1
|
Jiang S, Matuszewska M, Chen M, Hong Y, Chen Y, Wang Z, Zhuang H, Sun L, Zhu F, Wang H, Wu X, Ji S, Holmes MA, Ba X, Chen Y, Yu Y. Emergence and spread of ST5 methicillin-resistant Staphylococcus aureus with accessory gene regulator dysfunction: genomic insights and antibiotic resistance. Microbiol Res 2025; 297:128196. [PMID: 40311457 DOI: 10.1016/j.micres.2025.128196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 04/23/2025] [Accepted: 04/24/2025] [Indexed: 05/03/2025]
Abstract
The globally disseminated Staphylococcus aureus ST5 clone poses a major public health threat due to its multidrug resistance and virulence. Here, we identified an agr-dysfunctional (agrA-I238K) ST5 MRSA clone that has spread across East and Southeast Asia, with recent increases in China since its emergence in the 1970s. Comparative genomic analyses identified distinct single-nucleotide polymorphisms and mobile genetic elements linked to enhanced resistance and virulence. This clone exhibits resistance to seven antimicrobial classes, including third-generation tetracyclines and fusidic acid, and shares phenotypic and genetic similarities with the vancomycin-intermediate S. aureus Mu50 strain, including reduced susceptibility to vancomycin, teicoplanin, and daptomycin. The agrA-I238K mutation attenuates hemolytic activity, increases biofilm formation, and reduces daptomycin susceptibility, suggesting a key role in the clone's success. Our results demonstrate the important role of agrA-I238K mutation in the widespread distribution of agr-dysfunctional MRSA and highlight the importance of genomic surveillance in tracking the spread of agr-dysfunctional ST5 MRSA.
Collapse
Affiliation(s)
- Shengnan Jiang
- Centre of Laboratory Medicine, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China; Zhejiang University School of Medicine, Hangzhou, Hangzhou, Zhejiang, China; Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China
| | - Marta Matuszewska
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK; Wellcome Sanger Institute, University of Cambridge, Cambridge, UK
| | - Mengzhen Chen
- Hangzhou Center for Disease Control and Prevention, Hangzhou, China
| | - Yueqin Hong
- Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China; Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yiyi Chen
- Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China; Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhengan Wang
- Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China; Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Hemu Zhuang
- Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China; Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Lu Sun
- Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China; Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Feiteng Zhu
- Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China; Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Haiping Wang
- Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China; Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xueqing Wu
- Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China; Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Shujuan Ji
- Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China; Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Mark A Holmes
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Xiaoliang Ba
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK.
| | - Yan Chen
- Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China; Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| | - Yunsong Yu
- Centre of Laboratory Medicine, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China; Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China; Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
2
|
Karaca B, Kyalo Kilonzo N, Korkmaz Ş, Onar O, Yıldırım Ö, Çöleri Cihan A. The Dual Role of the Medicinal Mushroom Fomitopsis pinicola in Inhibiting Biofilm and Reducing Antibiotic Resistance of Methicillin-Resistant Staphylococcus aureus. Food Sci Nutr 2025; 13:e70355. [PMID: 40438095 PMCID: PMC12116333 DOI: 10.1002/fsn3.70355] [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: 03/03/2025] [Revised: 04/26/2025] [Accepted: 05/16/2025] [Indexed: 06/01/2025] Open
Abstract
This study investigates the antimicrobial, antibiofilm, and anti-quorum sensing activity of Fomitopsis pinicola against methicillin-resistant Staphylococcus aureus (MRSA) strains and its potential to improve the efficacy of conventional antibiotics and exert selective cytotoxic effects on cancer cells. Ethanolic extracts of F. pinicola were analyzed for antibacterial activity by MIC and time-kill assays. Synergistic interactions with antibiotics were quantified using checkerboard assays. Antibiofilm activity was analyzed on polystyrene and stainless-steel surfaces. Anti-quorum sensing activity was determined by the inhibition of violacein in Chromobacterium violaceum. Efflux pump inhibition was assessed by the accumulation of the ethidium bromide. The down-regulation of virulence genes (agrA, hla) was measured by qRT-PCR (quantitative real-time reverse-transcription PCR). FT-IR (Fourier transform infrared spectroscopy) spectroscopy characterized the bioactive compounds, and the cytotoxicity assays on HT-29 colon cancer and Vero cells evaluated selective toxicity. The extract showed strong antibacterial effects with a MIC of 312.5 μg/mL and concentration-dependent bactericidal activity. Synergistic interactions with antibiotics led to FIC indices ≤ 0.5. The extract significantly inhibited biofilm formation and eradicated already formed biofilms. Sub-MIC concentrations reduced quorum sensing by 85.01%, inhibited efflux pump activity, and down-regulated virulence-associated genes. FT-IR analysis confirmed the presence of triterpenoids and terpenoids. The extract displayed selective cytotoxicity on HT-29 cancer cells, showing strong inhibition, while normal Vero cells were spared. These results emphasize the potential of F. pinicola as a robust candidate for antimicrobial therapeutics, especially against biofilm-associated and multidrug-resistant pathogens, as well as a selective anticancer agent.
Collapse
Affiliation(s)
- Başar Karaca
- Department of Biology, Faculty of ScienceAnkara UniversityAnkaraTurkey
| | | | - Şilan Korkmaz
- Department of Biology, Faculty of ScienceAnkara UniversityAnkaraTurkey
| | - Okan Onar
- Department of Biology, Faculty of ScienceAnkara UniversityAnkaraTurkey
| | - Özlem Yıldırım
- Department of Biology, Faculty of ScienceAnkara UniversityAnkaraTurkey
| | - Arzu Çöleri Cihan
- Department of Biology, Faculty of ScienceAnkara UniversityAnkaraTurkey
| |
Collapse
|
3
|
Girma A. Staphylococcus aureus: Current perspectives on molecular pathogenesis and virulence. Cell Surf 2025; 13:100137. [PMID: 39758277 PMCID: PMC11699754 DOI: 10.1016/j.tcsw.2024.100137] [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: 11/07/2024] [Revised: 12/04/2024] [Accepted: 12/07/2024] [Indexed: 01/07/2025] Open
Abstract
Staphylococcus aureus has evolved a sophisticated regulatory system to control its virulence. One of the main roles of this interconnected network is to sense and respond to diverse environmental signals by altering the synthesis of virulence components required for survival in the host, including cell surface adhesins, extracellular enzymes and toxins. The accessory gene regulator (agr), a quorum sensing system that detects the local concentration of a cyclic peptide signaling molecule, is one of the well-studied of these S. aureus regulatory mechanisms. By using this system, S. aureus is able to sense its own population density and translate this information into a specific pattern of gene expression. In addition to Agr, this pathogen senses specific stimuli through various two-component systems and synchronizes responses with alternative sigma factors and cytoplasmic regulators of the SarA protein family. These different regulatory mechanisms combine host and environmental information into a network that guarantees the best possible response of pathogens to changing circumstances. In this article, an overview of the most significant and thoroughly studied regulatory systems of S. aureus is provided, along with a summary of their roles in host interactions.
Collapse
Affiliation(s)
- Abayeneh Girma
- Department of Biology, College of Natural and Computational Science, Mekdela Amba University, P.O. Box 32, Tulu Awuliya, Ethiopia
| |
Collapse
|
4
|
Yan Q, Liu X, Liu Y, Cai K, Huang J, Zhang B, Wang Z, Han Z, Wen J, Li H. Decoy EPS layers for trapping and killing bacteria. Int J Biol Macromol 2025:144479. [PMID: 40412689 DOI: 10.1016/j.ijbiomac.2025.144479] [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: 10/16/2024] [Revised: 04/04/2025] [Accepted: 05/20/2025] [Indexed: 05/27/2025]
Abstract
Here we report a novel strategy using bacterial extracellular polymeric substances (EPS) as decoys to enhance bacterial adhesion and contact-based antimicrobial activity. EPS extracted from Staphylococcus aureus and Bacillus subtilis was used to coat wafers as a conditioning layer to alter surface properties and facilitate bacterial aggregation. Results show that EPS downregulates quorum sensing-related genes (agr and atl in Staphylococcus aureus by 80.5 % and 86.6 %, respectively; fliC in Escherichia coli by ~58.3 %), suggesting that EPS facilitates energy-efficient adhesion independent of quorum sensing signals. Loading antibiotics (erythromycin, linezolid, levofloxacin) into the EPS layer further enhances adhesion and contact killing. Especially, the surfaces loaded with a levofloxacin concentration of 2 μg/mL exhibit a significant antimicrobial effect. For Staphylococcus aureus, the antimicrobial rate reaches 83.66 % after 4 h incubation but drops to 39.9 % after 8 h incubation. In contract, Escherichia coli exhibits greater sensitivity, with antibacterial activity increasing to 92.97 % after 8 h incubation. Laser confocal microscopy characterization further reveals that the antibiotic-loaded EPS surfaces possess remarkable contact bacteria-killing activity. Our results show the promising recruiting-killing efficacy of the antibiotics-loaded EPS against bacteria, which would give insight into exploring new antibacterial strategies for enhanced contact-antibacterial performances.
Collapse
Affiliation(s)
- Qi Yan
- Zhejiang-Japan Joint Laboratory for Antibacterial and Antifouling Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China; Zhejiang Engineering Research Center for Biomedical Materials Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China; Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo 315300, China
| | - Xiaomei Liu
- Zhejiang-Japan Joint Laboratory for Antibacterial and Antifouling Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China; Zhejiang Engineering Research Center for Biomedical Materials Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China.
| | - Yi Liu
- Zhejiang-Japan Joint Laboratory for Antibacterial and Antifouling Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China; Zhejiang Engineering Research Center for Biomedical Materials Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China
| | - Kedan Cai
- The Department of Nephrology, Ningbo No.2Hospital, Ningbo 315000, China
| | - Jing Huang
- Zhejiang-Japan Joint Laboratory for Antibacterial and Antifouling Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China; Zhejiang Engineering Research Center for Biomedical Materials Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China
| | - Botao Zhang
- Zhejiang-Japan Joint Laboratory for Antibacterial and Antifouling Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China; Zhejiang Engineering Research Center for Biomedical Materials Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China
| | - Zhijuan Wang
- Zhejiang-Japan Joint Laboratory for Antibacterial and Antifouling Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China; Zhejiang Engineering Research Center for Biomedical Materials Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China
| | - Zhuoyue Han
- Zhejiang-Japan Joint Laboratory for Antibacterial and Antifouling Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China; Zhejiang Engineering Research Center for Biomedical Materials Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China
| | - Jianxin Wen
- Zhejiang-Japan Joint Laboratory for Antibacterial and Antifouling Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China; Zhejiang Engineering Research Center for Biomedical Materials Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China
| | - Hua Li
- Zhejiang-Japan Joint Laboratory for Antibacterial and Antifouling Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China; Zhejiang Engineering Research Center for Biomedical Materials Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China.
| |
Collapse
|
5
|
Bodine SP, Muir TW. Molecular Mechanisms of Virulence Regulation in Staphylococcus aureus: A Journey into Reconstitutive Biochemistry. Acc Chem Res 2025; 58:1657-1669. [PMID: 40331756 DOI: 10.1021/acs.accounts.5c00117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
ConspectusMethodological development in the fields of genetics, chemical biology, and biochemistry over the last several decades has provided researchers with a diverse set of powerful tools to investigate biological processes. Leveraging these innovations in concert, scientists can now characterize biological pathways at a level of complexity ranging from systems biology down to molecular and atomic detail.Throughout this Account, we illustrate how discoveries made using these tools build on each other to develop a comprehensive understanding of biological pathways. Advancements in genetic sequencing facilitates association of genotypes and phenotypes, independent of biochemical mechanism. Through the biochemical reconstitution of the interactions between biological macromolecules─including the small molecules (ligands and metabolites) and proteins─that participate in these biological pathways, scientists can characterize the specific molecular features that link genotype and phenotype. This facilitates identification of targets within these pathways that can be manipulated to achieve a greater understanding of the biological process or to develop interventions to improve human health outcomes.Specifically, we describe how this toolbox was leveraged to discover and characterize the molecular biochemistry underlying control of pathogenicity in the Gram-positive bacterium Staphylococcus aureus. Concurrent with advancements in the investigative tools available to the scientific community, we and others reported on the genetic, molecular, and biochemical/biophysical components of this regulatory system. Virulence control in S. aureus is achieved through a chemical system of bacterial cell-to-cell communication indexed to local population density, referred to as quorum sensing (QS). We and our collaborators identified that this QS system is encoded in the accessory gene regulator (agr) operon and functions via the biosynthesis, secretion, and accumulation of a short peptide signaling molecule─the autoinducing peptide (AIP)─in the local environment correlated with the growth of S. aureus in the same biological niche. Above a threshold concentration, these AIPs bind and activate a cell-surface receptor to stimulate an intracellular response resulting in altered gene expression and bacterial group behaviors. We discovered that chemical modification of these AIPs often generates molecules that exhibit potent inhibition of agr QS, with demonstrated therapeutic potential to treat S. aureus infections. We went on to characterize the biochemical mechanism of signaling molecule biosynthesis and receptor activation in controlled systems through in vitro reconstitution of the constituent enzymes and substrates. Biochemical reconstitution enabled quantitative assessment of biophysical parameters. These efforts culminated in the comprehensive characterization and functional in vitro reconstitution of agr QS in a synthetic system in a minimal model at the interface of genotype, mechanism, and phenotype.
Collapse
Affiliation(s)
- Steven P Bodine
- Department of Chemistry, Princeton University, Princeton, New Jersey 08544, United States
| | - Tom W Muir
- Department of Chemistry, Princeton University, Princeton, New Jersey 08544, United States
| |
Collapse
|
6
|
Tram NDT, Xu J, Chan KH, Rajamani L, Ee PLR. Bacterial clustering biomaterials as anti-infective therapies. Biomaterials 2025; 316:123017. [PMID: 39708775 DOI: 10.1016/j.biomaterials.2024.123017] [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: 08/13/2024] [Revised: 11/23/2024] [Accepted: 12/13/2024] [Indexed: 12/23/2024]
Abstract
In Nature, bacterial clustering by host-released peptides or nucleic acids is an evolutionarily conserved immune defense strategy employed to prevent adhesion of pathogenic microbes, which is prerequisite for most infections. Synthetic anti-adhesion strategies present as non-lethal means of targeting bacteria and may potentially be used to avoid resistance against antimicrobial therapies. From bacteria-agglutinating biomolecules discovered in nature to synthetic designs involving peptides, cationic polymers and nanoparticles, the modes of actions appear broad and unconsolidated. Herein, we present a critical review and update of the state-of-the-art in synthetic bacteria-clustering designs with proposition of a more streamlined nomenclature and classification. Overall, this review aims to consolidate the conceptual framework in the field of bacterial clustering and highlight its potentials as an avenue for discovering novel antibacterial biomaterials.
Collapse
Affiliation(s)
- Nhan Dai Thien Tram
- Department of Pharmacy and Pharmaceutical Sciences, National University of Singapore, 18 Science Drive 4, Singapore, 117559, Singapore
| | - Jian Xu
- Department of Pharmacy and Pharmaceutical Sciences, National University of Singapore, 18 Science Drive 4, Singapore, 117559, Singapore
| | - Kiat Hwa Chan
- Division of Science, Yale-NUS College, 16 College Avenue West, Singapore, 138527, Singapore; NUS College, National University of Singapore, 18 College Avenue East, Singapore, 138593, Singapore
| | - Lakshminarayanan Rajamani
- Department of Pharmacy and Pharmaceutical Sciences, National University of Singapore, 18 Science Drive 4, Singapore, 117559, Singapore; Ocular Infections and Anti-Microbials Research Group, Singapore Eye Research Institute, Singapore, 169856, Singapore; Ophthalmology and Visual Sciences Academic Clinical Program, Duke-NUS Graduate Medical School, Singapore, 169857, Singapore
| | - Pui Lai Rachel Ee
- Department of Pharmacy and Pharmaceutical Sciences, National University of Singapore, 18 Science Drive 4, Singapore, 117559, Singapore.
| |
Collapse
|
7
|
Aruwa CE, Sabiu S. Staphylococcus aureus AgrA Modulators From South African Antimicrobial Plants. Chem Biodivers 2025:e202403220. [PMID: 40229205 DOI: 10.1002/cbdv.202403220] [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: 12/06/2024] [Revised: 03/25/2025] [Accepted: 04/14/2025] [Indexed: 04/16/2025]
Abstract
South African plants are an underutilized metabolite source for novel antimicrobials targeting key quorum sensing (QS) systems like the Staphylococcus aureus AgrA. AgrA-modulating metabolites constitute a new approach to reducing resistance development. This study created a library of 1211 metabolites from 266 South African plants with antimicrobial action. Metabolites' anti-QS activities were assessed using advanced computational methods. Molecular docking showed top five metabolites (hinokiflavone, asphodelin, elliptinone, mamegakinone, and robustaflavone). Most leads conformed to the Lipinski's rule of 5. Dynamics simulation revealed closest ∆Gbind for robustaflavone (-25.50 kcal/mol) and hinokiflavone (-20.94 kcal/mol) compared to the standard (-26.59 kcal/mol). Leads interacted with key residues (Asn62/201, Asp75/214, Arg94/233, and Asn95/234) and a high number of interactions that ensured thermodynamic stability and compactness. The identified leads could be key mechanistic contributors to the antimicrobial activity of their inherent plants. Both the plants and metabolites (robustaflavone and hinokiflavone) could be further explored following structural modification as QS inhibitors.
Collapse
Affiliation(s)
- Christiana Eleojo Aruwa
- Department of Biotechnology and Food Science, Faculty of Applied Sciences, Durban University of Technology, Durban, KwaZulu-Natal, South Africa
| | - Saheed Sabiu
- Department of Biotechnology and Food Science, Faculty of Applied Sciences, Durban University of Technology, Durban, KwaZulu-Natal, South Africa
| |
Collapse
|
8
|
Beenken KE, Smeltzer MS. Staphylococcus aureus Biofilm-Associated Infections: Have We Found a Clinically Relevant Target? Microorganisms 2025; 13:852. [PMID: 40284688 PMCID: PMC12029350 DOI: 10.3390/microorganisms13040852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 03/26/2025] [Accepted: 04/02/2025] [Indexed: 04/29/2025] Open
Abstract
Staphylococcus aureus is one of the most diverse bacterial pathogens. This is reflected in its ability to cause a wide array of infections and in genotypic and phenotypic differences between clinical isolates that extend beyond their antibiotic resistance status. Many S. aureus infections, including those involving indwelling medical devices, are therapeutically defined by the formation of a biofilm. This is reflected in the number of reports focusing on S. aureus biofilm formation and biofilm-associated infections. These infections are characterized by a level of intrinsic resistance that compromises conventional antibiotic therapy irrespective of acquired resistance, suggesting that an inhibitor of biofilm formation would have tremendous clinical value. Many reports have described large-scale screens aimed at identifying compounds that limit S. aureus biofilm formation, but relatively few examined whether the limitation was sufficient to overcome this intrinsic resistance. Similarly, while many of these reports examined the impact of putative inhibitors on S. aureus phenotypes, very few took a focused approach to identify and optimize an effective inhibitor of specific biofilm-associated targets. Such approaches are dependent on validating a target, hopefully one that is not restricted by the diversity of S. aureus as a bacterial pathogen. Rigorous biological validation of such a target would allow investigators to virtually screen vast chemical libraries to identify potential inhibitors that warrant further investigation based on their predicted function. Here, we summarize reports describing S. aureus regulatory loci implicated in biofilm formation to assess whether they are viable targets for the development of an anti-biofilm therapeutic strategy with an emphasis on whether sarA has been sufficiently validated to warrant consideration in this important clinical context.
Collapse
Affiliation(s)
- Karen E. Beenken
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Mark S. Smeltzer
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
- Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|
9
|
Qian L, He Y, Lian W, Ji Z, Tian Z, Wang C, Cao C, Shern T, Stedman T, Sun Y. AgrC biotinylation inhibits Staphylococcus aureus infection. PLoS One 2025; 20:e0318695. [PMID: 40193824 PMCID: PMC11991674 DOI: 10.1371/journal.pone.0318695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 01/20/2025] [Indexed: 04/09/2025] Open
Abstract
Staphylococcus aureus (S. aureus) is a leading cause of nosocomial infections, particularly among antibiotic-resistant strains. S. aureus virulence is governed by the accessory gene regulator (Agr) quorum sensing (QS) system, which relies on AgrC, a two-component histidine kinase, to detect secreted auto-inducing peptides (AIPs). Emerging evidence highlights the potential of inhibiting the interaction between AgrC and AIPs as a promising therapeutic strategy. Given the limited clinic methods in inhibiting AgrC, we hereby report a novel method utilizing TurboID, an engineered biotin ligase, to inhibit Agr C on S. aureus via its biotinylation. To achieve this goal, a fusion protein named TurboID-AgrD[Formula: see text] (Agr-ID) was designed to include an AgrC binding domain (AgrID[Formula: see text]) and a catalytic domain (TurboID) for AgrC biotinylation. By incubating with Alexa Fluor 647-conjugated streptavidin, the biotinylated AgrC on S. aureus was successfully visualized through fluorescence microscopy with 100x objective. We further confirmed the specific biotinylation of AgrC using Western Blotting, and biotinylated AgrC resulted in inhibiting the growth of S. aureus strains, including S. aureus 25923, S. aureus 43300, and S. aureus 6538 (MRSA). The downstream biological effect of AgrC biotinylation exhibited decreased virulence protein generation as monitored by the lower presence of apoptotic HEK 293T cells after incubating with S. aureus cell lysates and supernatant. The impaired colonizing features from biotinylated S. aureus 6538 were investigated by calculating the decreased ratio of cell death versus live HeLa cells. By further investigating the efficiency of the immune clearance of biotinylated S. aureus by mouse macrophages, we observed the enhanced uptake of S. aureus by murine macrophages in vivo. Overall, our work reveals that the biotinylation of AgrC can inhibit the growth and toxicity of S. aureus while simultaneously promoting the clearance of biotinylated S. aureus via macrophage phagocytosis.
Collapse
Affiliation(s)
- Lijuan Qian
- College of Biomedicine and Health, Anhui Science and Technology University, Anhui, China
- College of Agriculture, Anhui Science and Technology University, Anhui, China
| | - Yuxin He
- BS-united China Group, International Genetically Engineered Machine (iGEM) Team, Anhui Science and Technology University, Anhui, China
| | - Wenzhe Lian
- BS-united China Group, International Genetically Engineered Machine (iGEM) Team, Anhui Science and Technology University, Anhui, China
| | - Zhiyuan Ji
- BS-united China Group, International Genetically Engineered Machine (iGEM) Team, Anhui Science and Technology University, Anhui, China
| | - Ziming Tian
- BS-united China Group, International Genetically Engineered Machine (iGEM) Team, Anhui Science and Technology University, Anhui, China
| | - Chuyun Wang
- BS-united China Group, International Genetically Engineered Machine (iGEM) Team, Anhui Science and Technology University, Anhui, China
| | - Chen Cao
- BS-united China Group, International Genetically Engineered Machine (iGEM) Team, Anhui Science and Technology University, Anhui, China
| | - Tyler Shern
- Columbia College, Columbia University, New York, United States of America
| | - Teagan Stedman
- Graduate School of Arts and Sciences, Columbia University Irving Medical Center, New York, United States of America
| | - Yujun Sun
- College of Biomedicine and Health, Anhui Science and Technology University, Anhui, China
- College of Agriculture, Anhui Science and Technology University, Anhui, China
| |
Collapse
|
10
|
Yu H, Wang L, Liu X, Zheng J, Xiang H, Zheng Y, Lv D, Yang J, Zhang Y, Qiu J, Wang D. Mechanistic insights into the multitarget synergistic efficacy of farrerol and β-lactam antibiotics in combating methicillin-resistant Staphylococcus aureus. Antimicrob Agents Chemother 2025; 69:e0155124. [PMID: 40019240 PMCID: PMC11963547 DOI: 10.1128/aac.01551-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 02/03/2025] [Indexed: 03/01/2025] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA), a principal causative agent of infections worldwide, urgently requires innovative interventions to counter its increasing risk. The present study revealed the profound impact of farrerol (FA), a robust bioactive agent, on the virulence and resistance mechanisms of MRSA. Our in-depth investigation revealed that FA significantly mitigated the β-lactam resistance of MRSA USA300, an achievement attributed to its precise interference with the BlaZ and Pbp2a protein. Additionally, FA indirectly diminishes the oligomerization of PBP2a by disrupting pigment synthesis, further contributing to its efficacy. In addition, FA extends its functional footprint beyond resistance modulation, exhibiting substantial antivirulence efficacy through selective inhibition of the accessory gene regulator (Agr) system, thereby significantly curbing MRSA pathogenicity in A549 cell and murine models. This study comprehensively explored the multiple impacts of FA on MRSA, shedding light on its versatile role as a BlaZ suppressor, pigment synthesis regulator, and AgrA activity modulator. These intricate findings firmly position FA as a compelling therapeutic candidate for addressing MRSA infections in the clinic.
Collapse
Affiliation(s)
- Hangqian Yu
- College of Animal Science, Jilin University, Changchun, China
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Li Wang
- Clinical Medical College, Changchun University of Chinese Medicine, Changchun, China
| | - Xin Liu
- College of Animal Science, Jilin University, Changchun, China
| | - Jianze Zheng
- College of Animal Science, Jilin University, Changchun, China
| | - Hua Xiang
- College of Animal Medicine, Jilin Agricultural University, Changchun, China
| | - Yanyang Zheng
- College of Animal Medicine, Jilin Agricultural University, Changchun, China
| | - Dongmei Lv
- College of Animal Science, Jilin University, Changchun, China
| | - Jingjing Yang
- College of Animal Science, Jilin University, Changchun, China
| | - Yuxin Zhang
- College of Animal Science, Jilin University, Changchun, China
| | - Jiazhang Qiu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Dacheng Wang
- College of Animal Science, Jilin University, Changchun, China
| |
Collapse
|
11
|
Liu D, Lu Y, Li Z, Pang X, Gao X. Quorum Sensing: Not Just a Bridge Between Bacteria. Microbiologyopen 2025; 14:e70016. [PMID: 40159675 PMCID: PMC11955508 DOI: 10.1002/mbo3.70016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 02/18/2025] [Accepted: 03/07/2025] [Indexed: 04/02/2025] Open
Abstract
The study of quorum sensing (QS) has gained critical importance, offering insights into bacterial and microorganism communication. QS, regulated by autoinducers, synchronizes collective bacterial behaviors across diverse chemical signals and target genes. This review highlights innovative approaches to regulating QS, emphasizing the potential of quorum quenching and QS inhibitors to mitigate bacterial pathogenicity. These strategies have shown promise in aquaculture and plant resistance, disrupting QS pathways to combat infections. QS also provides opportunities for developing biosensors for early disease detection and preventing biofilm formation, which is critical to overcoming antimicrobial resistance. The applications of QS extend to cancer therapy, with targeted drug delivery systems utilizing QS mechanisms. Advancements in QS regulation, such as the use of nanomaterials, hydrogels, and microplastics, provide novel methods to modulate QS systems. This review explores the latest developments in QS, recognizing its significance in controlling bacterial behavior and its broad impacts on human health and disease management. Integrating these insights into therapeutic strategies and diagnostics represents a pivotal opportunity for medical progress.
Collapse
Affiliation(s)
- Derun Liu
- Medical Science and Technology Innovation CenterShandong First Medical University & Shandong Academy of Medical SciencesjinanChina
| | - Yonglin Lu
- Medical Science and Technology Innovation CenterShandong First Medical University & Shandong Academy of Medical SciencesjinanChina
| | - Ziyun Li
- State Key Laboratory of Microbial TechnologyShandong UniversityQingdaoChina
| | - Xin Pang
- State Key Laboratory of Microbial TechnologyShandong UniversityQingdaoChina
| | - Xueyan Gao
- Medical Science and Technology Innovation CenterShandong First Medical University & Shandong Academy of Medical SciencesjinanChina
- State Key Laboratory of Microbial TechnologyShandong UniversityQingdaoChina
| |
Collapse
|
12
|
Wang W, Yi X, Zhou R, Peng W, Huang J, Chen J, Bo R, Liu M, Li J. Tea tree oil nanoemulsion targets AgrA protein potentiates amoxicillin efficacy against methicillin-resistant Staphylococcus aureus. Int J Biol Macromol 2025; 292:139111. [PMID: 39733883 DOI: 10.1016/j.ijbiomac.2024.139111] [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: 10/21/2024] [Revised: 12/06/2024] [Accepted: 12/21/2024] [Indexed: 12/31/2024]
Abstract
The excessive utilization of antibiotics gives rise to the development of bacterial resistance, the deterioration of animal immune functions, the increase in mortality rates, and the undermining of human immunity. Therefore, there is an urgent necessity to explore new antimicrobial agents or alternatives to tackle bacterial resistance. We investigated tea tree oil (TTO), a pure natural plant essential oil extracted from Melaleuca leaves, which exerted efficient antibacterial activities. However, the poor solubility and high volatility of TTO limited the clinical application. Therefore, tea tree oil and Tween 80 were formulated into a stable nanoemulsion (Nano TTO). We attested that Nano TTO, as an antibiotic adjuvant, enhanced the antibacterial activity of amoxicillin (AMX) against methicillin-resistant Staphylococcus aureus (MRSA) and inhibited the formation of biofilms. Mechanistic studies proved that the Nano TTO potentiation effect on AMX was primarily the result of inhibition of the Agr expression by targeting the accessory regulator AgrA. Furthermore, Nano TTO effectively boosts the efficacy of amoxicillin in the mouse septicaemia model and mouse skin wound infection model. Overall, these results revealed the potential of Nano TTO as an adjuvant to evade multidrug-resistant bacterial pathogens and improve treatment outcomes for drug-resistant infections.
Collapse
Affiliation(s)
- Weimei Wang
- School of Veterinary Medicine, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, PR China
| | - Xiaobin Yi
- School of Veterinary Medicine, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, PR China
| | - Ruigang Zhou
- School of Veterinary Medicine, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, PR China
| | - Weilong Peng
- School of Veterinary Medicine, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, PR China
| | - Junjie Huang
- School of Veterinary Medicine, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, PR China
| | - Jun Chen
- School of Veterinary Medicine, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, PR China
| | - Ruonan Bo
- School of Veterinary Medicine, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu 225009, PR China
| | - Mingjiang Liu
- School of Veterinary Medicine, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu 225009, PR China
| | - Jingui Li
- School of Veterinary Medicine, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu 225009, PR China.
| |
Collapse
|
13
|
Chigozie VU, Saki M, Esimone CO. Molecular structural arrangement in quorum sensing and bacterial metabolic production. World J Microbiol Biotechnol 2025; 41:71. [PMID: 39939401 DOI: 10.1007/s11274-025-04280-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Accepted: 01/28/2025] [Indexed: 02/14/2025]
Abstract
Quorum sensing (QS) regulates bacterial behaviors such as biofilm formation, virulence, and metabolite production through signaling molecules like acyl-homoserine lactones (AHLs), peptides, and AI-2. These signals are pivotal in bacterial communication, influencing pathogenicity and industrial applications. This review explores the molecular architecture of QS signals and their role in metabolite production, emphasizing structural modifications that disrupt bacterial communication to control virulence and enhance industrial processes. Key findings highlight the development of synthetic QS analogs, engineered inhibitors, and microbial consortia as innovative tools in biotechnology and medicine. The review underscores the potential of molecular engineering in managing microbial behaviors and optimizing applications like biofuel production, bioplastics, and anti-virulence therapies. Additionally, cross-species signaling mechanisms, particularly involving AI-2, reveal new opportunities for regulating interspecies cooperation and competition. This synthesis aims to bridge molecular insights with practical applications, showcasing how QS-based technologies can drive advancements in microbial biotechnology and therapeutic strategies.
Collapse
Affiliation(s)
- Victor U Chigozie
- Department of Pharmaceutical Microbiology and Biotechnology, David Umahi Federal University of Health Sciences, Ohaozara, Ebonyi State, Nigeria.
- International Institute for Pharmaceutical Research (IIPR), Ohaozara, Ebonyi State, Nigeria.
| | - Morteza Saki
- Department of Microbiology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Charles O Esimone
- Department of Pharmaceutical Microbiology and Biotechnology, Nnamdi Azikiwe University, Awka, Nigeria
| |
Collapse
|
14
|
Marchi J, Minh CNN, Debarbieux L, Weitz JS. Multi-strain phage induced clearance of bacterial infections. PLoS Comput Biol 2025; 21:e1012793. [PMID: 39903766 PMCID: PMC11828373 DOI: 10.1371/journal.pcbi.1012793] [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: 09/30/2024] [Revised: 02/14/2025] [Accepted: 01/14/2025] [Indexed: 02/06/2025] Open
Abstract
Bacteriophage (or 'phage' - viruses that infect and kill bacteria) are increasingly considered as a therapeutic alternative to treat antibiotic-resistant bacterial infections. However, bacteria can evolve resistance to phage, presenting a significant challenge to the near- and long-term success of phage therapeutics. Application of mixtures of multiple phages (i.e., 'cocktails') has been proposed to limit the emergence of phage-resistant bacterial mutants that could lead to therapeutic failure. Here, we combine theory and computational models of in vivo phage therapy to study the efficacy of a phage cocktail, composed of two complementary phages motivated by the example of Pseudomonas aeruginosa facing two phages that exploit different surface receptors, LUZ19v and PAK_P1. As confirmed in a Luria-Delbrück fluctuation test, this motivating example serves as a model for instances where bacteria are extremely unlikely to develop simultaneous resistance mutations against both phages. We then quantify therapeutic outcomes given single- or double-phage treatment models, as a function of phage traits and host immune strength. Building upon prior work showing monophage therapy efficacy in immunocompetent hosts, here we show that phage cocktails comprised of phage targeting independent bacterial receptors can improve treatment outcome in immunocompromised hosts and reduce the chance that pathogens simultaneously evolve resistance against phage combinations. The finding of phage cocktail efficacy is qualitatively robust to differences in virus-bacteria interactions and host immune dynamics. Altogether, the combined use of theory and computational analysis highlights the influence of viral life history traits and receptor complementarity when designing and deploying phage cocktails in immunocompetent and immunocompromised hosts.
Collapse
Affiliation(s)
- Jacopo Marchi
- Department of Biology, University of Maryland, College Park, Maryland, United States of America
| | - Chau Nguyen Ngoc Minh
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Bacteriophage Bacterium Host, Paris, France
- Sorbonne Université, Collége Doctoral, Paris, France
| | - Laurent Debarbieux
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Bacteriophage Bacterium Host, Paris, France
| | - Joshua S Weitz
- Department of Biology, University of Maryland, College Park, Maryland, United States of America
- Department of Physics, University of Maryland, College Park, Maryland, United States of America
- University of Maryland Institute for Health Computing, North Bethesda, Maryland, United States of America
| |
Collapse
|
15
|
Beenken KE, Campbell MJ, Smeltzer MS. The ability of sarA to limit protease production plays a key role in the pathogenesis of Staphylococcus aureus osteomyelitis irrespective of the functional status of agr. Infect Immun 2025; 93:e0047324. [PMID: 39611695 PMCID: PMC11784413 DOI: 10.1128/iai.00473-24] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 11/04/2024] [Indexed: 11/30/2024] Open
Abstract
We demonstrate that mutation of the staphylococcal accessory regulator A (sarA) in the USA300 strain LAC limits virulence in a murine osteomyelitis model to a greater extent than mutation of the accessory gene regulator (agr) and that it does so irrespective of the functional status of agr. Protease production was decreased in the agr mutant but increased in sarA and sarA/agr mutants to a degree that limited biofilm formation. Extracellular protein A (eSpa) and full-length extracellular nuclease (Nuc1) were absent in the conditioned medium (CM) from sarA and sarA/agr mutants, and their abundance was restored in both mutants by eliminating protease production. Cytotoxicity of CM for osteoblasts and osteoclasts was also reduced in both mutants. Cytotoxicity was restored in a protease-deficient sarA mutant but not in the protease-deficient sarA/agr mutant. Reduced cytotoxicity was correlated with the reduced abundance of full-length α-toxin, LukF, and LukS in sarA and sarA/agr mutants. The abundance of these toxins in their full-length form was increased in the protease-deficient sarA mutant by comparison to LAC, demonstrating that mutation of sarA increases the production of these toxins but increased protease production limits their abundance in full-length and presumably functional forms. Most importantly, eliminating protease production enhanced the virulence of sarA and sarA/agr mutants, but had no impact in the agr mutant. We conclude that a key factor in the attenuation of LAC sarA and sarA/agr mutants in osteomyelitis is the increased production of extracellular proteases and its impact on virulence factors that contribute to biofilm formation and cytotoxicity.IMPORTANCEThe persistent emergence of antibiotic-resistant strains has rekindled interest in anti-virulence strategies to combat S. aureus infections. Numerous reports describe anti-virulence strategies focusing on key regulatory elements that globally influence virulence factor production, the two most commonly targeted being the accessory gene regulator (agr) and the staphylococcal accessory regulator A (sarA). We demonstrate that mutation of sarA limits virulence to a greater extent than mutation of agr and that this can be attributed to increased protease production in both sarA and sarA/agr mutants. This illustrates the critical role of sarA in protease-mediated post-translational regulation in S. aureus. It also suggests that an inhibitor of sarA would be more effective than an inhibitor of agr in overcoming the therapeutic recalcitrance of osteomyelitis and that such an inhibitor would remain effective even in the context of agr mutants known to arise in vivo during the transition from acute to chronic infection.
Collapse
Affiliation(s)
- Karen E. Beenken
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Mara J. Campbell
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Mark S. Smeltzer
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| |
Collapse
|
16
|
Xia Y, Hu Z, Jin Q, Chen Q, Zhao C, Qiang R, Xie Z, Li L, Zhang H. Structural characteristics, functions, and counteracting strategies of biofilms in Staphylococcus aureus. Comput Struct Biotechnol J 2025; 27:488-500. [PMID: 39916696 PMCID: PMC11799891 DOI: 10.1016/j.csbj.2025.01.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 02/09/2025] Open
Abstract
Background Staphylococcus aureus (S. aureus) is a prevalent pathogen associated with a wide range of infections, exhibiting significant antibiotic resistance and posing therapeutic challenges in clinical settings. The formation of biofilms contributes to the emergence of resistant strains, further diminishing the efficacy of antibiotics. This, in turn, leads to chronic and recurrent infections, ultimately increasing the healthcare burden. Consequently, preventing and eliminating biofilms has become a critical focus in clinical management and research. Aim of review This review systematically examines the mechanisms underlying biofilm formation in S. aureus and its contribution to antibiotic resistance, emphasizing the essential roles biofilms play in maintaining structural integrity and enhancing resistance. It also analyses the protective mechanisms that fortify S. aureus biofilms against antimicrobial treatments. Furthermore, the review provides a comprehensive overview of recent therapeutic innovations, including enzymatic therapy, nanotechnology, gene editing, and phage therapy. Key scientific concepts of review Emerging therapeutic strategies present novel approaches to combat S. aureus biofilm-associated infections through various mechanisms. This review discusses recent advancements in these therapies, their practical challenges in clinical application, and provides an in-depth analysis of each strategy's mechanisms and therapeutic potential. By mapping future research directions, this review aims to refine anti-biofilm strategies to control infection progression and effectively mitigate recurrence.
Collapse
Affiliation(s)
- Yanze Xia
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhenghui Hu
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Qiyuan Jin
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Qi Chen
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Chenhao Zhao
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Rui Qiang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Zonggang Xie
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Liubing Li
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Haifang Zhang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- MOE Key Laboratory of Geriatric Diseases and Immunology, Soochow University, Suzhou, China
| |
Collapse
|
17
|
Pengfei S, Yifan Y, Linhui L, Yimin L, Dan X, Shaowei G, Guanqing H, Yong W. Novel antibiotics against Staphylococcus aureus without detectable resistance by targeting proton motive force and FtsH. MedComm (Beijing) 2025; 6:e70046. [PMID: 39781293 PMCID: PMC11707430 DOI: 10.1002/mco2.70046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 11/06/2024] [Accepted: 11/21/2024] [Indexed: 01/12/2025] Open
Abstract
The increased prevalence of methicillin-resistant Staphylococcus aureus (MRSA) and its biofilms poses a great threat to human health. Especially, S. aureus-related osteomyelitis was hardly cured even by conventional antibiotics combined with surgical treatment. The development of novel structural antibiotics is urgently needed. By high-throughput screening and rational design, we identified a small molecule C218-0546 and its optimized analog STK848198 with great antimicrobial potential against MRSA avoiding resistance occurrence. And significant synergistical antimicrobial effects were found between the molecules and conventional antibiotics. Mechanisms studies by transcriptomics, fluorescent probes, molecule dynamics, and plasma surface resonance indicated that the proton motive force as well as FtsH are the main potential targets of these molecules. The compounds exhibited excellent in vivo pharmacokinetics, toxicity profiles, and antimicrobial activities in the abscess model as well as the peritonitis-sepsis model. In addition, STK848198 was found to be effective against MRSA biofilms by interacting with the quorum sensing system. STK848198 also showed in vivo efficacy in the periprosthetic joint infection model. In all, our study identified a class of antimicrobials with novel scaffolds that could be potential alternatives for the treatment of MRSA and its biofilm-related infections.
Collapse
Affiliation(s)
- She Pengfei
- Department of Laboratory MedicineThe Third Xiangya Hospital of Central South UniversityChangshaHunanChina
| | - Yang Yifan
- Department of Laboratory MedicineThe Third Xiangya Hospital of Central South UniversityChangshaHunanChina
| | - Li Linhui
- Department of Laboratory MedicineThe Third Xiangya Hospital of Central South UniversityChangshaHunanChina
| | - Li Yimin
- Department of Laboratory MedicineThe Affiliated Changsha Hospital of Xiangya School of Medicine (The First Hospital of Changsha)Central South UniversityChangshaHunanChina
| | - Xiao Dan
- Department of Laboratory MedicineThe Affiliated Changsha Hospital of Xiangya School of Medicine (The First Hospital of Changsha)Central South UniversityChangshaHunanChina
| | - Guo Shaowei
- Department of Laboratory MedicineThe Affiliated Changsha Hospital of Xiangya School of Medicine (The First Hospital of Changsha)Central South UniversityChangshaHunanChina
| | - Huang Guanqing
- Department of Laboratory MedicineThe Affiliated Changsha Hospital of Xiangya School of Medicine (The First Hospital of Changsha)Central South UniversityChangshaHunanChina
| | - Wu Yong
- Department of Laboratory MedicineThe Affiliated Changsha Hospital of Xiangya School of Medicine (The First Hospital of Changsha)Central South UniversityChangshaHunanChina
| |
Collapse
|
18
|
Ingmer H, Leisner JJ, Fulaz S. Forssman and the staphylococcal hemolysins. APMIS 2025; 133:e13459. [PMID: 39188243 PMCID: PMC11669744 DOI: 10.1111/apm.13459] [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: 06/12/2024] [Accepted: 08/05/2024] [Indexed: 08/28/2024]
Abstract
Forssman was a Swedish pathologist and microbiologist who, in the 1920s and 1930s conducted a long series of experiments that led to unique insights into surface antigens of blood cells, as well as added to the discrimination of toxins produced by staphylococci that lyse red blood cells. This review takes offset in the studies published by Forssman in APMIS addressing the hemolytic properties of staphylococcal toxins displayed against erythrocytes of animal and human origin. In light of current knowledge, we will discuss the insights we now have and how they may pave the way for curing infections with pathogenic staphylococci, including Staphylococcus aureus.
Collapse
Affiliation(s)
- Hanne Ingmer
- Department of Veterinary and Animal ScienceUniversity of CopenhagenCopenhagenDenmark
| | - Jørgen J. Leisner
- Department of Veterinary and Animal ScienceUniversity of CopenhagenCopenhagenDenmark
| | - Stephanie Fulaz
- Department of Veterinary and Animal ScienceUniversity of CopenhagenCopenhagenDenmark
| |
Collapse
|
19
|
Meng Q, Wang X, Huang X, Li C, Yu Z, Li P, Liu X, Wen Z. Repurposing Benzbromarone as an Antibacterial Agent against Gram-Positive Bacteria. ACS Infect Dis 2024; 10:4208-4221. [PMID: 39561096 DOI: 10.1021/acsinfecdis.4c00495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2024]
Abstract
The rise of antibiotic-resistant Gram-positive pathogens, particularly methicillin-resistant Staphylococcus aureus (MRSA), presents a significant challenge in clinical settings. There is a critical need for new antibacterial agents to combat these resistant strains. Our study reveals that the uricosuric drug Benzbromarone (Benz) exhibits potent antibacterial activity against Gram-positive pathogens, with minimum inhibitory concentrations (MICs) ranging from 8 to 32 μg/mL and minimum bactericidal concentrations (MBCs) ranging from 32 to 128 μg/mL against clinical isolates of S. aureus, S. epidermidis, Enterococcus faecalis, and Streptococcus agalactiae. Furthermore, Benz significantly inhibits biofilm formation at subinhibitory concentrations and eradicates mature biofilms at higher concentrations. Benz also suppresses the hemolytic activity of S. aureus, indicating its potential to reduce virulence. Proteomic and in vitro induced resistance analyses indicate that Benz inhibits protein synthesis and turnover. Additionally, Benz induces membrane depolarization and increases membrane permeability, likely by targeting the membrane phospholipid phosphatidylethanolamine (PE). In the mouse wound infection model, Benz promotes wound healing and significantly reduces bacterial load. These findings suggest that Benz is a promising candidate for developing new antibacterial therapies against Gram-positive bacterial infections.
Collapse
Affiliation(s)
- Qingyin Meng
- Department of Infectious Diseases and Shenzhen Key Laboratory for Endogenous Infections, Shenzhen Nanshan People's Hospital, Shenzhen University Medical School, No. 89 Taoyuan Road, Nanshan District, Shenzhen 518052, China
| | - Xueting Wang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Nation Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518060, China
- Guangdong Key Laboratory for New Technology Research of Vegetables, Vegetable Research Institute, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
| | - Xuancheng Huang
- Department of Infectious Diseases and Shenzhen Key Laboratory for Endogenous Infections, Shenzhen Nanshan People's Hospital, Shenzhen University Medical School, No. 89 Taoyuan Road, Nanshan District, Shenzhen 518052, China
| | - Congcong Li
- Department of Infectious Diseases and Shenzhen Key Laboratory for Endogenous Infections, Shenzhen Nanshan People's Hospital, Shenzhen University Medical School, No. 89 Taoyuan Road, Nanshan District, Shenzhen 518052, China
| | - Zhijian Yu
- Department of Infectious Diseases and Shenzhen Key Laboratory for Endogenous Infections, Shenzhen Nanshan People's Hospital, Shenzhen University Medical School, No. 89 Taoyuan Road, Nanshan District, Shenzhen 518052, China
| | - Peiyu Li
- Department of Infectious Diseases and Shenzhen Key Laboratory for Endogenous Infections, Shenzhen Nanshan People's Hospital, Shenzhen University Medical School, No. 89 Taoyuan Road, Nanshan District, Shenzhen 518052, China
| | - Xiaoju Liu
- Department of Infectious Diseases and Shenzhen Key Laboratory for Endogenous Infections, Shenzhen Nanshan People's Hospital, Shenzhen University Medical School, No. 89 Taoyuan Road, Nanshan District, Shenzhen 518052, China
| | - Zewen Wen
- Department of Infectious Diseases and Shenzhen Key Laboratory for Endogenous Infections, Shenzhen Nanshan People's Hospital, Shenzhen University Medical School, No. 89 Taoyuan Road, Nanshan District, Shenzhen 518052, China
| |
Collapse
|
20
|
De Jesus R, Iqbal S, Mundra S, AlKendi R. Heterogenous bioluminescence patterns, cell viability, and biofilm formation of Photobacterium leiognathi strains exposed to ground microplastics. FRONTIERS IN TOXICOLOGY 2024; 6:1479549. [PMID: 39665083 PMCID: PMC11631867 DOI: 10.3389/ftox.2024.1479549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 10/30/2024] [Indexed: 12/13/2024] Open
Abstract
Microplastics (MPs) have been detected in various aquatic environments and negatively affect organisms, including marine luminous bacteria. This study investigated the differences in bioluminescence patterns, cell viability, and biofilm formation of Photobacterium leiognathi strains (LB01 and LB09) when exposed to various concentrations of ground microplastics (GMPs; 0.25%, 0.50%, 1%, or 2% [w/v] per mL) at 22°C or 30°C for 3.1 days (75 h) and 7 days. The strains exhibited heterogenous responses, including variable bioluminescence patterns, cell viability, and biofilm formation, due to the GMPs having effects such as hormesis and bioluminescence quenching. Moreover, the bioluminescence and cell viability differed between the two strains, possibly involving distinct cellular mechanisms, suggesting that GMPs affect factors that influence quorum sensing. Furthermore, the biofilm formation of LB01 and LB09 was observed following exposure to GMPs. Both strains showed increased biofilm formation at higher GMP concentrations (1% and 2%) after 3.1 days at 30°C and 22°C. However, in the 7-day experiment, LB01 significantly (p < 0.05) increased biofilms at 22°C, while LB09 significantly (p < 0.05) produced biofilms at 30°C. These findings highlight the strain-specific responses of Phb. leiognathi to MP pollutants. Therefore, this study underscores the importance of evaluating MPs as environmental stressors on marine microorganisms and their role in the ecophysiological repercussions of plastic pollution in aquatic environments.
Collapse
Affiliation(s)
- Rener De Jesus
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Sameera Iqbal
- Khalifa Center for Genetic Engineering and Biotechnology, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Sunil Mundra
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
- Khalifa Center for Genetic Engineering and Biotechnology, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Ruwaya AlKendi
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
21
|
Arévalo-Jaimes BV, Torrents E. Candida albicans enhances Staphylococcus aureus virulence by progressive generation of new phenotypes. CURRENT RESEARCH IN MICROBIAL SCIENCES 2024; 7:100316. [PMID: 39649408 PMCID: PMC11621768 DOI: 10.1016/j.crmicr.2024.100316] [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] [Indexed: 12/10/2024] Open
Abstract
Candida albicans and Staphylococcus aureus have been co-isolated from several biofilm-associated diseases, including those related to medical devices. This association confers advantages to both microorganisms, resulting in detrimental effects on the host. To elucidate this phenomenon, the present study investigated colony changes derived from non-physical interactions between C. albicans and S. aureus. We performed proximity assays by confronting colonies of the yeast and the bacteria on agar plates at six different distances for 9-10 days. We found that colony variants of S. aureus originated progressively after prolonged exposure to C. albicans proximity, specifically in response to pH neutralization of the media by the fungi. The new phenotypes of S. aureus were more virulent in a Galleria mellonella larvae model compared to colonies grown without C. albicans influence. This event was associated with an upregulation of RNA III and agrA expression, suggesting a role for α-toxin. Our findings indicate that C. albicans enhances S. aureus virulence by inducing the formation of more aggressive colonies. This highlights the importance of understanding the intricate connection between environmental responses, virulence and, fitness in S. aureus pathogenesis.
Collapse
Affiliation(s)
- Betsy Verónica Arévalo-Jaimes
- Bacterial infections and antimicrobial therapies group, Institute for Bioengineering of Catalonia (IBEC), Baldiri Reixac Street 10, 08037, Barcelona, Spain
- Microbiology Section, Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Diagonal Street 647, 08028, Barcelona, Spain
| | - Eduard Torrents
- Bacterial infections and antimicrobial therapies group, Institute for Bioengineering of Catalonia (IBEC), Baldiri Reixac Street 10, 08037, Barcelona, Spain
- Microbiology Section, Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Diagonal Street 647, 08028, Barcelona, Spain
| |
Collapse
|
22
|
Faleye OO, Lee JH, Kim YG, Faleye OS, Lee J. Antibiofilm and antivirulence potentials of iodinated fmoc-phenylalanine against Staphylococcus aureus. Microb Pathog 2024; 197:107080. [PMID: 39454802 DOI: 10.1016/j.micpath.2024.107080] [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: 05/08/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 10/28/2024]
Abstract
Staphylococcus aureus poses significant risks to public health due to its ability to form biofilm and produce virulence factors, contributing to the increase in antibiotic resistance and treatment complications. This emphasizes the urgent need for novel antimicrobial controls. Based on the premise that halogenation improves antimicrobial efficacy, this study investigated the ability of halogenated phenylalanine to effectively inhibit S. aureus biofilm formation and virulence activities. Among 29 halogenated compounds, Fmoc-4-iodo-phenylalanine (Fmoc-Iodo-Phe) displayed the highest antibiofilm effect against S. aureus, achieving 94.3 % reduction at 50 μg/mL. Microscopic studies confirmed its ability to prevent and disrupt mature biofilms. At 10 μg/mL, Fmoc-Iodo-Phe markedly inhibited virulence factors, such as cell surface hydrophobicity, hemolysin and slime production. It showed low propensity for resistance development and effectively inhibited biofilms formed by methicillin-resistant S. aureus (MRSA) and S. epidermidis, but was inactive against Gram-negative bacteria. Gene expression analysis complemented by molecular docking suggest that Fmoc-Iodo-Phe could target the AgrA quorum sensing cascade due to strong interactions with key residues at its DNA binding sites. Notably, it was non-cytotoxic in Caenorhabditis elegans model and satisfied drug-likeliness criteria based on ADMET prediction. Therefore, our findings position Fmoc-Iodo-Phe as a promising antimicrobial candidate against S. aureus infections, underscoring its potential as an alternative to traditional antibiotics.
Collapse
Affiliation(s)
| | - Jin-Hyung Lee
- School of Chemical Engineering, Yeungnam University, 280 Daehak-Ro, Gyeongsan, 38541, Republic of Korea
| | - Yong-Guy Kim
- School of Chemical Engineering, Yeungnam University, 280 Daehak-Ro, Gyeongsan, 38541, Republic of Korea
| | - Olajide Sunday Faleye
- School of Chemical Engineering, Yeungnam University, 280 Daehak-Ro, Gyeongsan, 38541, Republic of Korea
| | - Jintae Lee
- School of Chemical Engineering, Yeungnam University, 280 Daehak-Ro, Gyeongsan, 38541, Republic of Korea.
| |
Collapse
|
23
|
Cui S, Kim E. Quorum sensing and antibiotic resistance in polymicrobial infections. Commun Integr Biol 2024; 17:2415598. [PMID: 39430726 PMCID: PMC11487952 DOI: 10.1080/19420889.2024.2415598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 09/20/2024] [Accepted: 10/07/2024] [Indexed: 10/22/2024] Open
Abstract
Quorum sensing (QS) is a critical bacterial communication system regulating behaviors like biofilm formation, virulence, and antibiotic resistance. This review highlights QS's role in polymicrobial infections, where bacterial species interactions enhance antibiotic resistance. We examine QS mechanisms, such as acyl-homoserine lactones (AHLs) in Gram-negative bacteria and autoinducing peptides (AIPs) in Gram-positive bacteria, and their impact on biofilm-associated antibiotic resistance. The challenges uniquely associated with polymicrobial infections, such as those found in cystic fibrosis lung infections, chronic wound infections, and medical device infections, are also summarized. Furthermore, we explore various laboratory models, including flow cells and dual-species culture models, used to study QS interactions in polymicrobial environments. The review also discusses promising quorum sensing inhibitors (QSIs), such as furanones and AHL analogs, which have demonstrated efficacy in reducing biofilm formation and virulence in laboratory and clinical studies. By addressing the interplay between QS and antibiotic resistance, this paper aims to advance therapeutic strategies that disrupt bacterial communication and improve antibiotic efficacy, ultimately mitigating the global challenge of antibiotic resistance in polymicrobial infections.
Collapse
Affiliation(s)
- Sunny Cui
- Department of Biological Sciences, Dartmouth College, Hanover, NH, USA
| | - Esther Kim
- Arts and Science, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
24
|
Saha S, Ghosh M. Computational exploration of natural compounds targeting Staphylococcus aureus: inhibiting AgrA promoter binding for antimicrobial intervention. J Biomol Struct Dyn 2024; 42:8256-8267. [PMID: 37578046 DOI: 10.1080/07391102.2023.2246566] [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: 06/12/2023] [Accepted: 07/30/2023] [Indexed: 08/15/2023]
Abstract
Staphylococcus aureus is a highly virulent nosocomial pathogen that poses a significant threat to individuals exposed to healthcare settings. Due to its sophisticated machinery for producing virulence factors, S. aureus can cause severe and potentially fatal infections in humans. This study focuses on the response regulator AgrA, which plays a crucial role in regulating the production of virulence factors in S. aureus. The objective is to identify natural compounds that can inhibit the binding of AgrA to its promoter site, thus inhibiting the expression of virulence genes. To achieve this, a pharmacophore model was generated using known drugs and applied to screen the ZINC natural product database. The resulting compounds were subjected to molecular docking-based virtual screening against the C-terminal DNA binding domain of AgrA. Three compounds, namely ZINC000077269178, ZINC000051012304, and ZINC000004266026, were shortlisted based on their strong affinity for key residues involved in DNA binding and transcription initiation. Subsequently, the unbound and ligand-bound complexes were subjected to a 200 ns molecular dynamics simulation to assess their conformational stability. Various analyses, including RMSD, RMSF, Rg, SASA, Principal Component Analysis, and Gibbs free energy landscape, were conducted on the simulation trajectory. The RMSD profile indicated similar fluctuations in both bound and unbound structures, while the Rg profile demonstrated the compactness of the protein without any unfolding during the simulation. Furthermore, Principal component analysis revealed that ligand binding reduced the overall atomic motion of the protein whereas free energy landscape suggested the energy variations obtained in complexes.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Subhadip Saha
- Department of Biotechnology, National Institute of Technology Durgapur, Durgapur, India
| | - Monidipa Ghosh
- Department of Biotechnology, National Institute of Technology Durgapur, Durgapur, India
| |
Collapse
|
25
|
Manu P, Nketia PB, Osei-Poku P, Kwarteng A. Computational Mutagenesis and Inhibition of Staphylococcus aureus AgrA LytTR Domain Using Phenazine Scaffolds: Insight From a Biophysical Study. BIOMED RESEARCH INTERNATIONAL 2024; 2024:8843954. [PMID: 39328594 PMCID: PMC11424843 DOI: 10.1155/2024/8843954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 08/29/2024] [Indexed: 09/28/2024]
Abstract
Biofilm formation by Staphylococcus aureus is a major challenge in clinical settings due to its role in persistent infections. The AgrA protein, a key regulator in biofilm development, is a promising target for therapeutic intervention. This study investigates the antibiofilm potential of halogenated phenazine compounds by targeting AgrA and explores their molecular interactions to provide insights for drug development. We employed molecular docking, molecular dynamics simulations, and computational mutagenesis to evaluate the binding of halogenated phenazine compounds (C1 to C7, HP, and HP-14) to AgrA. Binding free energy analysis was performed to assess the affinity of these compounds for the AgrA-DNA complex. Additionally, the impact of these compounds on AgrA's structural conformation and salt bridge interactions was examined. The binding-free energy analysis revealed that all compounds enhance binding affinity compared to the Apo form of AgrA, which has a ΔGbind of -80.75 kcal/mol. The strongest binding affinities were observed with compounds C7 (-113.84 kcal/mol), HP-14 (-115.23 kcal/mol), and HP (-112.28 kcal/mol), highlighting their effectiveness. Molecular dynamics simulations demonstrated that these compounds bind at the hydrophobic cleft of AgrA, disrupting essential salt bridge interactions between His174-Glu163 and His174-Glu226. This disruption led to structural conformational changes and reduced DNA binding affinity, aligning with experimental findings on biofilm inhibition. The halogenated phenazine compounds effectively inhibit biofilm formation by targeting AgrA, disrupting its DNA-binding function. The study supports the potential of these compounds as antibiofilm agents and provides a foundation for rational drug design targeting the AgrA-DNA interaction. Future research should focus on further optimizing these lead compounds and exploring additional active sites on AgrA to develop novel treatments for biofilm-associated infections.
Collapse
Affiliation(s)
- Prince Manu
- Department of ChemistryKwame Nkrumah University of Science and Technology, Kumasi, Ghana
- Kumasi Centre for Collaborative Research in Tropical MedicineKwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Prisca Baah Nketia
- Department of ChemistryKwame Nkrumah University of Science and Technology, Kumasi, Ghana
- Kumasi Centre for Collaborative Research in Tropical MedicineKwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Priscilla Osei-Poku
- Kumasi Centre for Collaborative Research in Tropical MedicineKwame Nkrumah University of Science and Technology, Kumasi, Ghana
- Department of Biochemistry and BiotechnologyKwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Alexander Kwarteng
- Kumasi Centre for Collaborative Research in Tropical MedicineKwame Nkrumah University of Science and Technology, Kumasi, Ghana
- Department of Biochemistry and BiotechnologyKwame Nkrumah University of Science and Technology, Kumasi, Ghana
| |
Collapse
|
26
|
Marchi J, Ngoc Minh CN, Debarbieux L, Weitz JS. Multi-strain phage induced clearance of bacterial infections. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.07.611814. [PMID: 39282405 PMCID: PMC11398464 DOI: 10.1101/2024.09.07.611814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/20/2024]
Abstract
Bacteriophage (or 'phage' - viruses that infect and kill bacteria) are increasingly considered as a therapeutic alternative to treat antibiotic-resistant bacterial infections. However, bacteria can evolve resistance to phage, presenting a significant challenge to the near- and long-term success of phage therapeutics. Application of mixtures of multiple phage (i.e., 'cocktails') have been proposed to limit the emergence of phage-resistant bacterial mutants that could lead to therapeutic failure. Here, we combine theory and computational models of in vivo phage therapy to study the efficacy of a phage cocktail, composed of two complementary phages motivated by the example of Pseudomonas aeruginosa facing two phages that exploit different surface receptors, LUZ19v and PAK_P1. As confirmed in a Luria-Delbrück fluctuation test, this motivating example serves as a model for instances where bacteria are extremely unlikely to develop simultaneous resistance mutations against both phages. We then quantify therapeutic outcomes given single- or double-phage treatment models, as a function of phage traits and host immune strength. Building upon prior work showing monophage therapy efficacy in immunocompetent hosts, here we show that phage cocktails comprised of phage targeting independent bacterial receptors can improve treatment outcome in immunocompromised hosts and reduce the chance that pathogens simultaneously evolve resistance against phage combinations. The finding of phage cocktail efficacy is qualitatively robust to differences in virus-bacteria interactions and host immune dynamics. Altogether, the combined use of theory and computational analysis highlights the influence of viral life history traits and receptor complementarity when designing and deploying phage cocktails in immunocompetent and immunocompromised hosts.
Collapse
Affiliation(s)
- Jacopo Marchi
- Department of Biology, University of Maryland, College Park, MD, USA
| | - Chau Nguyen Ngoc Minh
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Bacteriophage Bacterium Host, Paris, France and Sorbonne Université, Collège Doctoral, Paris, France
| | - Laurent Debarbieux
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Bacteriophage Bacterium Host, Paris, France
| | - Joshua S. Weitz
- Department of Biology, University of Maryland, College Park, MD USA
- Department of Physics, University of Maryland, College Park, MD USA
- University of Maryland Institute for Health Computing, North Bethesda, MD and Institut de Biologie, École Normale Supérieure, Paris, France
| |
Collapse
|
27
|
Zhu J, Liang Z, Yao H, Wu Z. Identifying Cell-Penetrating Peptides for Effectively Delivering Antimicrobial Molecules into Streptococcus suis. Antibiotics (Basel) 2024; 13:725. [PMID: 39200025 PMCID: PMC11350675 DOI: 10.3390/antibiotics13080725] [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: 06/28/2024] [Revised: 07/27/2024] [Accepted: 07/31/2024] [Indexed: 09/01/2024] Open
Abstract
Cell-penetrating peptides (CPPs) are promising carriers to effectively transport antisense oligonucleotides (ASOs), including peptide nucleic acids (PNAs), into bacterial cells to combat multidrug-resistant bacterial infections, demonstrating significant therapeutic potential. Streptococcus suis, a Gram-positive bacterium, is a major bacterial pathogen in pigs and an emerging zoonotic pathogen. In this study, through the combination of super-resolution structured illumination microscopy (SR-SIM), flow cytometry analysis, and toxicity analysis assays, we investigated the suitability of four CPPs for delivering PNAs into S. suis cells: HIV-1 TAT efficiently penetrated S. suis cells with low toxicity against S. suis; (RXR)4XB had high penetration efficiency with inherent toxicity against S. suis; (KFF)3K showed lower penetration efficiency than HIV-1 TAT and (RXR)4XB; K8 failed to penetrate S. suis cells. HIV-1 TAT-conjugated PNA specific for the essential gyrase A subunit gene (TAT-anti-gyrA PNA) effectively inhibited the growth of S. suis. TAT-anti-gyrA PNA exhibited a significant bactericidal effect on serotypes 2, 4, 5, 7, and 9 strains of S. suis, which are known to cause human infections. Our study demonstrates the potential of CPP-ASO conjugates as new antimicrobial compounds for combating S. suis infections. Furthermore, our findings demonstrate that applying SR-SIM and flow cytometry analysis provides a convenient, intuitive, and cost-effective approach to identifying suitable CPPs for delivering cargo molecules into bacterial cells.
Collapse
Affiliation(s)
- Jinlu Zhu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210014, China; (J.Z.); (Z.L.); (H.Y.)
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210014, China
- WOAH Reference Lab for Swine Streptococcosis, Nanjing 210014, China
| | - Zijing Liang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210014, China; (J.Z.); (Z.L.); (H.Y.)
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210014, China
- WOAH Reference Lab for Swine Streptococcosis, Nanjing 210014, China
| | - Huochun Yao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210014, China; (J.Z.); (Z.L.); (H.Y.)
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210014, China
- WOAH Reference Lab for Swine Streptococcosis, Nanjing 210014, China
| | - Zongfu Wu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210014, China; (J.Z.); (Z.L.); (H.Y.)
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210014, China
- WOAH Reference Lab for Swine Streptococcosis, Nanjing 210014, China
- Guangdong Provincial Key Laboratory of Research on the Technology of Pig-Breeding and Pig-Disease Prevention, Guangzhou 511400, China
| |
Collapse
|
28
|
Braun C, Badiou C, Guironnet-Paquet A, Iwata M, Lenief V, Mosnier A, Beauclair C, Renucci E, Bouschon P, Cuzin R, Briend Y, Patra V, Patot S, Scharschmidt TC, van Wamel W, Lemmens N, Nakajima S, Vandenesh F, Nicolas JF, Lina G, Nosbaum A, Vocanson M. Staphylococcus aureus-specific skin resident memory T cells protect against bacteria colonization but exacerbate atopic dermatitis-like flares in mice. J Allergy Clin Immunol 2024; 154:355-374. [PMID: 38734386 DOI: 10.1016/j.jaci.2024.03.032] [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: 10/27/2023] [Revised: 02/15/2024] [Accepted: 03/26/2024] [Indexed: 05/13/2024]
Abstract
BACKGROUND The contribution of Staphylococcus aureus to the exacerbation of atopic dermatitis (AD) is widely documented, but its role as a primary trigger of AD skin symptoms remains poorly explored. OBJECTIVES This study sought to reappraise the main bacterial factors and underlying immune mechanisms by which S aureus triggers AD-like inflammation. METHODS This study capitalized on a preclinical model, in which different clinical isolates were applied in the absence of any prior experimental skin injury. RESULTS The development of S aureus-induced dermatitis depended on the nature of the S aureus strain, its viability, the concentration of the applied bacterial suspension, the production of secreted and nonsecreted factors, as well as the activation of accessory gene regulatory quorum sensing system. In addition, the rising dermatitis, which exhibited the well-documented AD cytokine signature, was significantly inhibited in inflammasome adaptor apoptosis-associated speck-like protein containing a CARD domain- and monocyte/macrophage-deficient animals, but not in T- and B-cell-deficient mice, suggesting a major role for the innate response in the induction of skin inflammation. However, bacterial exposure generated a robust adaptive immune response against S aureus, and an accumulation of S aureus-specific γδ and CD4+ tissue resident memory T cells at the site of previous dermatitis. The latter both contributed to worsen the flares of AD-like dermatitis on new bacteria exposures, but also, protected the mice from persistent bacterial colonization. CONCLUSIONS These data highlight the induction of unique AD-like inflammation, with the generation of proinflammatory but protective tissue resident memory T cells in a context of natural exposure to pathogenic S aureus strains.
Collapse
Affiliation(s)
- Camille Braun
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, Unité Mixte de Recherche 5308, Centre national de la recherche scientifique, Ecole Normale Supérieure de Lyon, Lyon, France; Service de Pédiatrie, Pneumologie, Allergologie, Mucoviscidose, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, Bron, France
| | - Cédric Badiou
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, Unité Mixte de Recherche 5308, Centre national de la recherche scientifique, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Aurélie Guironnet-Paquet
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, Unité Mixte de Recherche 5308, Centre national de la recherche scientifique, Ecole Normale Supérieure de Lyon, Lyon, France; Etablissement Français du Sang Auvergne Rhône-Alpes, Apheresis Unit, Hôpital Lyon Sud, Pierre-Bénite, France
| | - Masashi Iwata
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Vanina Lenief
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, Unité Mixte de Recherche 5308, Centre national de la recherche scientifique, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Amandine Mosnier
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, Unité Mixte de Recherche 5308, Centre national de la recherche scientifique, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Charlotte Beauclair
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, Unité Mixte de Recherche 5308, Centre national de la recherche scientifique, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Emilie Renucci
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, Unité Mixte de Recherche 5308, Centre national de la recherche scientifique, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Pauline Bouschon
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, Unité Mixte de Recherche 5308, Centre national de la recherche scientifique, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Roxane Cuzin
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, Unité Mixte de Recherche 5308, Centre national de la recherche scientifique, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Yoann Briend
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, Unité Mixte de Recherche 5308, Centre national de la recherche scientifique, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Vijaykumar Patra
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, Unité Mixte de Recherche 5308, Centre national de la recherche scientifique, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Sabine Patot
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, Unité Mixte de Recherche 5308, Centre national de la recherche scientifique, Ecole Normale Supérieure de Lyon, Lyon, France
| | | | - Willem van Wamel
- Department of Medical Microbiology and Infectious Diseases, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Nicole Lemmens
- Department of Medical Microbiology and Infectious Diseases, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Saeko Nakajima
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - François Vandenesh
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, Unité Mixte de Recherche 5308, Centre national de la recherche scientifique, Ecole Normale Supérieure de Lyon, Lyon, France; Service de Microbiologie Clinique, Groupement Hospitalier Nord, Hospices Civils de Lyon, Bron, France; Centre National de Référence des Staphylocoques, Hospices Civils de Lyon, Lyon, France
| | - Jean-François Nicolas
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, Unité Mixte de Recherche 5308, Centre national de la recherche scientifique, Ecole Normale Supérieure de Lyon, Lyon, France; Service d'Allergologie et Immunologie Clinique, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Pierre-Bénite, France
| | - Gérard Lina
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, Unité Mixte de Recherche 5308, Centre national de la recherche scientifique, Ecole Normale Supérieure de Lyon, Lyon, France; Service de Microbiologie Clinique, Groupement Hospitalier Nord, Hospices Civils de Lyon, Bron, France; Centre National de Référence des Staphylocoques, Hospices Civils de Lyon, Lyon, France
| | - Audrey Nosbaum
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, Unité Mixte de Recherche 5308, Centre national de la recherche scientifique, Ecole Normale Supérieure de Lyon, Lyon, France; Service d'Allergologie et Immunologie Clinique, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Pierre-Bénite, France
| | - Marc Vocanson
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, Unité Mixte de Recherche 5308, Centre national de la recherche scientifique, Ecole Normale Supérieure de Lyon, Lyon, France.
| |
Collapse
|
29
|
Wang J, Yang JY, Durairaj P, Wen WH, Sabapathi N, Yang L, Wang B, Jia AQ. Discovery and evaluation of 3-(2-isocyanobenzyl)-1 H-indole derivatives as potential quorum sensing inhibitors for the control of Pseudomonas aeruginosa infections in vitro. RSC Med Chem 2024; 15:d4md00354c. [PMID: 39185452 PMCID: PMC11342129 DOI: 10.1039/d4md00354c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 07/17/2024] [Indexed: 08/27/2024] Open
Abstract
Quorum sensing (QS) inhibition stands out as an innovative therapeutic strategy for combating infections caused by drug-resistant pathogens. In this study, we assessed the potential of 3-(2-isocyanobenzyl)-1H-indole derivatives as novel quorum sensing inhibitors (QSIs). Initial screenings of their QS inhibitory activities were conducted against Pseudomonas aeruginosa PAO1 and Chromobacterium violaceum CV026. Notably, six 3-(2-isocyanobenzyl)-1H-indole derivatives (4, 12, 25, 28, 32, and 33) exhibited promising QS, biofilms, and pyocyanin inhibitory activities under minimum inhibitory concentrations (MICs) against P. aeruginosa PAO1. Among them, 3-(2-isocyano-6-methylbenzyl)-1H-indole (IMBI, 32) emerged as the most promising candidate, demonstrating superior biofilm and pyocyanin inhibition. Further comprehensive studies revealed that derivative 32 at 25 μg mL-1 inhibited biofilm formation by 70% against P. aeruginosa PAO1, as confirmed by scanning electron microscopy (SEM). Additionally, derivative 32 substantially increased the susceptibility of mature biofilms, leading to a 57% destruction of biofilm architecture. In terms of interfering with virulence factors in P. aeruginosa PAO1, derivative 32 (25 μg mL-1) displayed remarkable inhibitory effects on pyocyanin, protease, and extracellular polysaccharides (EPS) by 73%, 51%, and 37%, respectively, exceeding the positive control resveratrol (RSV). Derivative 32 at 25 μg mL-1 also exhibited effective inhibition of swimming and swarming motilities. Moreover, it downregulated the expressions of QS-related genes, including lasI, lasR, rhlI, rhlR, pqsR, sdhB, sucD, sodB, and PA5439, by 1.82- to 10.87-fold. Molecular docking, molecular dynamics simulations (MD), and energy calculations further supported the stable binding of 32 to LasR, RhlI, RhlR, EsaL, and PqsR antagonizing the expression of QS-linked traits. Evaluation of the toxicity of derivative 32 on HEK293T cells via CCK-8 assay demonstrated low cytotoxicity. Overall, this study underscores the efficacy of derivative 32 in inhibiting virulence factors in P. aeruginosa. Derivative 32 emerges as a potential QSI for controlling P. aeruginosa PAO1 infections in vitro and an anti-biofilm agent for restoring or enhancing drug sensitivity in drug-resistant pathogens.
Collapse
Affiliation(s)
- Jiang Wang
- Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University Haikou 570311 China +86 898 68622476
| | - Jing-Yi Yang
- Hainan Branch, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University Sanya 572022 China
| | - Pradeepraj Durairaj
- Center for Translational Research, Shenzhen Bay Laboratory Shenzhen 518132 China
- FAMU-FSU College of Engineering, National High Magnetic Field Laboratory, Florida State University Tallahassee Florida 32310 USA
| | - Wei-Huan Wen
- Center for Translational Research, Shenzhen Bay Laboratory Shenzhen 518132 China
| | - Nadana Sabapathi
- Center for Translational Research, Shenzhen Bay Laboratory Shenzhen 518132 China
| | - Liang Yang
- School of Medicine, Southern University of Science and Technology Shenzhen 518055 China
| | - Bo Wang
- Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University Haikou 570311 China +86 898 68622476
| | - Ai-Qun Jia
- Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University Haikou 570311 China +86 898 68622476
| |
Collapse
|
30
|
Hetta HF, Ramadan YN, Rashed ZI, Alharbi AA, Alsharef S, Alkindy TT, Alkhamali A, Albalawi AS, Battah B, Donadu MG. Quorum Sensing Inhibitors: An Alternative Strategy to Win the Battle against Multidrug-Resistant (MDR) Bacteria. Molecules 2024; 29:3466. [PMID: 39124871 PMCID: PMC11313800 DOI: 10.3390/molecules29153466] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/29/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024] Open
Abstract
Antibiotic resistance is a major problem and a major global health concern. In total, there are 16 million deaths yearly from infectious diseases, and at least 65% of infectious diseases are caused by microbial communities that proliferate through the formation of biofilms. Antibiotic overuse has resulted in the evolution of multidrug-resistant (MDR) microbial strains. As a result, there is now much more interest in non-antibiotic therapies for bacterial infections. Among these revolutionary, non-traditional medications is quorum sensing inhibitors (QSIs). Bacterial cell-to-cell communication is known as quorum sensing (QS), and it is mediated by tiny diffusible signaling molecules known as autoinducers (AIs). QS is dependent on the density of the bacterial population. QS is used by Gram-negative and Gram-positive bacteria to control a wide range of processes; in both scenarios, QS entails the synthesis, identification, and reaction to signaling chemicals, also known as auto-inducers. Since the usual processes regulated by QS are the expression of virulence factors and the creation of biofilms, QS is being investigated as an alternative solution to antibiotic resistance. Consequently, the use of QS-inhibiting agents, such as QSIs and quorum quenching (QQ) enzymes, to interfere with QS seems like a good strategy to prevent bacterial infections. This review sheds light on QS inhibition strategy and mechanisms and discusses how using this approach can aid in winning the battle against resistant bacteria.
Collapse
Affiliation(s)
- Helal F. Hetta
- Division of Microbiology, Immunology and Biotechnology, Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (A.A.A.); (S.A.); (T.T.A.)
| | - Yasmin N. Ramadan
- Department of Microbiology and Immunology, Faculty of Pharmacy, Assiut University, Assiut 71515, Egypt; (Y.N.R.); (Z.I.R.)
| | - Zainab I. Rashed
- Department of Microbiology and Immunology, Faculty of Pharmacy, Assiut University, Assiut 71515, Egypt; (Y.N.R.); (Z.I.R.)
| | - Ahmad A. Alharbi
- Division of Microbiology, Immunology and Biotechnology, Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (A.A.A.); (S.A.); (T.T.A.)
| | - Shomokh Alsharef
- Division of Microbiology, Immunology and Biotechnology, Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (A.A.A.); (S.A.); (T.T.A.)
| | - Tala T. Alkindy
- Division of Microbiology, Immunology and Biotechnology, Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (A.A.A.); (S.A.); (T.T.A.)
| | - Alanoud Alkhamali
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (A.A.); (A.S.A.)
| | - Abdullah S. Albalawi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (A.A.); (A.S.A.)
| | - Basem Battah
- Department of Biochemistry and Microbiology, Faculty of Pharmacy, Antioch Syrian Private University, Maaret Siadnaya 22734, Syria
| | - Matthew G. Donadu
- Hospital Pharmacy, Giovanni Paolo II Hospital, ASL Gallura, 07026 Olbia, Italy;
- Department of Medicine, Surgery and Pharmacy, Scuola di Specializzazione in Farmacia Ospedaliera, University of Sassari, 07100 Sassari, Italy
| |
Collapse
|
31
|
Liu C, Hou J, Ren X, Guo X, Wang B, Song W, Wang L, Wang G. Norwogonin aids in fighting MRSA-induced pneumonia by targeting agrA C to inhibit α-hemolysin production. World J Microbiol Biotechnol 2024; 40:265. [PMID: 38990361 DOI: 10.1007/s11274-024-04052-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 06/12/2024] [Indexed: 07/12/2024]
Abstract
The increasing prevalence of infections related to methicillin-resistant Staphylococcus aureus (MRSA) necessitates the exploration of innovative therapeutic strategies that diverge from conventional antibiotic treatments. This is imperative to effectively combat resistance and manage these infections. The adoption of antivirulence strategies has emerged as a particularly promising avenue. This approach applies a heightened selective pressure on pathogens, thereby diminishing the likelihood of bacteria evolving resistance to antibiotics. In our pursuit of novel therapeutics for treating MRSA infections, we have focused on agents that inhibit the virulence of S. aureus without impeding its growth, aiming to minimize the development of drug resistance. α-Hemolysin, a critical virulence factor encoded by the hla gene, is a cytotoxin that forms pores in host cell membranes and plays a pivotal role in the progression of disease during bacterial infections. Herein, we identified that norwogonin could effectively inhibit Hla production via targeting agrAC, a crucial protein in quorum sensing, resulting in dose-dependent inhibition of hemolytic activity without suppressing S. aureus growth. In vitro assays illustrated that norwogonin decreased the thermal stability of agrAC, providing evidence of interaction between norwogonin and agrAC. Meanwhile, norwogonin alleviated Hla-mediated A549 cell damage and reduced lactate dehydrogenase release. In vivo studies suggested that norwogonin treatment blocked the establishment of a mouse model of pneumonia caused by S. aureus USA300. Notably, norwogonin enhanced the antibacterial potency of oxacillin. In conclusion, norwogonin is a promising candidate for treating S. aureus infections, offering a novel alternative to traditional antibiotics by targeting virulence factors and enhancing the efficacy of existing treatments.
Collapse
Affiliation(s)
- Chang Liu
- Jilin University School of Pharmaceutical Sciences, Jilin University, Changchun, China
- Clinical Medical College, Changchun University of Chinese Medicine, Changchun, China
| | - Juan Hou
- Clinical Medical College, Changchun University of Chinese Medicine, Changchun, China
| | - Xinran Ren
- Jilin University School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Xuerui Guo
- Jilin University School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Bingmei Wang
- Clinical Medical College, Changchun University of Chinese Medicine, Changchun, China
| | - Wu Song
- Clinical Medical College, Changchun University of Chinese Medicine, Changchun, China.
| | - Li Wang
- Clinical Medical College, Changchun University of Chinese Medicine, Changchun, China.
| | - Guangshu Wang
- Jilin University School of Pharmaceutical Sciences, Jilin University, Changchun, China.
| |
Collapse
|
32
|
Vadakkan K, Sathishkumar K, Kuttiyachan Urumbil S, Ponnenkunnathu Govindankutty S, Kumar Ngangbam A, Devi Nongmaithem B. A review of chemical signaling mechanisms underlying quorum sensing and its inhibition in Staphylococcus aureus. Bioorg Chem 2024; 148:107465. [PMID: 38761705 DOI: 10.1016/j.bioorg.2024.107465] [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: 03/24/2024] [Revised: 04/29/2024] [Accepted: 05/14/2024] [Indexed: 05/20/2024]
Abstract
Staphylococcus aureus is a significant bacterium responsible for multiple infections and is a primary cause of fatalities among patients in hospital environments. The advent of pathogenic bacteria such as methicillin-resistant S. aureus revealed the shortcomings of employing antibiotics to treat bacterial infectious diseases. Quorum sensing enhances S. aureus's survivability through signaling processes. Targeting the key components of quorum sensing has drawn much interest nowadays as a promising strategy for combating infections caused by bacteria. Concentrating on the accessory gene regulator quorum-sensing mechanism is the most commonly suggested anti-virulence approach for S.aureus. Quorum quenching is a common strategy for controlling illnesses triggered by microorganisms since it reduces the pathogenicity of bacteria and improves bacterial biofilm susceptibility to antibiotics, thus providing an intriguing prospect for drug discovery. Quorum sensing inhibition reduces selective stresses and constrains the emergence of antibiotic resistance while limiting bacterial pathogenicity. This review examines the quorum sensing mechanisms involved in S. aureus, quorum sensing targets and gene regulation, environmental factors affecting quorum sensing, quorum sensing inhibition, natural products as quorum sensing inhibitory agents and novel therapeutical strategies to target quorum sensing in S. aureus as drug developing technique to augment conventional antibiotic approaches.
Collapse
Affiliation(s)
- Kayeen Vadakkan
- Department of Biotechnology, St. Mary's College (Autonomous), Thrissur, Kerala 680020, India; Manipur International University, Imphal, Manipur 795140, India.
| | - Kuppusamy Sathishkumar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Thandalam, Chennai, Tamil Nadu 602105, India
| | | | | | | | | |
Collapse
|
33
|
Santajit S, Tunyong W, Horpet D, Binmut A, Kong-Ngoen T, Wisessaowapak C, Thavorasak T, Pumirat P, Indrawattana N. Unveiling the Antimicrobial, Anti-Biofilm, and Anti-Quorum-Sensing Potential of Paederia foetida Linn. Leaf Extract against Staphylococcus aureus: An Integrated In Vitro-In Silico Investigation. Antibiotics (Basel) 2024; 13:613. [PMID: 39061295 PMCID: PMC11273848 DOI: 10.3390/antibiotics13070613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 06/18/2024] [Accepted: 06/19/2024] [Indexed: 07/28/2024] Open
Abstract
Antimicrobial resistance poses a global health threat, with Staphylococcus aureus emerging as a notorious pathogen capable of forming stubborn biofilms and regulating virulence through quorum sensing (QS). In the quest for novel therapeutic strategies, this groundbreaking study unveils the therapeutic potential of Paederia foetida Linn., an Asian medicinal plant containing various bioactive compounds, contributing to its antimicrobial activities, in the battle against S. aureus. Through a comprehensive approach, we investigated the effect of ethanolic P. foetida leaf extract on S. aureus biofilms, QS, and antimicrobial activity. The extract exhibited promising inhibitory effects against S. aureus including the biofilm-forming strain and MRSA. Real-time PCR analysis revealed significant downregulation of key virulence and biofilm genes, suggesting interference with QS. Biofilm assays quantified the extract's ability to disrupt and prevent biofilm formation. LC-MS/MS analysis identified quercetin and kaempferol glycosides as potential bioactive constituents, while molecular docking studies explored their binding to the QS transcriptional regulator SarA. Computational ADMET predictions highlighted favorable intestinal absorption but potential P-glycoprotein interactions limiting oral bioavailability. While promising anti-virulence effects were demonstrated, the high molecular weights and excessive hydrogen bond donors/acceptors of the flavonoid glycosides raise concerns regarding drug-likeness and permeability. This integrated study offers valuable insights for developing novel anti-virulence strategies to combat antimicrobial resistance.
Collapse
Affiliation(s)
- Sirijan Santajit
- Department of Medical Technology, School of Allied Health Sciences, Walailak University, Tha Sala 80160, Thailand; (S.S.); (D.H.); (A.B.)
- Research Center in Tropical Pathobiology, Walailak University, Tha Sala 80160, Thailand
| | - Witawat Tunyong
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand; (W.T.); (T.K.-N.); (P.P.)
| | - Dararat Horpet
- Department of Medical Technology, School of Allied Health Sciences, Walailak University, Tha Sala 80160, Thailand; (S.S.); (D.H.); (A.B.)
| | - Asma Binmut
- Department of Medical Technology, School of Allied Health Sciences, Walailak University, Tha Sala 80160, Thailand; (S.S.); (D.H.); (A.B.)
| | - Thida Kong-Ngoen
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand; (W.T.); (T.K.-N.); (P.P.)
| | | | - Techit Thavorasak
- Center of Research Excellence in Therapeutic Proteins and Antibody Engineering, Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand;
| | - Pornpan Pumirat
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand; (W.T.); (T.K.-N.); (P.P.)
| | - Nitaya Indrawattana
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand; (W.T.); (T.K.-N.); (P.P.)
- Siriraj Center of Research Excellence in Allergy and Immunology, Department of Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| |
Collapse
|
34
|
Eisenbraun EL, Vulpis TD, Prosser BN, Horswill AR, Blackwell HE. Synthetic Peptides Capable of Potent Multigroup Staphylococcal Quorum Sensing Activation and Inhibition in Both Cultures and Biofilm Communities. J Am Chem Soc 2024; 146:15941-15954. [PMID: 38832917 PMCID: PMC11321086 DOI: 10.1021/jacs.4c02694] [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] [Indexed: 06/06/2024]
Abstract
The pathogen Staphylococcus epidermidis uses a chemical signaling process, i.e., quorum sensing (QS), to form robust biofilms and cause human infection. Many questions remain about QS in S. epidermidis, as it uses this intercellular communication pathway to both negatively and positively regulate virulence traits. Herein, we report synthetic multigroup agonists and antagonists of the S. epidermidis accessory gene regulator (agr) QS system capable of potent superactivation and complete inhibition, respectively. These macrocyclic peptides maintain full efficacy across the three major agr specificity groups, and their activity can be "mode-switched" from agonist to antagonist via subtle residue-specific structural changes. We describe the design and synthesis of these non-native peptides and demonstrate that they can appreciably decrease biofilm formation on abiotic surfaces, underscoring the potential for agr agonism as a route to block S. epidermidis virulence. Additionally, we show that both the S. epidermidis agonists and antagonists are active in S. aureus, another common pathogen with a related agr system, yet only as antagonists. This result not only revealed one of the most potent agr inhibitors known in S. aureus but also highlighted differences in the mechanisms of agr agonism and antagonism between these related bacteria. Finally, our investigations reveal unexpected inhibitory behavior for certain S. epidermidis agr agonists at sub-activating concentrations, an observation that can be leveraged for the design of future probes with enhanced potencies. Together, these peptides provide a powerful tool set to interrogate the role of QS in S. epidermidis infections and in Staphylococcal pathogenicity in general.
Collapse
Affiliation(s)
- Emma L. Eisenbraun
- Department of Chemistry, University of Wisconsin–Madison, 1101 University Ave., Madison, WI 53706, USA
| | - Troy D. Vulpis
- Department of Chemistry, University of Wisconsin–Madison, 1101 University Ave., Madison, WI 53706, USA
| | - Brendan N. Prosser
- Department of Chemistry, University of Wisconsin–Madison, 1101 University Ave., Madison, WI 53706, USA
| | - Alexander R. Horswill
- Department of Immunology and Microbiology, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA
| | - Helen E. Blackwell
- Department of Chemistry, University of Wisconsin–Madison, 1101 University Ave., Madison, WI 53706, USA
| |
Collapse
|
35
|
Inagaki R, Koshiba A, Nasuno E, Kato N. Eliminating extracellular autoinducing peptide signals inhibits the Staphylococcus aureus quorum sensing agr system. Biochem Biophys Res Commun 2024; 711:149912. [PMID: 38615572 DOI: 10.1016/j.bbrc.2024.149912] [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: 03/11/2024] [Accepted: 04/05/2024] [Indexed: 04/16/2024]
Abstract
An accessory gene regulator (agr) in the quorum sensing (QS) system in Staphylococcus aureus contributes to host infection, virulence factor production, and resistance to oxidative damage. Artificially maintaining the inactive state of agr QS impedes the host infection strategy of S. aureus and inhibits toxin production. The QS system performs intercellular signal transduction, which is activated by the mature autoinducer peptide (AIP). It is released from cells after AgrD peptide processing as an intercellular signal associated with increased bacterial cell density. This study evaluated the effectiveness of inhibiting agr QS wherein AIP trap carriers were made to coexist when culturing Staphylococcus aureus. Immersing a nitrocellulose (NC) membrane in Staphylococcus aureus ATCC 12600 culture inhibited QS-dependent α-hemolysin production, which significantly reduced the hemolysis ratio of sheep red blood cells by the culture supernatant. A quartz crystal microbalance analysis supported AIP adsorption onto the NC membrane. Adding the NC membrane during culture was found to maintain the expression levels of the agr QS gene agrA and α-hemolysin gene hla lower than that when it was not added. Eliminating extracellular AIP signals allowed agr QS to remain inactive and prevented QS-dependent α-hemolysin expression. Isolating intercellular signals secreted outside the cell is an effective strategy to suppress gene expression in bacterial cells that collaborate via intercellular signaling.
Collapse
Affiliation(s)
- Ruki Inagaki
- Graduate School of Regional Development and Creativity, Utsunomiya University, 7-1-2 Yoto, Utsunomiya, Tochigi, 321-8585, Japan
| | - Ayaka Koshiba
- Graduate School of Regional Development and Creativity, Utsunomiya University, 7-1-2 Yoto, Utsunomiya, Tochigi, 321-8585, Japan
| | - Eri Nasuno
- Graduate School of Regional Development and Creativity, Utsunomiya University, 7-1-2 Yoto, Utsunomiya, Tochigi, 321-8585, Japan.
| | - Norihiro Kato
- Graduate School of Regional Development and Creativity, Utsunomiya University, 7-1-2 Yoto, Utsunomiya, Tochigi, 321-8585, Japan.
| |
Collapse
|
36
|
Sabino YNV, de Araújo Domingues KC, O'Connor PM, Marques PH, Santos EH, Tótola MR, Abreu LM, de Queiroz MV, Cotter PD, Mantovani HC. Bacillus velezensis iturins inhibit the hemolytic activity of Staphylococcus aureus. Sci Rep 2024; 14:9469. [PMID: 38658583 PMCID: PMC11043418 DOI: 10.1038/s41598-024-58973-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 04/05/2024] [Indexed: 04/26/2024] Open
Abstract
Bovine mastitis caused by S. aureus has a major economic impact on the dairy sector. With the crucial need for new therapies, anti-virulence strategies have gained attention as alternatives to antibiotics. Here we aimed to identify novel compounds that inhibit the production/activity of hemolysins, a virulence factor of S. aureus associated with mastitis severity. We screened Bacillus strains obtained from diverse sources for compounds showing anti-hemolytic activity. Our results demonstrate that lipopeptides produced by Bacillus spp. completely prevented the hemolytic activity of S. aureus at certain concentrations. Following purification, both iturins, fengycins, and surfactins were able to reduce hemolysis caused by S. aureus, with iturins showing the highest anti-hemolytic activity (up to 76% reduction). The lipopeptides showed an effect at the post-translational level. Molecular docking simulations demonstrated that these compounds can bind to hemolysin, possibly interfering with enzyme action. Lastly, molecular dynamics analysis indicated general stability of important residues for hemolysin activity as well as the presence of hydrogen bonds between iturins and these residues, with longevous interactions. Our data reveals, for the first time, an anti-hemolytic activity of lipopeptides and highlights the potential application of iturins as an anti-virulence therapy to control bovine mastitis caused by S. aureus.
Collapse
Affiliation(s)
| | | | | | - Pedro Henrique Marques
- Department of Microbiology, Immunology and Parasitology, Federal University of Triângulo Mineiro, Uberaba, MG, Brazil
| | - Eduardo Horta Santos
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | | | | | - Paul D Cotter
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, Cork, Ireland
- APC Microbiome Ireland, Cork, Ireland
| | | |
Collapse
|
37
|
Srivastava A, Verma N, Kumar V, Apoorva P, Agarwal V. Biofilm inhibition/eradication: exploring strategies and confronting challenges in combatting biofilm. Arch Microbiol 2024; 206:212. [PMID: 38616221 DOI: 10.1007/s00203-024-03938-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 03/04/2024] [Accepted: 03/20/2024] [Indexed: 04/16/2024]
Abstract
Biofilms are complex communities of microorganisms enclosed in a self-produced extracellular matrix, posing a significant threat to different sectors, including healthcare and industry. This review provides an overview of the challenges faced due to biofilm formation and different novel strategies that can combat biofilm formation. Bacteria inside the biofilm exhibit increased resistance against different antimicrobial agents, including conventional antibiotics, which can lead to severe problems in livestock and animals, including humans. In addition, biofilm formation also imposes heavy economic pressure on industries. Hence it becomes necessary to explore newer alternatives to eradicate biofilms effectively without applying selection pressure on the bacteria. Excessive usage of antibiotics may also lead to an increase in the number of resistant strains as bacteria employ an advanced antimicrobial resistance mechanism. This review provides insight into multifaceted technologies like quorum sensing inhibition, enzymes, antimicrobial peptides, bacteriophage, phytocompounds, and nanotechnology to neutralize biofilms without developing antimicrobial resistance (AMR). Furthermore, it will pave the way for developing newer therapeutic agents to deal with biofilms more efficiently.
Collapse
Affiliation(s)
- Anmol Srivastava
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj, 211004, Uttar Pradesh, India
| | - Nidhi Verma
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj, 211004, Uttar Pradesh, India
| | - Vivek Kumar
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj, 211004, Uttar Pradesh, India
| | - Pragati Apoorva
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj, 211004, Uttar Pradesh, India
| | - Vishnu Agarwal
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj, 211004, Uttar Pradesh, India.
| |
Collapse
|
38
|
Yamaguchi J, Manome T, Hara Y, Yamazaki Y, Nakamura Y, Ishibashi M, Takaya A. Physalin H, physalin B, and isophysalin B suppress the quorum-sensing function of Staphylococcus aureus by binding to AgrA. Front Pharmacol 2024; 15:1365815. [PMID: 38659576 PMCID: PMC11039898 DOI: 10.3389/fphar.2024.1365815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 03/25/2024] [Indexed: 04/26/2024] Open
Abstract
The virulence of Staphylococcus aureus, including methicillin-resistant S. aureus (MRSA), depends on the expression of toxins and virulence factors controlled by the quorum-sensing (QS) system, encoded on the virulence accessory gene regulator (agr) locus. The aim of this study was to identify a phytochemical that inhibits Agr-QS function and to elucidate its mechanism. We screened 577 compounds and identified physalin H, physalin B, and isophysalin B--phytochemicals belonging to physalins found in plants of the Solanaceae family--as novel Agr-QS modulators. Biological analyses and in vitro protein-DNA binding assays suggested that these physalins suppress gene expression related to the Agr-QS system by inhibiting binding of the key response regulator AgrA to the agr promoters, reducing the function of hemolytic toxins downstream of these genes in MRSA. Furthermore, although physalin F suppressed gene expression in the Agr-QS system, its anti-hemolytic activity was lower than that of physalins H, B, and isophysalin B. Conversely, five physalins isolated from the same plant with the ability to suppress Agr-QS did not reduce bacterial Agr-QS activity but inhibited AgrA binding to DNA in vitro. A docking simulation revealed that physalin interacts with the DNA-binding site of AgrA in three docking states. The carbonyl oxygens at C-1 and C-18 of physalins, which can suppress Agr-QS, were directed to residues N201 and R198 of AgrA, respectively, whereas these carbonyl oxygens of physalins, without Agr-QS suppression activity, were oriented in different directions. Next, 100-ns molecular dynamics simulations revealed that the hydrogen bond formed between the carbonyl oxygen at C-15 of physalins and L186 of AgrA functions as an anchor, sustaining the interaction between the carbonyl oxygen at C-1 of physalins and N201 of AgrA. Thus, these results suggest that physalin H, physalin B, and isophysalin B inhibit the interaction of AgrA with the agr promoters by binding to the DNA-binding site of AgrA, suppressing the Agr-QS function of S. aureus. Physalins that suppress the Agr-QS function are proposed as potential lead compounds in the anti-virulence strategy for MRSA infections.
Collapse
Affiliation(s)
- Junpei Yamaguchi
- Department of Infection Control Science, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Teruhisa Manome
- Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
- Laboratory of Natural Products Chemistry, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Yasumasa Hara
- Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
- Faculty of Agriculture, Kagawa University, Takamatsu, Japan
| | - Yuriko Yamazaki
- Cutaneous Allergy and Host Defense, Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Department of Dermatology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yuumi Nakamura
- Cutaneous Allergy and Host Defense, Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Department of Dermatology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Masami Ishibashi
- Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
- School of Pharmacy at Fukuoka, International University of Health and Welfare, Okawa, Japan
| | - Akiko Takaya
- Department of Infection Control Science, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
- Medical Mycology Research Center, Chiba University, Chiba, Japan
- Plant Molecular Science Center, Chiba University, Chiba, Japan
| |
Collapse
|
39
|
Aboelnaga N, Elsayed SW, Abdelsalam NA, Salem S, Saif NA, Elsayed M, Ayman S, Nasr M, Elhadidy M. Deciphering the dynamics of methicillin-resistant Staphylococcus aureus biofilm formation: from molecular signaling to nanotherapeutic advances. Cell Commun Signal 2024; 22:188. [PMID: 38519959 PMCID: PMC10958940 DOI: 10.1186/s12964-024-01511-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 02/01/2024] [Indexed: 03/25/2024] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) represents a global threat, necessitating the development of effective solutions to combat this emerging superbug. In response to selective pressures within healthcare, community, and livestock settings, MRSA has evolved increased biofilm formation as a multifaceted virulence and defensive mechanism, enabling the bacterium to thrive in harsh conditions. This review discusses the molecular mechanisms contributing to biofilm formation across its developmental stages, hence representing a step forward in developing promising strategies for impeding or eradicating biofilms. During staphylococcal biofilm development, cell wall-anchored proteins attach bacterial cells to biotic or abiotic surfaces; extracellular polymeric substances build scaffolds for biofilm formation; the cidABC operon controls cell lysis within the biofilm, and proteases facilitate dispersal. Beside the three main sequential stages of biofilm formation (attachment, maturation, and dispersal), this review unveils two unique developmental stages in the biofilm formation process for MRSA; multiplication and exodus. We also highlighted the quorum sensing as a cell-to-cell communication process, allowing distant bacterial cells to adapt to the conditions surrounding the bacterial biofilm. In S. aureus, the quorum sensing process is mediated by autoinducing peptides (AIPs) as signaling molecules, with the accessory gene regulator system playing a pivotal role in orchestrating the production of AIPs and various virulence factors. Several quorum inhibitors showed promising anti-virulence and antibiofilm effects that vary in type and function according to the targeted molecule. Disrupting the biofilm architecture and eradicating sessile bacterial cells are crucial steps to prevent colonization on other surfaces or organs. In this context, nanoparticles emerge as efficient carriers for delivering antimicrobial and antibiofilm agents throughout the biofilm architecture. Although metal-based nanoparticles have been previously used in combatting biofilms, its non-degradability and toxicity within the human body presents a real challenge. Therefore, organic nanoparticles in conjunction with quorum inhibitors have been proposed as a promising strategy against biofilms. As nanotherapeutics continue to gain recognition as an antibiofilm strategy, the development of more antibiofilm nanotherapeutics could offer a promising solution to combat biofilm-mediated resistance.
Collapse
Affiliation(s)
- Nirmeen Aboelnaga
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, Egypt
| | - Salma W Elsayed
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, Egypt
- Department of Microbiology & Immunology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Nehal Adel Abdelsalam
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, Egypt
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Salma Salem
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, Egypt
| | - Nehal A Saif
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, Egypt
| | - Manar Elsayed
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, Egypt
| | - Shehab Ayman
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, Egypt
| | - Maha Nasr
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Mohamed Elhadidy
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt.
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, Egypt.
- Department of Bacteriology, Mycology and Immunology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt.
| |
Collapse
|
40
|
Kameswaran S, Gujjala S, Zhang S, Kondeti S, Mahalingam S, Bangeppagari M, Bellemkonda R. Quenching and quorum sensing in bacterial bio-films. Res Microbiol 2024; 175:104085. [PMID: 37268165 DOI: 10.1016/j.resmic.2023.104085] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/24/2023] [Accepted: 05/25/2023] [Indexed: 06/04/2023]
Abstract
Quorum sensing (QS) is the ability of bacteria to monitor their population density and adjust gene expression accordingly. QS-regulated processes include host-microbe interactions, horizontal gene transfer, and multicellular behaviours (such as the growth and development of biofilm). The creation, transfer, and perception of bacterial chemicals known as autoinducers or QS signals are necessary for QS signalling (e.g. N-acylhomoserine lactones). Quorum quenching (QQ), another name for the disruption of QS signalling, comprises a wide range of events and mechanisms that are described and analysed in this study. In order to better comprehend the targets of the QQ phenomena that organisms have naturally developed and are currently being actively researched from practical perspectives, we first surveyed the diversity of QS-signals and QS-associated responses. Next, the mechanisms, molecular players, and targets related to QS interference are discussed, with a focus on natural QQ enzymes and compounds that function as QS inhibitors. To illustrate the processes and biological functions of QS inhibition in microbe-microbe and host-microbe interactions, a few QQ paradigms are described in detail. Finally, certain QQ techniques are offered as potential instruments in a variety of industries, including agriculture, medical, aquaculture, crop production, and anti-biofouling areas.
Collapse
Affiliation(s)
- Srinivasan Kameswaran
- Department of Botany, Vikrama Simhapuri University College, Kavali, Andhra Pradesh, India
| | - Sudhakara Gujjala
- Department of Biochemistry, Sri Krishnadevaray a University, Ananthapuram, Andhra Pradesh, India
| | - Shaoqing Zhang
- School of Chemistry and Civil Engineering, Shaoguan University, Shaoguan, 512005, PR China
| | - Suresh Kondeti
- Multi-Disciplinary Research Unit, Nizam's Institute of Medical Sciences, Hyderabad, 500082, India
| | - Sundararajan Mahalingam
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Manjunatha Bangeppagari
- Department of Cell Biology & Molecular Genetics, Sri Devaraj Urs Academy of Higher Education and Research (Deemed to Be University), Tamaka, Kolar, 563103, Karnataka, India
| | - Ramesh Bellemkonda
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
41
|
Elfaky MA. Unveiling the hidden language of bacteria: anti-quorum sensing strategies for gram-negative bacteria infection control. Arch Microbiol 2024; 206:124. [PMID: 38409503 DOI: 10.1007/s00203-024-03900-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 02/04/2024] [Accepted: 02/16/2024] [Indexed: 02/28/2024]
Abstract
Quorum sensing (QS) is a communication mechanism employed by many bacteria to regulate gene expression in a population density-dependent manner. It plays a crucial role in coordinating various bacterial behaviors, including biofilm formation, virulence factor production, and antibiotic resistance. However, the dysregulation of QS can lead to detrimental effects, making it an attractive target for developing novel therapeutic strategies. Anti-QS approaches aim to interfere with QS signaling pathways, inhibiting the communication between bacteria, and disrupting their coordinated activities. Various strategies have been explored to achieve this goal. Advances in understanding QS mechanisms and the discovery of new targets have paved the way for the development of innovative anti-QS approaches. Combining multiple anti-QS strategies or utilizing them in combination with traditional antibiotics holds great promise for combating bacterial infections and addressing the challenges posed by antibiotic resistance. Anti-QS approaches offer a diverse range of strategies including natural compounds, antibody-mediated quorum quenching (QQ), computer-aided drug design for QQ, repurposing of Drugs approved by FDA as anti-QS agents and modulating quorum-sensing molecules which were discussed in detail in this review. This review, comprehensively and for the first time, sheds light on the significance of diverse anti-QS strategies in solving antimicrobial resistance problem in Gram-negative microbial infection.
Collapse
Affiliation(s)
- Mahmoud A Elfaky
- Department of Natural Products, Faculty of Pharmacy, King Abdulaziz University, Jeddah, 21589, Saudi Arabia.
- Centre for Artificial Intelligence in Precision Medicines, King Abdulaziz University, Jeddah, 21589, Saudi Arabia.
| |
Collapse
|
42
|
Vinodhini V, Kavitha M. Deciphering agr quorum sensing in Staphylococcus aureus: insights and therapeutic prospects. Mol Biol Rep 2024; 51:155. [PMID: 38252331 DOI: 10.1007/s11033-023-08930-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 10/10/2023] [Indexed: 01/23/2024]
Abstract
The emergence of superbugs like methicillin-resistant Staphylococcus aureus exposed the limitations of treating microbial infections using antibiotics. At present, the discovery of novel and convincing therapeutic methods are being executed increasingly as possible substitutes to conventional antibiotic therapies. The quorum sensing helps Staphylococcus aureus become more viable through their signaling mechanisms. In recent years, targeting the prominent factors of quorum sensing has obtained remarkable attention as a futuristic approach to dealing with bacterial pathogenicity. The standard antibiotic therapy intends to inhibit the organism by targeting specific molecules and afford a chance for the evolution of antibiotic resistance. This prompts the development of novel therapeutic strategies like inhibiting quorum sensing that can limit bacterial virulence by decreasing the selective pressure, thereby restricting antibiotic resistance evolution. This review furnishes new insights into the accessory gene regulator quorum sensing in Staphylococcus aureus and its inhibition by targeting the genes that regulate the operon. Further, this review comprehensively explores the inhibitors reported up to date and their specific targets and discusses their potentially ineffective alternative therapy against methicillin-resistant Staphylococcus aureus.
Collapse
Affiliation(s)
- V Vinodhini
- School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - M Kavitha
- School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India.
| |
Collapse
|
43
|
Joy A, Seethi V F, Cyriac MC, Habeeb J, Sudhakaran S, Shah S. Modelling of AgrA inhibitors to combat anti-microbial resistance in Staphylococcus aureus. J Biomol Struct Dyn 2024; 42:551-558. [PMID: 37166373 DOI: 10.1080/07391102.2023.2203260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 03/15/2023] [Indexed: 05/12/2023]
Abstract
Staphylococcus aureus is a Gram-positive bacterium found on human skin that causes skin and soft tissue infections, as well as pneumonia, osteomyelitis, and endocarditis. The prevalence of antibiotic resistant strains has made the treatments less effective. An efficient alternate method for battling these contagious diseases is anti-virulence strategy. The AgrA protein, a key activator of Accessory Gene Regulator system in S. aureus, is vital to the virulence of the organism and, consequently, its pathogenesis. Using a Machine Learning algorithm, the Support Vector Machine (SVM), and a ligand-based pharmacophore modelling method, prediction models of AgrA inhibitors were developed. The metrics of the SVM model were inadequate, hence it was not used for virtual screening. For ligand-based pharmacophore modelling, 14 of 29 compounds were removed from the active set due to a lack of shared pharmacophore properties, and 504 compounds were designated as decoys. A 3D pharmacophore model was created using LigandScout 4.4.5, with a fit score of 57.48, including a positive ionizable group, one hydrogen bond donor, and three hydrogen bond acceptors. The model after further validation was used to virtually screen an external database which resulted in six hits. These compounds were docked with the AgrA domain crystal structure to determine the inhibitor activity. Further, each docked complex was subjected to a 100 ns molecular dynamics simulation. CID238 and CID20510252 demonstrated potent inhibitory binding interactions and hence can be used to develop AgrA inhibitors in future after proper validation.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Amitha Joy
- Department of Biotechnology, Sahrdaya College of Engineering and Technology, Thrissur, Kerala, India
| | | | - Marria C Cyriac
- Department of Biotechnology, Sahrdaya College of Engineering and Technology, Thrissur, Kerala, India
| | - Jasmin Habeeb
- Division of Crop Improvement, ICAR-Central Plantation Crops Research Institute, Kasaragod, Kerala, India
| | | | - Shaheen Shah
- Genomics Central [MaGenomics], Thrissur, Kerala, India
| |
Collapse
|
44
|
Sabino YNV, Araújo Domingues KCD, Mathur H, Gómez-Mascaraque LG, Drouin G, Martínez-Abad A, Tótola MR, Abreu LM, Cotter PD, Mantovani HC. Exopolysaccharides produced by Bacillus spp. inhibit biofilm formation by Staphylococcus aureus strains associated with bovine mastitis. Int J Biol Macromol 2023; 253:126689. [PMID: 37678679 DOI: 10.1016/j.ijbiomac.2023.126689] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 08/22/2023] [Accepted: 09/02/2023] [Indexed: 09/09/2023]
Abstract
Bovine mastitis is a costly disease in the dairy sector worldwide. Here the objective was to identify and characterize anti-biofilm compounds produced by Bacillus spp. against S. aureus associated with bovine mastitis. Results showed that cell-free supernatants of three Bacillus strains (out of 33 analysed) reduced S. aureus biofilm formation by approximately 40 % without affecting bacterial growth. The anti-biofilm activity was associated with exopolysaccharides (EPS) secreted by Bacillus spp. The EPS decreased S. aureus biofilm formation in a dose-dependent manner, inhibiting biofilm formation by 83 % at 1 mg/mL. The EPS also showed some biofilm disruption activity (up to 36.4 %), which may be partially mediated by increased expression of the aur gene. The characterization of EPS produced by Bacillus velezensis 87 and B. velezensis TR47II revealed macromolecules with molecular weights of 31.2 and 33.7 kDa, respectively. These macromolecules were composed mainly of glucose (mean = 218.5 μg/mg) and mannose (mean = 241.5 μg/mg) and had similar functional groups (pyranose ring, beta-type glycosidic linkage, and alkynes) as revealed by FT-IR. In conclusion, this study shows the potential applications of EPS produced by B. velezensis as an anti-biofilm compound that could contribute to the treatment of bovine mastitis caused by S. aureus.
Collapse
Affiliation(s)
| | | | - Harsh Mathur
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, Ireland
| | | | - Gaetan Drouin
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, Ireland
| | | | - Marcos Rogério Tótola
- Department of Microbiology, Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brazil
| | - Lucas Magalhães Abreu
- Department of Phytopathology, Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brazil
| | - Paul D Cotter
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, Ireland; APC Microbiome Ireland, Cork, Ireland
| | - Hilario Cuquetto Mantovani
- Department of Microbiology, Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brazil; Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
45
|
Lin H, Song L, Zhou S, Fan C, Zhang M, Huang R, Zhou R, Qiu J, Ma S, He J. A Hybrid Antimicrobial Peptide Targeting Staphylococcus aureus with a Dual Function of Inhibiting Quorum Sensing Signaling and an Antibacterial Effect. J Med Chem 2023; 66:17105-17117. [PMID: 38099725 DOI: 10.1021/acs.jmedchem.3c02027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
Community-associated methicillin-resistant Staphylococcus aureus (MRSA) is now a major cause of bacterial infection. Antivirulence therapy does not stimulate evolution of a pathogen toward a resistant phenotype, providing a novel method to treat infectious diseases. Here, we used a cyclic peptide of CP7, an AIP-III variant that specifically inhibited the virulence and biofilm formation of Staphylococcus aureus (S. aureus) in a nonbiocidal manner, to conjugate with a broad-spectrum antimicrobial peptide (AMP) via two N-termini to obtain a hybrid AMP called CP7-FP13-2. This peptide not only specifically inhibited the production of virulence of S. aureus at low micromolar concentrations but also killed S. aureus, including MRSA, by disrupting the integrity of the bacterial cell membrane. In addition, CP7-FP13-2 inhibited the formation of the S. aureus biofilm and showed good antimicrobial efficacy against the S. aureus-infected Kunming mice model. Therefore, this study provides a promising strategy against the resistance and virulence of S. aureus.
Collapse
Affiliation(s)
- Haixing Lin
- Group of peptides and natural products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, P. R. China
- Department of Urology, Tongren Municipal People's Hospital, 120 Taoyuan Avenue, Tongren, Guizhou 554300, P. R. China
| | - Li Song
- Group of peptides and natural products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, P. R. China
| | - Shaofen Zhou
- Group of peptides and natural products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, P. R. China
| | - Cuiqiong Fan
- Group of peptides and natural products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, P. R. China
| | - Minna Zhang
- Department of Nephrology, Tongren Municipal People's Hospital, 120 Taoyuan Avenue, Tongren, Guizhou 554300, P. R. China
| | - Ruifeng Huang
- Group of peptides and natural products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, P. R. China
| | - Runhong Zhou
- Group of peptides and natural products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, P. R. China
| | - Jingnan Qiu
- Group of peptides and natural products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, P. R. China
| | - Shuaiqi Ma
- Group of peptides and natural products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, P. R. China
| | - Jian He
- Group of peptides and natural products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, P. R. China
| |
Collapse
|
46
|
Hassan RM, Abd El-Maksoud MS, Ghannam IAY, El-Azzouny AAS, Aboul-Enein MN. Synthetic non-toxic anti-biofilm agents as a strategy in combating bacterial resistance. Eur J Med Chem 2023; 262:115867. [PMID: 37866335 DOI: 10.1016/j.ejmech.2023.115867] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/26/2023] [Accepted: 10/09/2023] [Indexed: 10/24/2023]
Abstract
The tremendous increase in the bacterial resistance to the available antibiotics is a serious problem for the treatment of various infections. Biofilm formation in bacteria significantly contributes to the bacterial survival in host cells, and is considered as an crucial factor, responsible for bacterial resistance. The response of the bacterial cells in the biofilm to antibiotics is completely different from that of the free floating planktonic cells of the same strain. The anti-biofilm agents that could inhibit the biofilm production without affecting the bacterial growth, apply less selective pressure over the bacterial strains than the traditional antibiotics; thus the development of bacterial resistance would be of low incidence. Many attempts have been performed to discover novel agents capable of interfering with the bacterial biofilm life cycle, and several compounds have shown promising activities in suppressing the biofilm production or in dispersing mature existing biofilms. This review describes the different chemical classes that have anti-biofilm effects against different Gram-positive and Gram-negative bacteria without affecting the bacterial growth.
Collapse
Affiliation(s)
- Rasha Mohamed Hassan
- Medicinal and Pharmaceutical Chemistry Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre (ID: 60014618), P.O. 12622, Dokki, Giza, Egypt.
| | - Mohamed Samir Abd El-Maksoud
- Medicinal and Pharmaceutical Chemistry Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre (ID: 60014618), P.O. 12622, Dokki, Giza, Egypt
| | - Iman Ahmed Youssef Ghannam
- Chemistry of Natural and Microbial Products Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre, Dokki, Cairo, 12622, Egypt
| | - Aida Abdel-Sattar El-Azzouny
- Medicinal and Pharmaceutical Chemistry Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre (ID: 60014618), P.O. 12622, Dokki, Giza, Egypt
| | - Mohamed Nabil Aboul-Enein
- Medicinal and Pharmaceutical Chemistry Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre (ID: 60014618), P.O. 12622, Dokki, Giza, Egypt.
| |
Collapse
|
47
|
Wang J, Yang JY, Durairaj P, Wang W, Tang S, Wang D, Cai CY, Jia AQ. Developing 3-(2-Isocyano-6-methylbenzyl)-1 H-indole Derivatives to Enhance the Susceptibility of Serratia marcescens by Occluding Quorum Sensing. ACS Infect Dis 2023; 9:2607-2621. [PMID: 37971550 PMCID: PMC10715256 DOI: 10.1021/acsinfecdis.3c00433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/29/2023] [Accepted: 11/01/2023] [Indexed: 11/19/2023]
Abstract
Quorum sensing (QS) inhibition is recognized as a novel antimicrobial target for infections caused by drug-resistant pathogens and is an attractive strategy for antipathogenic agent development. We designed and synthesized three parts of 3-(2-isocyanobenzyl)-1H-indole derivatives and tested their activity as novel quorum sensing inhibitors (QSIs). 3-(2-Isocyanobenzyl)-1H-indole derivatives demonstrated promising QS, biofilms, and prodigiosin inhibitory activities against Serratia marcescens at subminimum inhibitory concentrations (sub-MICs). In particular, 3-(2-isocyano-6-methylbenzyl)-1H-indole (IMBI, 32) was identified as the best candidate based on several screening assays, including biofilm and prodigiosin inhibition. Further studies demonstrated that exposure to IMBI at 1.56 μg/mL to S. marcescens NJ01 significantly inhibited the formation of biofilms by 42%. The IMBI treatment on S. marcescens NJ01 notably enhanced the susceptibility of the formed biofilms, destroying the architecture of the biofilms by up to 40%, as evidenced by scanning electron microscopy (SEM) and confocal laser scanning microscopy (CLSM). For interference of virulence factors in S. marcescens NJ01, IMBI at 3.12 μg/mL inhibited the activity of protease and extracellular polysaccharides (EPS) by 17% and 51%, respectively, which were higher than that of the positive control vanillic acid (VAN). Furthermore, IMBI downregulated the expression of QS- and biofilm-related genes fimA, bsmA, pigP, flhC, rssB, fimC, and rsmA by 1.02- to 2.74-fold. To confirm these findings, molecular docking was performed, which indicated that the binding of IMBI to SmaR, RhlI, RhlR, LasR, and CviR could antagonize the expression of QS-linked traits. In addition, molecular dynamic simulations (MD) and energy calculations indicated that the binding of receptors with IMBI was extremely stable. The biofilms of S. marcescens NJ01 were markedly reduced by 50% when IMBI (0.39 μg/mL) was combined with kanamycin (0.15 μg/mL). In conclusion, this study highlights the potency of IMBI in inhibiting the virulence factors of S. marcescens. IMBI has all the potential to be developed as an effective and efficient QS inhibitor and antibiofilm agent in order to restore or improve antimicrobial drug sensitivity.
Collapse
Affiliation(s)
- Jiang Wang
- Key
Laboratory of Tropical Biological Resources of Ministry of Education,
School of Pharmaceutical Sciences, Hainan
University, Haikou 570228, China
- Hainan
General Hospital, Hainan Affiliated Hospital
of Hainan Medical University, Haikou 570311, China
- Center
for Translational Research, Shenzhen Bay
Laboratory, Shenzhen 518132, China
| | - Jing-Yi Yang
- Key
Laboratory of Tropical Biological Resources of Ministry of Education,
School of Pharmaceutical Sciences, Hainan
University, Haikou 570228, China
| | - Pradeepraj Durairaj
- Center
for Translational Research, Shenzhen Bay
Laboratory, Shenzhen 518132, China
| | - Wei Wang
- Key
Laboratory of Tropical Biological Resources of Ministry of Education,
School of Pharmaceutical Sciences, Hainan
University, Haikou 570228, China
| | - Shi Tang
- Key
Laboratory of Tropical Biological Resources of Ministry of Education,
School of Pharmaceutical Sciences, Hainan
University, Haikou 570228, China
| | - Dayong Wang
- Key
Laboratory of Tropical Biological Resources of Ministry of Education,
School of Pharmaceutical Sciences, Hainan
University, Haikou 570228, China
| | - Chao-Yun Cai
- Center
for Translational Research, Shenzhen Bay
Laboratory, Shenzhen 518132, China
| | - Ai-Qun Jia
- Hainan
General Hospital, Hainan Affiliated Hospital
of Hainan Medical University, Haikou 570311, China
| |
Collapse
|
48
|
Chen F, Di H, Wang Y, Peng C, Chen R, Pan H, Yang CG, Liang H, Lan L. The enzyme activity of sortase A is regulated by phosphorylation in Staphylococcus aureus. Virulence 2023; 14:2171641. [PMID: 36694285 PMCID: PMC9928477 DOI: 10.1080/21505594.2023.2171641] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
In many Gram-positive bacteria, the transpeptidase enzyme sortase A (SrtA) anchors surface proteins to cell wall and plays a critical role in the bacterial pathogenesis. Here, we show that in Staphylococcus aureus, an important human pathogen, the SrtA is phosphorylated by serine/threonine protein kinase Stk1. S. aureus SrtA can also be phosphorylated by small-molecule phosphodonor acetyl phosphate (AcP) in vitro. We determined that various amino acid residues of S. aureus SrtA are subject to phosphorylation, primarily on its catalytic site residue cysteine-184 in the context of a bacterial cell lysate. Both Stk1 and AcP-mediated phosphorylation inhibited the enzyme activity of SrtA in vitro. Consequently, deletion of gene (i.e. stp1) encoding serine/threonine phosphatase Stp1, the corresponding phosphatase of Stk1, caused an increase in the phosphorylation level of SrtA. The stp1 deletion mutant mimicked the phenotypic traits of srtA deletion mutant (i.e. attenuated growth where either haemoglobin or haem as a sole iron source and reduced liver infections in a mouse model of systemic infection). Importantly, the phenotypic defects of the stp1 deletion mutant can be alleviated by overexpressing srtA. Taken together, our finding suggests that phosphorylation plays an important role in modulating the activity of SrtA in S. aureus.
Collapse
Affiliation(s)
- Feifei Chen
- College of Life Science, Northwest University, Xi’an, China,State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China,Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
| | - Hongxia Di
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China,University of Chinese Academy of Sciences, Beijing, China
| | - Yanhui Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China,University of Chinese Academy of Sciences, Beijing, China
| | - Chao Peng
- National Facility for Protein Science in Shanghai, Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Science, Shanghai, China
| | - Rongrong Chen
- Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China,University of Chinese Academy of Sciences, Beijing, China
| | - Huiwen Pan
- Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China,University of Chinese Academy of Sciences, Beijing, China
| | - Cai-Guang Yang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China,Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China,University of Chinese Academy of Sciences, Beijing, China
| | - Haihua Liang
- College of Life Science, Northwest University, Xi’an, China,School of Medicine, Southern University of Science and Technology, Shenzhen, China,Haihua Liang School of Medicine Southern University of Science and Technology, Shenzhen, China
| | - Lefu Lan
- College of Life Science, Northwest University, Xi’an, China,State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China,Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China,University of Chinese Academy of Sciences, Beijing, China,CONTACT Lefu Lan
| |
Collapse
|
49
|
Gao P, Wei Y, Hou S, Lai PM, Liu H, Tai SSC, Tang VYM, Prakash PH, Sze KH, Chen JHK, Sun H, Li X, Kao RYT. SaeR as a novel target for antivirulence therapy against Staphylococcus aureus. Emerg Microbes Infect 2023; 12:2254415. [PMID: 37671453 PMCID: PMC10494732 DOI: 10.1080/22221751.2023.2254415] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 08/27/2023] [Accepted: 08/28/2023] [Indexed: 09/07/2023]
Abstract
Staphylococcus aureus is a major human pathogen responsible for a wide range of clinical infections. SaeRS is one of the two-component systems in S. aureus that modulate multiple virulence factors. Although SaeR is required for S. aureus to develop an infection, inhibitors have not been reported. Using an in vivo knockdown method, we demonstrated that SaeR is targetable for the discovery of antivirulence agent. HR3744 was discovered through a high-throughput screening utilizing a GFP-Lux dual reporter system driven by saeP1 promoter. The antivirulence efficacy of HR3744 was tested using Western blot, Quantitative Polymerase Chain Reaction, leucotoxicity, and haemolysis tests. In electrophoresis mobility shift assay, HR3744 inhibited SaeR-DNA probe binding. WaterLOGSY-NMR test showed HR3744 directly interacted with SaeR's DNA-binding domain. When SaeR was deleted, HR3744 lost its antivirulence property, validating the target specificity. Virtual docking and mutagenesis were used to confirm the target's specificity. When Glu159 was changed to Asn, the bacteria developed resistance to HR3744. A structure-activity relationship study revealed that a molecule with a slight modification did not inhibit SaeR, indicating the selectivity of HR3744. Interestingly, we found that SAV13, an analogue of HR3744, was four times more potent than HR3744 and demonstrated identical antivirulence properties and target specificity. In a mouse bacteraemia model, both HR3744 and SAV13 exhibited in vivo effectiveness. Collectively, we identified the first SaeR inhibitor, which exhibited in vitro and in vivo antivirulence properties, and proved that SaeR could be a novel target for developing antivirulence drugs against S. aureus infections.
Collapse
Affiliation(s)
- Peng Gao
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Yuanxin Wei
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Suying Hou
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Pok-Man Lai
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Han Liu
- Morningside Laboratory for Chemical Biology and Department of Chemistry, The University of Hong Kong, Hong Kong, People’s Republic of China
| | - Sherlock Shing Chiu Tai
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Victor Yat Man Tang
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Pradeep Halebeedu Prakash
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Kong-Hung Sze
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Jonathan Hon Kwan Chen
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Hongzhe Sun
- Morningside Laboratory for Chemical Biology and Department of Chemistry, The University of Hong Kong, Hong Kong, People’s Republic of China
| | - Xuechen Li
- Morningside Laboratory for Chemical Biology and Department of Chemistry, The University of Hong Kong, Hong Kong, People’s Republic of China
| | - Richard Yi-Tsun Kao
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| |
Collapse
|
50
|
Tao Z, Ke K, Shi D, Zhu L. Development of a dual fluorescent reporter system to identify inhibitors of Staphylococcus aureus virulence factors. Appl Environ Microbiol 2023; 89:e0097823. [PMID: 37889047 PMCID: PMC10686081 DOI: 10.1128/aem.00978-23] [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: 06/16/2023] [Accepted: 09/15/2023] [Indexed: 10/28/2023] Open
Abstract
IMPORTANCE Staphylococcus aureus is a formidable pathogen responsible for a wide range of infections, and the emergence of antibiotic-resistant strains has posed significant challenges in treating these infections. In this study, we have established a novel dual reporter system capable of concurrently monitoring the activities of two critical virulence regulators in S. aureus. By incorporating both reporters into a single screening platform, we provide a time- and cost-efficient approach for assessing the activity of compounds against two distinct targets in a single screening round. This innovative dual reporter system presents a promising strategy for the identification of molecules capable of modulating virulence gene expression in S. aureus, potentially expediting the development of antivirulence therapies.
Collapse
Affiliation(s)
- Zhanhua Tao
- Institute of Eco-Environmental Research, Guangxi Academy of Sciences, Nanning, China
- Guangxi Key Laboratory of Marine Natural Products and Combinatorial Biosynthesis Chemistry, Nanning, China
| | - Ke Ke
- Guangxi Academy of Sciences, Nanning, China
| | | | - Libo Zhu
- Guangxi Academy of Sciences, Nanning, China
| |
Collapse
|