szczecin, Piastów 45, 70-311 Szczecin, Poland; (M.W.); (D.C.-J.); (R.D.)
| | - Adam Junka
- Department of Pharmaceutical Microbiology and Parasitology, Faculty of Pharmacy, Medical University of Wroclaw, Borowska 211a, 50-534 Wrocław, Poland
- Laboratory of Microbiology, Łukasiewicz Research Network–PORT Polish Center for Technology Development, 54-066 Wrocław, Poland
- Correspondence: (A.J.); (K.F.); Tel.: +48-88-922-93-41 (A.J.); +48-91-449-6714 (K.F.)
| | - Radosław Drozd
- Department of Microbiology and Biotechnology, Faculty of Biotechnology and Animal Husbandry, West Pomeranian University of Technology in Szczecin, Piastów 45, 70-311 Szczecin, Poland; (M.W.); (D.C.-J.); (R.D.)
| | - Marcin Wardach
- Faculty of Electrical Engineering, West Pomeranian University of Technology in Szczecin, Sikorskiego 37, 70-313 Szczecin, Poland;
| | - Paweł Migdał
- Department of Environment, Hygiene and Animal Welfare, Faculty of Biology and Animal Science, Wroclaw University of Environmental and Life Sciences, Chełmońskiego 38C, 51-630 Wrocław, Poland;
| | - Patrycja Szymczyk-Ziółkowska
- Centre for Advanced Manufacturing Technologies (CAMT/FPC), Faculty of Mechanical Engineering, Wroclaw University of Science and Technology, Łukasiewicza 5, 50-371 Wrocław, Poland;
| | - Daniel Styburski
- Laboratory of Chromatography and Mass Spectroscopy, Faculty of Biotechnology and Animal Husbandry, West Pomeranian University of Technology in Szczecin, Klemensa Janickiego 29, 71-270 Szczecin, Poland;
| | - Karol Fijałkowski
- Department of Microbiology and Biotechnology, Faculty of Biotechnology and Animal Husbandry, West Pomeranian University of Technology in Szczecin, Piastów 45, 70-311 Szczecin, Poland; (M.W.); (D.C.-J.); (R.D.)
- Correspondence: (A.J.); (K.F.); Tel.: +48-88-922-93-41 (A.J.); +48-91-449-6714 (K.F.)
| |
Collapse
|
42
|
Douglas EJA, Duggan S, Brignoli T, Massey RC. The MpsB protein contributes to both the toxicity and immune evasion capacity of Staphylococcus aureus. MICROBIOLOGY-SGM 2021; 167. [PMID: 34618666 PMCID: PMC8698210 DOI: 10.1099/mic.0.001096] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Understanding the role specific bacterial factors play in the development of severe disease in humans is critical if new approaches to tackle such infections are to be developed. In this study we focus on genes we have found to be associated with patient outcome following bacteraemia caused by the major human pathogen Staphylococcus aureus. By examining the contribution these genes make to the ability of the bacteria to survive exposure to the antibacterial factors found in serum, we identify three novel serum resistance-associated genes, mdeA, mpsB and yycH. Detailed analysis of an MpsB mutant supports its previous association with the slow growing small colony variant (SCV) phenotype of S. aureus, and we demonstrate that the effect this mutation has on membrane potential prevents the activation of the Agr quorum sensing system, and as a consequence the mutant bacteria do not produce cytolytic toxins. Given the importance of both toxin production and immune evasion for the ability of S. aureus to cause disease, we believe that these findings explain the role of the mpsB gene as a mortality-associated locus during human disease.
Collapse
Affiliation(s)
- Edward J A Douglas
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
| | - Seána Duggan
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
| | - Tarcisio Brignoli
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
| | - Ruth C Massey
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK.,Schools of Microbiology and Medicine, University College Cork and APC Microbiome Ireland, Cork, Ireland
| |
Collapse
|