1
|
Chen XW, Chen HQ, Wu JH, Wang ZH, Zhou YQ, Tian SQ, Peng B. Isoniazid potentiates tigecycline to kill methicillin-resistant Staphylococcus aureus. Emerg Microbes Infect 2025; 14:2434587. [PMID: 39585340 DOI: 10.1080/22221751.2024.2434587] [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: 11/26/2024]
Abstract
Therapeutic option for treating methicillin-resistant Staphylococcus aureus (MRSA) infection is urgently required since its resistance to a broad spectrum of currently available antibiotics. Here, we report that isoniazid is able to potentiate the killing efficacy of tigecycline to MRSA. The combination of isoniazid and tigecycline reduces the minimal inhibitory concentration of clinic MRSA strains to tigecycline. The killing activity of tigecycline is further confirmed by killing experiments and murine infection model. We further demonstrate the mechanism that isoniazid increases intracellular accumulation of tigecycline by promoting the influx but limiting the efflux of tigecycline through proton motive force. We also show that isoniazid and tigecycline synergize to increase the abundance of isoniazid-NAD adduct, which in turn damage cell membrane, possibly contributing to the disruption of PMF. Whereas phosphatidylethanolamine and cardiolipin are able to abrogate the synergistic effect of isoniazid plus tigecycline. Thus our study provides a new perspective that antibiotics, e.g. isoniazid, once recognized only to target Mycobacterium tuberculosis, can be repurposed as antibiotic adjuvant to tigecycline, expanding our choice of antibiotic-antibiotic combinations in treating bacterial infectious diseases.
Collapse
Affiliation(s)
- Xuan-Wei Chen
- State Key Laboratory of Bio-Control, Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, Guangzhou, People's Republic of China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, People's Republic of China
| | - Hao-Qing Chen
- Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Jia-Han Wu
- State Key Laboratory of Bio-Control, Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Zhi-Han Wang
- State Key Laboratory of Bio-Control, Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yu-Qing Zhou
- State Key Laboratory of Bio-Control, Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Si-Qi Tian
- State Key Laboratory of Bio-Control, Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Bo Peng
- State Key Laboratory of Bio-Control, Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, Guangzhou, People's Republic of China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, People's Republic of China
| |
Collapse
|
2
|
Zhou L, Xiang M, Xin Y, Gao S, Xu K, Zhang J, Lu X, Tang W. Design and synthesis of benzothiazole aryl urea derivatives as potent anti-staphylococcal agents targeting autolysin-mediated peptidoglycan hydrolases. Eur J Med Chem 2025; 292:117715. [PMID: 40324299 DOI: 10.1016/j.ejmech.2025.117715] [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: 02/25/2025] [Revised: 04/28/2025] [Accepted: 04/29/2025] [Indexed: 05/07/2025]
Abstract
Novel benzothiazole aryl ureas were designed and synthesized as anti-MRSA agents targeting peptidoglycan (PG) hydrolases (autolysins). Structural simplification of prior benzothiazole-urea hybrids yielded compounds 4a, 7a and 11a bearing p-CF3 on phenyl ring demonstrating narrow-spectrum activity against Gram-positive bacteria including clinical methicillin-resistant S. aureus (MRSA). The primary autolysin in S. aureus, AtlA, mediates peptidoglycan hydrolase activity critical for bacterial growth, division, and cell wall remodeling. Mechanistic studies revealed that 4a down-regulated autolysin-related genes RNAIII and walR, disrupting peptidoglycan homeostasis. Knockout of atlA (a key autolysin gene) impaired 4a's efficacy, confirming autolysins as critical targets. Docking indicated that 4a binds to AtlA via hydrogen bonds, Pi-Pi, and hydrophobic interactions. In vivo, 4a significantly reduced bacterial load in a murine abdominal infection model, outperforming vancomycin at 10 mg/kg with lower cytotoxicity. Additionally, 4a disrupted MRSA biofilms, suppressed hemolytic toxin production, and alleviated inflammation in infected mice. These findings underscore AtlA as a promising therapeutic target and highlight benzothiazole phenyl urea as a scaffold for developing innovative anti-staphylococcal agents.
Collapse
Affiliation(s)
- Long Zhou
- School of Pharmacy, Department of Medical Microbiology, Anhui Medical University, Hefei, 230032, China
| | - Miaoqing Xiang
- Anhui Province Key Laboratory of Occupational Health, Anhui No. 2 Provincial People's Hospital, Hefei, 230022, China
| | - Yu Xin
- School of Pharmacy, Department of Medical Microbiology, Anhui Medical University, Hefei, 230032, China
| | - Shan Gao
- School of Pharmacy, Department of Medical Microbiology, Anhui Medical University, Hefei, 230032, China
| | - Kehan Xu
- School of Pharmacy, Department of Medical Microbiology, Anhui Medical University, Hefei, 230032, China
| | - Jing Zhang
- Anhui Province Key Laboratory of Occupational Health, Anhui No. 2 Provincial People's Hospital, Hefei, 230022, China
| | - Xueer Lu
- Department of Clinical Laboratory, Hefei Third People's Hospital, Hefei Third Clinical College of Anhui Medical University, Hefei, 230022, China.
| | - Wenjian Tang
- School of Pharmacy, Department of Medical Microbiology, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
3
|
Bhati R, Saifi AP, Sangwan M, Mahur P, Sharma A, Singh AK, Muthukumaran J, Jain M. Computational insights into the inhibition of cell division in Staphylococcus aureus: Towards novel therapeutics. Comput Biol Chem 2025; 117:108391. [PMID: 40037019 DOI: 10.1016/j.compbiolchem.2025.108391] [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: 11/29/2024] [Revised: 02/14/2025] [Accepted: 02/15/2025] [Indexed: 03/06/2025]
Abstract
Staphylococcus aureus, a gram-positive bacterium, causes infective endocarditis, osteoarticular, skin, and respiratory infections. The emergence of multidrug-resistant strains, particularly Methicillin-resistant Staphylococcus aureus (MRSA), has caused a 21-35 % rise in bloodstream infections, complicating treatment strategies. Filamentous temperature-sensitive protein Z (FtsZ), a critical protein involved in bacterial cell division, forms a Z-ring at the division site, making it a key target for novel antibacterial therapies. In this study, 1165 phytochemicals were screened, and three lead molecules namely, Aromadendrin, Leucopelargonidin, and 7-Deacetoxy-7-oxogedunin were identified based on their favorable physicochemical properties, drug-likeness, and estimated binding affinities (- 11.73 kcal/mol, - 10.77 kcal/mol, and - 10.38 kcal/mol, respectively) against FtsZ. 100 ns Molecular dynamics simulations conducted in triplicates confirmed the stability of the FtsZ-ligand complexes.Binding free energy calculations revealed that IMPHY003535 (Leucopelargonidin) exhibited the most favorable binding free energy (-27.25 kcal/mol), followed by 7-Deacetoxy-7-oxogedunin (-15.31 kcal/mol) and Aromadendrin (-13.38 kcal/mol). Leucopelargonidin emerged as the most promising inhibitor, highlighting its potential as a lead compound for developing antibacterial agents targeting FtsZ. These findings demonstrate the significant role of phytochemicals in combating antibiotic resistance and the importance of further optimization, including in vivo studies, to assess their therapeutic potential, which could provide new treatment avenues to overcome bacterial resistance mechanisms.
Collapse
Affiliation(s)
- Roopali Bhati
- Department of Biotechnology, Sharda School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Ayesha Parvez Saifi
- Department of Biotechnology, Sharda School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Manisha Sangwan
- Department of Biotechnology, Sharda School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Pragati Mahur
- Department of Biotechnology, Sharda School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Abhishek Sharma
- Department of Biotechnology, Sharda School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Amit Kumar Singh
- Department of Biotechnology, Sharda School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Jayaraman Muthukumaran
- Department of Biotechnology, Sharda School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Monika Jain
- Department of Biotechnology, Sharda School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh, India.
| |
Collapse
|
4
|
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
|
5
|
Xiong Y, Hao Y, Ding L, Li J, Cao Y, Fan S, Guan F, He L. Hesperidin-loaded self-assembled supramolecular hydrogel based on quaternized chitosan as efficient photothermal antibacterial dressing for MRSA-infected wound healing. Carbohydr Polym 2025; 359:123577. [PMID: 40306783 DOI: 10.1016/j.carbpol.2025.123577] [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: 12/24/2024] [Revised: 03/25/2025] [Accepted: 04/04/2025] [Indexed: 05/02/2025]
Abstract
The management of infected chronic wounds is one of the urgent challenges. Herein, a hesperidin (Hes)-loaded self-assembled supramolecular hydrogel based on quaternized chitosan (CQHP) has been developed as an efficient photothermal antibacterial dressing for MRSA-infected wound healing. Specifically, CQHP hydrogels fabricated through the dynamic noncovalent interactions among CM-β-CD grafted QCS, Hes, proline and Fe3+, exhibited injectable and self-healing behaviors, along with adhesion, antioxidant, hemostatic and protein adsorption performance, satisfying the essential feature as chronic wound dressing. Of note, superior photothermal effect generated from the Hes-Fe3+ has been demonstrated, which endowed the CQHP hydrogels effectively and rapidly eliminate the E. coli, S. aureus and MRSA through photothermal therapy, thereby avoiding the use of antibiotics or photothermal conversion nanomaterials in hydrogels and substantially reducing the biological toxicity. Furtherly, sustained antibacterial performance in the absence of NIR can be achieved through the inherent antibacterial activities of Hes. Importantly, the developed CQHP hydrogels significantly promoted the closure of acute full-thickness scratch wounds, and exhibited remarkable better therapeutic effect on MRSA-infected wound than commercial 3M transparent film, by efficiency and sustained antibacterial activity, reducing inflammation, enhancing angiogenesis and collagen deposition, highlighting its promising application in the MRSA-infected wound healing with high efficiency, quality and security.
Collapse
Affiliation(s)
- Yamin Xiong
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yi Hao
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Linlin Ding
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Jinjin Li
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yuan Cao
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Shuhui Fan
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Fangxia Guan
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Leiliang He
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
6
|
Zou J, Cui W, Deng N, Li C, Yang W, Ye X, Yao F, Zhang T, Xiao J, Ma C, Wu L, Dong D, Chen J, Guo C, Liu A, Wu H. Fate reversal: Exosome-driven macrophage rejuvenation and bacterial-responsive drug release for infection immunotherapy in diabetes. J Control Release 2025; 382:113730. [PMID: 40250625 DOI: 10.1016/j.jconrel.2025.113730] [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: 12/17/2024] [Revised: 03/27/2025] [Accepted: 04/11/2025] [Indexed: 04/20/2025]
Abstract
Superficial surgical site infection (SSI) is a significant risk factor for the development of periprosthetic joint infection (PJI), particularly in diabetic patients. A high-glucose microenvironment is observed to compromise phagocytosis by inducing cellular senescence, which leads to impaired antibacterial immune function. Exosomes derived from umbilical cord stem cells (H-Exos) can reverse the immunosuppressive microenvironment by rejuvenating senescent cells, thereby terminating excessive, persistent, and ineffective inflammatory responses. Thus, a novel exosome-based immunotherapeutic antibacterial strategy to reverse fate is proposed. Vancomycin & lysostaphin-loaded exosomes are incorporated in a customizable microneedle patch (ExoV-ExoL@MN) for controlled release, enabling tailored treatments for diverse clinical scenarios. While rejuvenating macrophage senescent phenotype, the antibiotics encapsulated within exosomes can be responsively released by the hemolysin secreted by bacteria, triggering rapid bacterial killing. Post-infection clearance, they induce a shift from M1 to M2 macrophage polarization, thereby enhancing anti-inflammatory and reparative responses. Furthermore, the components can be mixed on demand and at any time, allowing for real-time customization and fabrication directly at the clinic (fabrication@clinic). This strategy reverses the immunosuppressive microenvironment by rejuvenating senescent macrophages and effectively combats bacterial invasion into deep tissues through bacteria-responsive antibiotic release, providing a promising approach for preventing and treating SSI-induced PJI.
Collapse
Affiliation(s)
- Jiaxuan Zou
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310002, PR China; Orthopedics Research Institute of Zhejiang University, Hangzhou 310002, PR China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University, Hangzhou 310002, PR China; Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou 310002, PR China
| | - Wushi Cui
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310002, PR China; Orthopedics Research Institute of Zhejiang University, Hangzhou 310002, PR China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University, Hangzhou 310002, PR China; Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou 310002, PR China
| | - Niping Deng
- School of Engineering, Westlake University, Hangzhou 310024, PR China
| | - Congsun Li
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310002, PR China; Orthopedics Research Institute of Zhejiang University, Hangzhou 310002, PR China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University, Hangzhou 310002, PR China; Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou 310002, PR China
| | - Weinan Yang
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310002, PR China; Orthopedics Research Institute of Zhejiang University, Hangzhou 310002, PR China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University, Hangzhou 310002, PR China; Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou 310002, PR China
| | - Xiaojun Ye
- Department of Ultrasound, Hangzhou Women's Hospital, Hangzhou 310008, PR China
| | - Feng Yao
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310002, PR China; Orthopedics Research Institute of Zhejiang University, Hangzhou 310002, PR China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University, Hangzhou 310002, PR China; Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou 310002, PR China
| | - Tao Zhang
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310002, PR China; Orthopedics Research Institute of Zhejiang University, Hangzhou 310002, PR China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University, Hangzhou 310002, PR China; Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou 310002, PR China; Department of Orthopedics, Lishui Hospital, Zhejiang University School of Medicine, Lishui 323000, PR China
| | - Jian Xiao
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310002, PR China; Orthopedics Research Institute of Zhejiang University, Hangzhou 310002, PR China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University, Hangzhou 310002, PR China; Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou 310002, PR China; Department of Orthopedics, The First People's Hospital of Jiashan, Jiaxing 314100, PR China
| | - Chiyuan Ma
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310002, PR China; Orthopedics Research Institute of Zhejiang University, Hangzhou 310002, PR China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University, Hangzhou 310002, PR China; Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou 310002, PR China
| | - Lingfeng Wu
- Department of Orthopedics, Lishui Hospital, Zhejiang University School of Medicine, Lishui 323000, PR China
| | - Dahai Dong
- Department of Orthopedics, Suichang County People's Hospital in Zhejiang Province, Lishui 323300, PR China
| | - Jing Chen
- Institute of Medical Sciences, The Second Hospital and Shandong University Center for Orthopaedics, Cheeloo College of Medicine, Shandong University, Jinan 250033, PR China.
| | - Chengchen Guo
- School of Engineering, Westlake University, Hangzhou 310024, PR China.
| | - An Liu
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310002, PR China; Orthopedics Research Institute of Zhejiang University, Hangzhou 310002, PR China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University, Hangzhou 310002, PR China; Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou 310002, PR China.
| | - Haobo Wu
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310002, PR China; Orthopedics Research Institute of Zhejiang University, Hangzhou 310002, PR China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University, Hangzhou 310002, PR China; Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou 310002, PR China.
| |
Collapse
|
7
|
Nagib M, Sayed AM, Korany AH, Abdelkader K, Shari FH, Mackay WG, Rateb ME. Human Defensins: Structure, Function, and Potential as Therapeutic Antimicrobial Agents with Highlights Against SARS CoV-2. Probiotics Antimicrob Proteins 2025; 17:1563-1583. [PMID: 39693007 PMCID: PMC12055905 DOI: 10.1007/s12602-024-10436-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/12/2024] [Indexed: 12/19/2024]
Abstract
The human defensins are a group of cationic antimicrobial peptides that range in size from 2 to 5 kDa and share a common structural motif of six disulphide-linked cysteines. Several naturally occurring human α- and β-defensins have been identified over the past two decades. They have a wide variety of antimicrobial effects, and their potential to avoid the development of resistance to antimicrobial treatment makes them attractive as therapeutic agents. Human defensins have recently been the focus of medical and molecular biology studies due to their promising application in medicine and the pharmaceutical industry. This work aims to provide a comprehensive summary of the current developments of human defensins, including their identification, categorization, molecular features, expression, modes of action, and potential application in medical settings. Current obstacles and future opportunities for using human defensins are also covered. Furthermore, we shed light on the potential of this class as an antiviral agent, particularly against SARS CoV-2, by providing an in silico-based investigation of their plausible mechanisms of action.
Collapse
Affiliation(s)
- Maryam Nagib
- School of Computing Engineering and Physical Sciences, University of the West of Scotland, Paisley, Scotland, PA12BE, UK
| | - Ahmed M Sayed
- Department of Pharmacognosy, College of Pharmacy, Almaaqal University, Basrah, 61014, Iraq
| | - Ahmed H Korany
- Department of Microbiology and Immunology, Faculty of Pharmacy, Nahda University, Beni Suef, 62513, Egypt
| | - Karim Abdelkader
- Department of Microbiology and Immunology, Faculty of Pharmacy, Beni-Suef University, Beni Suef, 62511, Egypt
| | - Falah H Shari
- Department of Clinical Biochemistry, College of Pharmacy, Almaaqal University, Basrah, 61014, Iraq
| | - William G Mackay
- School of Health and Life Sciences, University of the West of Scotland, Blantyre, Glasgow, G72 0LH, UK
| | - Mostafa E Rateb
- School of Computing Engineering and Physical Sciences, University of the West of Scotland, Paisley, Scotland, PA12BE, UK.
| |
Collapse
|
8
|
Menezes Dantas DD, Macêdo NS, Sousa Silveira ZD, Santos Barbosa CRD, Muniz DF, Bezerra AH, Sousa JTD, Alencar GG, Morais Oliveira-Tintino CDD, Tintino SR, da Rocha MN, Marinho ES, Marinho MM, Dos Santos HS, Melo Coutinho HD, Cunha FABD. Naringenin as potentiator of norfloxacin efficacy through inhibition of the NorA efflux pump in Staphylococcus aureus. Microb Pathog 2025; 203:107504. [PMID: 40154849 DOI: 10.1016/j.micpath.2025.107504] [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: 10/07/2024] [Revised: 03/07/2025] [Accepted: 03/21/2025] [Indexed: 04/01/2025]
Abstract
Bacterial resistance is a major challenge in the treatment of Staphylococcus aureus infections, with efflux mechanisms highlighted as reducing the efficacy of antibiotics. In this study, we investigated the potential of naringenin, a natural flavonoid, as an antibacterial agent and efflux pump inhibitor in S. aureus strains 1199 and 1199B. The studies used minimum inhibitory concentration (MIC) assays, ethidium bromide (EtBr) fluorescence emission enhancement assays, cell membrane permeability assays, and in silico molecular docking and ADME prediction assays. Naringenin showed no relevant antibacterial activity (MIC ≥1024 μg/mL). However, it potentiated the effect of norfloxacin and EtBr, reducing their MICs and increasing the fluorescence emission of EtBr, suggesting a possible inhibition of the NorA efflux pump. Bacterial membrane permeability was not significantly affected. Molecular docking assays indicated that naringenin interacts with the chlorpromazine binding site and has more favorable affinity energy than the chlorpromazine-NorA complex. ADME prediction showed favorable physicochemical properties, good oral absorption, metabolic stability and central nervous system safety. Therefore, naringenin demonstrates the potential to reverse the efficacy of norfloxacin in S. aureus by associating with efflux inhibition through effective interactions with the NorA protein, suggesting its therapeutic potential against bacterial resistance.
Collapse
Affiliation(s)
- Debora de Menezes Dantas
- Postgraduate Program in Biological Chemistry, Department of Biological Chemistry, Regional University of Cariri - URCA, Crato, CE, Brazil; Semi-arid Bioprospecting Laboratory and Alternative Methods, Pimenta Campus, Regional University of Cariri - URCA, Crato, CE, Brazil.
| | - Nair Silva Macêdo
- Postgraduate Program in Biological Chemistry, Department of Biological Chemistry, Regional University of Cariri - URCA, Crato, CE, Brazil; Semi-arid Bioprospecting Laboratory and Alternative Methods, Pimenta Campus, Regional University of Cariri - URCA, Crato, CE, Brazil
| | - Zildene de Sousa Silveira
- Graduate Program in Biological Sciences- PPGCB, Federal University of Pernambuco - UFPE, Recife, Pernambuco, Brazil; Semi-arid Bioprospecting Laboratory and Alternative Methods, Pimenta Campus, Regional University of Cariri - URCA, Crato, CE, Brazil
| | - Cristina Rodrigues Dos Santos Barbosa
- Postdoctoral Intern at the Semiarid Bioprospecting Laboratory (LABSEMA), Regional University of Cariri - URCA, Crato, CE, Brazil; Semi-arid Bioprospecting Laboratory and Alternative Methods, Pimenta Campus, Regional University of Cariri - URCA, Crato, CE, Brazil
| | - Débora Feitosa Muniz
- Postgraduate Program in Biological Chemistry, Department of Biological Chemistry, Regional University of Cariri - URCA, Crato, CE, Brazil; Semi-arid Bioprospecting Laboratory and Alternative Methods, Pimenta Campus, Regional University of Cariri - URCA, Crato, CE, Brazil
| | - Antônio Henrique Bezerra
- Postgraduate Program in Biological Chemistry, Department of Biological Chemistry, Regional University of Cariri - URCA, Crato, CE, Brazil; Semi-arid Bioprospecting Laboratory and Alternative Methods, Pimenta Campus, Regional University of Cariri - URCA, Crato, CE, Brazil
| | - Josivânia Teixeira de Sousa
- Semi-arid Bioprospecting Laboratory and Alternative Methods, Pimenta Campus, Regional University of Cariri - URCA, Crato, CE, Brazil
| | - Gabriel Gonçalves Alencar
- Laboratory of Microbiology and Molecular Biology (LMBM), Regional University of Cariri - URCA, Crato, 63105-000, CE, Brazil
| | - Cícera Datiane de Morais Oliveira-Tintino
- Laboratory of Microbiology and Molecular Biology (LMBM), Regional University of Cariri - URCA, Crato, 63105-000, CE, Brazil; Postdoctoral Intern at the Laboratory of Microbiology and Molecular Biology (LMBM), Regional University of Cariri - URCA, Crato, CE, Brazil
| | - Saulo Relison Tintino
- Laboratory of Microbiology and Molecular Biology (LMBM), Regional University of Cariri - URCA, Crato, 63105-000, CE, Brazil; Biological Chemistry Department, Pimenta Campus, Regional University of Cariri - URCA, Crato, CE, Brazil
| | - Matheus Nunes da Rocha
- Ceará State University, Postgraduate Program in Natural Sciences, Laboratory of Chemistry of Natural Products, Fortaleza, CE, Brazil
| | - Emmanuel Silva Marinho
- Ceará State University, Postgraduate Program in Natural Sciences, Laboratory of Chemistry of Natural Products, Fortaleza, CE, Brazil
| | | | - Hélcio Silva Dos Santos
- Biological Chemistry Department, Pimenta Campus, Regional University of Cariri - URCA, Crato, CE, Brazil
| | - Henrique Douglas Melo Coutinho
- Laboratory of Microbiology and Molecular Biology (LMBM), Regional University of Cariri - URCA, Crato, 63105-000, CE, Brazil; Biological Chemistry Department, Pimenta Campus, Regional University of Cariri - URCA, Crato, CE, Brazil
| | - Francisco Assis Bezerra da Cunha
- Semi-arid Bioprospecting Laboratory and Alternative Methods, Pimenta Campus, Regional University of Cariri - URCA, Crato, CE, Brazil; Biological Chemistry Department, Pimenta Campus, Regional University of Cariri - URCA, Crato, CE, Brazil
| |
Collapse
|
9
|
Vera-Yunca D, Matias C, Vingsbo Lundberg C, Friberg LE. Model-based translation of the PKPD-relationship for linezolid and vancomycin on methicillin-resistant Staphylococcus aureus: from in vitro time-kill experiments to a mouse pneumonia model. J Antimicrob Chemother 2025:dkaf140. [PMID: 40343749 DOI: 10.1093/jac/dkaf140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Accepted: 04/23/2025] [Indexed: 05/11/2025] Open
Abstract
OBJECTIVES MRSA is one of the main pathogens that cause nosocomial pneumonia. Based on longitudinal in vitro and in vivo data, a pharmacokinetic-pharmacodynamic (PKPD) model was built to quantify the effect of two control antibiotics (LZD and VAN) for Gram-positive bacteria in a standardized mouse pneumonia model. METHODS The PKPD model was developed for data generated on the MRSA strain 160 079 in static in vitro time-kill experiments and thereafter adjusted to fit data from lungs of neutropenic mice administered with single or multiple doses of LZD (0.5-40 mg/kg) or VAN (1-40 mg/kg). Simulations with human PK were run to predict antibacterial response in patients. RESULTS Bacterial regrowth observed in vitro when exposed to VAN concentrations was described by an adaptive resistance model. The selected MRSA isolate showed good virulence in the mouse pneumonia model. Bacterial load in lungs decreased up to 2-log with respect to control mice after LZD and VAN treatment. A 70%-75% lower killing rate was estimated for the in vivo data when compared with in vitro. Simulations displayed bacterial stasis at 24 h for patients infected with bacteria with MICs below the clinical breakpoint for both drugs after administering standard-of-care dosing regimens. CONCLUSIONS A translational workflow allowed us to build a PKPD model with both in vitro and in vivo data that characterized bacterial dynamics following LZD and VAN exposure, showing that this approach can inform the development of antibiotics. We also showcased the first successful use of the standardized mouse pneumonia model for Gram-positive bacteria.
Collapse
Affiliation(s)
| | - Carina Matias
- Bacteria, Parasites & Fungi, Statens Serum Institut, Copenhagen, Denmark
| | | | - Lena E Friberg
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
| |
Collapse
|
10
|
Xu H, He D, Tao H. A biomimetic nano-NET strategy for the treatment of MRSA-related implant-associated infection. RSC Adv 2025; 15:14821-14837. [PMID: 40337221 PMCID: PMC12057620 DOI: 10.1039/d5ra00367a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 04/24/2025] [Indexed: 05/09/2025] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) has spread across diverse global environments, and MRSA-related infection is a major threat to public health. Implant-associated infection (IAI) caused by MRSA remains a tough global clinical problem. Conventional antibiotic therapy has limited efficacy in treating MRSA-related IAI, and antibiotic abuse has resulted in the emergence of multidrug-resistant bacteria. Hence, there is a necessity to explore more effective approaches to deal with MRSA-related IAI. Herein, inspired by neutrophil extracellular traps (NETs) released by neutrophils to kill microorganisms, this study proposes a novel biomimetic nano-NET strategy using an epsilon-poly-l-lysine-coated CuO2 nanoplatform, denoted as PCPNAs. The function-adaptive nanoplatform exhibited excellent Fenton-like performance, including robust ROS generation and GSH scavenging ability. PCPNAs showed >90% cell viability in mammalian cells and reduced bacterial burden by 7.65 log10 CFU in vitro. Moreover, the positively charged PCPNAs could easily bind to negatively charged MRSA cells through charge-coupling and simultaneously exerted a trapping effect on MRSA cells. Notably, PCPNAs self-assembled into web-like structures to physically trap and kill biofilm bacteria, achieving 99.58% biofilm eradication. Furthermore, PCPNAs showed satisfactory biocompatibility in vivo and displayed ideal anti-bacterial and anti-inflammatory effects in a mouse model with implant-associated infection. With further development and optimization, the biomimetic nano-NET strategy based on PCPNAs provides a new therapeutic option for the treatment of MRSA-related implant-associated infection.
Collapse
Affiliation(s)
- Huan Xu
- Department of Orthopedics Surgery, Lishui Hospital, Zhejiang University School of Medicine No. 289, Kuocang Road Lishui Zhejiang 323000 China
| | - Dengwei He
- Department of Orthopedics Surgery, Lishui Hospital, Zhejiang University School of Medicine No. 289, Kuocang Road Lishui Zhejiang 323000 China
| | - Huimin Tao
- Department of Orthopedics Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine No. 88, Jiefang Road Hangzhou Zhejiang 310009 China
| |
Collapse
|
11
|
Devkar HU, Juyal K, Thakur NL, Kaur P, Parmar K, Pullapanthula R, Narayanan S. Antimicrobial Potential of Marine Sponge-Associated Bacillus velezensis and Stutzerimonas stutzeri from the Indian Coast: A Genome Mining and Metabolite Profiling Approach. Curr Microbiol 2025; 82:280. [PMID: 40327113 DOI: 10.1007/s00284-025-04262-6] [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: 12/01/2024] [Accepted: 04/25/2025] [Indexed: 05/07/2025]
Abstract
Antimicrobial resistance (AMR) is one of the leading health crises worldwide that demands new antimicrobials to enter the clinical pipeline. Marine sponges are a rich source of promising bioactive compounds. Due to their sessile nature and filter-feeding lifestyle, sponges are prone to attack by competitors, predators, and pathogens. To combat these threats, they produce a diverse array of bioactive compounds. Notably, the microbial communities residing within the sponges make many of these beneficial compounds. Twenty-one bacterial isolates from various marine sponges from the Indian coast were selected for this study. The bacterial isolates were fermented to obtain crude extracts, which were then screened against critical bacterial pathogens. Based on the MIC (minimum inhibitory concentration) results, two isolates, Bacillus velezensis NIO_002 and Stutzerimonas stutzeri NIO_003 showing good activity, were characterized by morphological, biochemical, and molecular methods. Genome mining predicted multiple antibiotic biosynthetic gene clusters, most of which showed a high degree of similarity to known gene clusters, and some with low or no similarity which may be indicative of novel gene clusters. LC-MS (liquid chromatography-mass spectrometry) data revealed the putative presence of certain antibacterial compounds previously reported in the literature. To our knowledge, this is the first study to report the antimicrobial activity of marine sponge-associated Bacillus velezensis and Stutzerimonas stutzeri strains characterized by whole genome sequencing, thereby indicating the novelty of our strains. This study emphasizes the potential of our bacterial isolates for further development as a source of promising antibiotics to address the escalating challenge of drug-resistant pathogens.
Collapse
Affiliation(s)
- Heena U Devkar
- CSIR- National Institute of Oceanography, Dona Paula, 403004, Goa, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Kartik Juyal
- CSIR- National Institute of Oceanography, Dona Paula, 403004, Goa, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Narsinh L Thakur
- CSIR- National Institute of Oceanography, Dona Paula, 403004, Goa, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| | - Parvinder Kaur
- Foundation for Neglected Disease Research, Bangalore, 561203, Karnataka, India
| | - Keyur Parmar
- National Institute of Pharmaceutical Education and Research, Guwahati, 781101, Assam, India
| | | | - Shridhar Narayanan
- Foundation for Neglected Disease Research, Bangalore, 561203, Karnataka, India
| |
Collapse
|
12
|
Korotetskiy IS, Shilov SV, Kuznetsova TV, Zubenko N, Ivanova L, Reva ON. Epigenetic background of lineage-specific gene expression landscapes of four Staphylococcus aureus hospital isolates. PLoS One 2025; 20:e0322006. [PMID: 40323905 PMCID: PMC12052166 DOI: 10.1371/journal.pone.0322006] [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: 11/27/2024] [Accepted: 03/16/2025] [Indexed: 05/07/2025] Open
Abstract
Bacteria with similar genomes can exhibit different phenotypes due to alternative gene expression patterns. In this study, we analysed four antibiotic-resistant Staphylococcus aureus hospital isolates using transcriptomics, PacBio genome sequencing, and methylomics analyses. Transcriptomic data were obtained from cultures exposed to gentamicin, the iodine-alanine complex CC-196, and their combination. We observed strain-specific expression patterns of core and accessory genes that remained stable under antimicrobial stress - a phenomenon we term the Clonal Gene Expression Stability (CGES) that is the main discovery of the paper. An involvement of epigenetic mechanisms in stabilization of the CGES was hypothesized and statistically verified. Canonical methylation patterns controlled by type I restriction-modification systems accounted for ~ 10% of epigenetically modified adenine residues, whereas multiple non-canonically modified adenines were distributed sporadically due to imperfect DNA targeting by methyltransferases. Protein-coding sequences were characterized by a significantly lower frequency of modified nucleotides. Epigenetic modifications near transcription start codons showed a statistically significant negative association with gene expression levels. While the role of epigenetic modifications in gene regulation remains debatable, variations in non-canonical modification patterns may serve as markers of CGES.
Collapse
Affiliation(s)
- Ilya S. Korotetskiy
- Virology laboratory, JSC Scientific Center for Anti-Infectious Drugs, Almaty, Kazakhstan
- LLC International Engineering and Technological University, Almaty, Kazakhstan
- LLP Research and Production Association Kazpharmacom, Almaty, Kazakhstan
| | - Sergey V. Shilov
- Virology laboratory, JSC Scientific Center for Anti-Infectious Drugs, Almaty, Kazakhstan
| | - Tatyana V. Kuznetsova
- Virology laboratory, JSC Scientific Center for Anti-Infectious Drugs, Almaty, Kazakhstan
| | - Natalya Zubenko
- Virology laboratory, JSC Scientific Center for Anti-Infectious Drugs, Almaty, Kazakhstan
| | - Lyudmila Ivanova
- Virology laboratory, JSC Scientific Center for Anti-Infectious Drugs, Almaty, Kazakhstan
| | - Oleg N. Reva
- Centre for Bioinformatics and Computational Biology, Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
13
|
Zhao X, Wang S, Wang J, Bai X, Yang Z, Guo H, Wu L, Liu C, Yu X, Du J. Construction of a thiophene-based conjugated polymer/TP-PCN S-scheme to enhance visible-light-driven photocatalytic activity: Promotion of wound healing in super-resistant bacterial infections. JOURNAL OF HAZARDOUS MATERIALS 2025; 488:137429. [PMID: 39892129 DOI: 10.1016/j.jhazmat.2025.137429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/18/2025] [Accepted: 01/27/2025] [Indexed: 02/03/2025]
Abstract
S-scheme heterojunctions have garnered significant attention in the field of photocatalytic antimicrobials due to their superior charge separation efficiency and higher redox capacity. In this study, an innovative linear conjugated polymer (PCO) was combined with fragmented carbon nitride (TP-PCN) to create PCO/TP-PCN organic-organic S-scheme heterojunctions, which markedly enhanced the photocatalytic antimicrobial performance. The composite (PCO-7/TP-PCN) demonstrated the ability to combat bacterial infections under visible light irradiation, effectively eradicating approximately 2.16 × 107 cfu/ml MRSA within 6 min. This exceptional photocatalytic performance can be attributed to the successful formation of an S-scheme heterojunction between PCO and TP-PCN, as well as the interaction of surface functional groups of PCO-7/TP-PCN with bacteria. Results from UV-Vis-NIR DRS and in situ-XPS experiments indicated a significant enhancement in carrier transport rate through the establishment of a built-in electric field and energy band bending at the interface. In vitro and in vivo experiments further demonstrated that PCO-7/TP-PCN not only exhibited potent antimicrobial activity under visible light irradiation but also promoted angiogenesis to inhibit inflammatory responses. Therefore, it can be concluded that this organic-organic S-scheme heterojunction photocatalyst holds great potential for development as a promising new generation of efficient antimicrobial materials, which could aid in preventing bacterial infection of wounds and ensuring effective wound healing.
Collapse
Affiliation(s)
- Xin Zhao
- Key Laboratory of Preparation and Application of Environmental Friendly Materials, Ministry of Education, Jilin Normal University, Changchun 130103, PR China; College of Chemistry, Jilin Normal University, Siping 136000, PR China
| | - Simiao Wang
- College of Chemistry, Jilin Normal University, Siping 136000, PR China
| | - Jiayu Wang
- Department of Biological Science, School of Life Science, Jilin Normal University, Siping, Jilin 136000, PR China
| | - Xinyue Bai
- College of Chemistry, Jilin Normal University, Siping 136000, PR China
| | - Zhongwei Yang
- Institute for Advanced Interdisciplinary Research (iAIR), University of Jinan, Jinan 250022, PR China
| | - Haiyong Guo
- Department of Biological Science, School of Life Science, Jilin Normal University, Siping, Jilin 136000, PR China
| | - Lina Wu
- College of Chemistry, Baicheng Normal University, Baicheng 137000, PR China.
| | - Chunbo Liu
- Key Laboratory of Preparation and Application of Environmental Friendly Materials, Ministry of Education, Jilin Normal University, Changchun 130103, PR China; College of Engineering, Jilin Normal University, Siping 136000, PR China.
| | - Xin Yu
- Institute for Advanced Interdisciplinary Research (iAIR), University of Jinan, Jinan 250022, PR China.
| | - Juan Du
- Key Laboratory of Preparation and Application of Environmental Friendly Materials, Ministry of Education, Jilin Normal University, Changchun 130103, PR China; College of Chemistry, Jilin Normal University, Siping 136000, PR China.
| |
Collapse
|
14
|
Fan Z, Liang C, Zhang J, Li Y, Tan L, Deng H, Wang P, Wang J, Kang J, Zhu Y, Fu H, Tao J. Multimodal Synergistic Strategies for Diabetic Wound Healing Using Glucose Oxidase Nanocomposites: Therapeutic Mechanisms and Nanomaterial Design. Int J Nanomedicine 2025; 20:5727-5762. [PMID: 40337147 PMCID: PMC12056316 DOI: 10.2147/ijn.s515057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 04/21/2025] [Indexed: 05/09/2025] Open
Abstract
Diabetic wounds (DWs) are characterized by high blood glucose levels, and one of the primary strategies for regulating blood glucose is the use of glucose oxidase (GOx). This enzyme catalyzes the oxidation of glucose to produce D-gluconic acid, consuming oxygen and generating hydrogen peroxide (H₂O₂) in the process. In DWs, this reaction not only effectively reduces glucose concentrations at the wound site but also provides an antibacterial effect through the release of H₂O₂. Based on this principle, combining glucose oxidase with other therapeutic approaches to develop multimodal wound treatment strategies has garnered significant research attention. Additionally, the abundance of binding sites on the GOx molecular surface enables the construction of multifunctional GOx-based nanocomposites. This review uniquely integrates emerging nanomaterial designs with cascade therapeutic strategies, offering insights into overcoming challenges in diabetic wound healing. Recently, multifunctional nanocomposites have gained attention for integrating multiple therapeutic modalities, relying on cascade mechanisms of multimodal synergistic therapies to tackle complex challenges in DWs treatment. However, there is currently no systematic review that comprehensively elaborates on the construction of these nanocomposites and the specific applications of multimodal treatment strategies in DWs healing. To fill this gap in the field, this review provides a comprehensive overview of these nanomaterials, starting with a systematic exploration of cascade and synergistic therapeutic mechanisms centered on GOx-catalyzed reactions. It highlights applications in photothermal therapy (PTT), photodynamic therapy (PDT), and gas therapy (GT), summarizes the design of nanocarriers, and discusses challenges in DWs healing and future development directions. The findings discussed provide a pathway for the development of clinically viable, cost-effective therapies for chronic wounds.
Collapse
Affiliation(s)
- Zaiwei Fan
- Department of Orthopedics, The Second Affiliated Hospital, Jiangxi Medical College Nanchang University, Nanchang, Jiangxi, 330006, People’s Republic of China
| | - Chengzhi Liang
- Department of Orthopedics, The Second Affiliated Hospital, Jiangxi Medical College Nanchang University, Nanchang, Jiangxi, 330006, People’s Republic of China
| | - Jiayu Zhang
- Department of ECG, Sir Run Run Shaw Hospital, Sir Run Run Shaw Institute of Clinical Medicine of Zhejiang University, Hangzhou, People’s Republic of China
| | - Yiming Li
- School of Traditional Chinese Medicine, Nanchang Medical College, Nanchang, Jiangxi, 330052, People’s Republic of China
| | - Lihua Tan
- Department of Orthopedics, The Second Affiliated Hospital, Jiangxi Medical College Nanchang University, Nanchang, Jiangxi, 330006, People’s Republic of China
| | - Hui Deng
- Department of Orthopedics, The Second Affiliated Hospital, Jiangxi Medical College Nanchang University, Nanchang, Jiangxi, 330006, People’s Republic of China
| | - Pinkai Wang
- Department of Orthopedics, The Second Affiliated Hospital, Jiangxi Medical College Nanchang University, Nanchang, Jiangxi, 330006, People’s Republic of China
| | - Jialiang Wang
- Department of Orthopedics, The Second Affiliated Hospital, Jiangxi Medical College Nanchang University, Nanchang, Jiangxi, 330006, People’s Republic of China
| | - Jiawei Kang
- Department of Orthopedics, The Second Affiliated Hospital, Jiangxi Medical College Nanchang University, Nanchang, Jiangxi, 330006, People’s Republic of China
| | - Yudan Zhu
- Department of Orthopedics, The Second Affiliated Hospital, Jiangxi Medical College Nanchang University, Nanchang, Jiangxi, 330006, People’s Republic of China
| | - Hang Fu
- Department of Orthopedics, The Second Affiliated Hospital, Jiangxi Medical College Nanchang University, Nanchang, Jiangxi, 330006, People’s Republic of China
| | - Jun Tao
- Department of Orthopedics, The Second Affiliated Hospital, Jiangxi Medical College Nanchang University, Nanchang, Jiangxi, 330006, People’s Republic of China
| |
Collapse
|
15
|
Jiang Y, Wang Y, Bai Y, Yuan L, Dai QB, Zhu Q, Zhao R, Liu MF, Liu P. Genomic and phenotypic adaptations of methicillin resistant Staphylococcus aureus during vancomycin therapy. Sci Rep 2025; 15:15346. [PMID: 40316685 PMCID: PMC12048576 DOI: 10.1038/s41598-025-99639-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Accepted: 04/22/2025] [Indexed: 05/04/2025] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) poses a significant global health challenge, particularly associated with serious infections such as bacteremia. Lipoglycopeptide antibiotics, including vancomycin, dalbavancin, and daptomycin, are critical in MRSA treatment. In this study, we analyzed two MRSA isolates (XF1 and XF2) from a bacteremia patient treated with vancomycin. Antimicrobial susceptibility testing revealed that XF1 was sensitive to vancomycin, dalbavancin, and daptomycin, whereas XF2 exhibited 8- to 16-fold higher minimum inhibitory concentrations for these antibiotics, alongside a 4- to 8-fold reduction in resistance to β-lactam antibiotics, demonstrating the "β-lactam seesaw effect". Whole-genome sequencing confirmed their isogenic nature (ST59-SCCmecIV-t172), identifying seven mutations in XF2, including those in walK (G223S), vraR (D88Y), clpX (P64L), and ltaS (L62P), as well as a frameshift mutation in mgt (S39fs), likely contributing to resistance. Transmission electron microscopy and autolysis assays demonstrated that XF2 had a thicker cell wall and a slower autolysis rate compared to XF1. Phenotypic analysis showed that XF2 exhibited reduced growth rate, diminished virulence, and enhanced biofilm formation compared to XF1. Gene expression analysis supported these findings, revealing significant alterations in pathways related to cell wall metabolism, autolysis, and virulence regulation. These adaptations highlight the genomic and phenotypic plasticity of MRSA under antibiotic pressure, enabling resistance and persistence. This study underscores the urgent need for enhanced surveillance and alternative therapeutic strategies, including exploiting the β-lactam seesaw effect, to combat lipoglycopeptide-nonsusceptible MRSA.
Collapse
Affiliation(s)
- Yiyue Jiang
- Department of Clinical Laboratory, Medical Center of Burn plastic and wound repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Ying Wang
- Department of Clinical Laboratory, Medical Center of Burn plastic and wound repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
- School of Public Health, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - YunXue Bai
- Department of Clinical Laboratory, Medical Center of Burn plastic and wound repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Lei Yuan
- Department of Clinical Laboratory, Medical Center of Burn plastic and wound repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Qian-Bin Dai
- Department of Clinical Laboratory, Medical Center of Burn plastic and wound repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Qing Zhu
- Department of Clinical Laboratory, Medical Center of Burn plastic and wound repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Rui Zhao
- Department of Clinical Laboratory, Medical Center of Burn plastic and wound repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Mei-Fang Liu
- Department of Clinical Laboratory, Medical Center of Burn plastic and wound repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Peng Liu
- Department of Clinical Laboratory, Medical Center of Burn plastic and wound repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
16
|
Wang R, Zhao C, Guo D, Wang Y, Sun L, Liu X, Sun Y, Liu D, Guan J, Wang L, Wang B. Disarming the Pathogenicity of Methicillin-Resistant Staphylococcus aureus via Osmundacetone-Mediated Inhibition of Sortase A. Microb Biotechnol 2025; 18:e70119. [PMID: 40358044 DOI: 10.1111/1751-7915.70119] [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: 10/10/2024] [Revised: 01/07/2025] [Accepted: 02/13/2025] [Indexed: 05/15/2025] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is a major global health threat due to its resistance to multiple antibiotics, making conventional treatments ineffective. The rise in antibiotic resistance highlights the urgent need for new therapies. Sortase A (SrtA), a key virulence factor in Staphylococcus aureus (S. aureus), facilitates bacterial adhesion and infection by anchoring surface proteins to host cells, making it a promising drug target. In this study, we investigated the potential of osmundacetone (OSC), a natural compound from Osmundae Rhizoma, as an SrtA inhibitor. Using fluorescence resonance energy transfer (FRET), OSC was found to inhibit SrtA with an IC50 of 1.29 μg/mL (7.24 μM). Further in vitro assays confirmed the effectiveness of OSC in inhibiting SrtA-mediated bacterial adhesion, invasion and biofilm formation. Fluorescence quenching and molecular docking pinpointed the binding site of OSC on SrtA. In vivo, OSC improved survival rates in MRSA-infected mice and Galleria mellonella (G. mellonella) while reducing bacterial loads in infected tissues. These results suggest OSC as a promising candidate for anti-MRSA therapies.
Collapse
Affiliation(s)
- Rong Wang
- Changchun University of Chinese Medicine, Changchun, China
| | - Chunhui Zhao
- Changchun University of Chinese Medicine, Changchun, China
| | - Dongbin Guo
- Changchun University of Chinese Medicine, Changchun, China
| | - Yueying Wang
- Changchun University of Chinese Medicine, Changchun, China
| | - Luanbiao Sun
- China-Japan Union Hospital of Jilin University, Jilin University, Changchun, China
| | - Xinyao Liu
- Changchun University of Chinese Medicine, Changchun, China
| | - Yun Sun
- Changchun University of Chinese Medicine, Changchun, China
| | - Da Liu
- Changchun University of Chinese Medicine, Changchun, China
| | - Jiyu Guan
- State Key·Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Jilin University Changchun, China
| | - Li Wang
- Changchun University of Chinese Medicine, Changchun, China
| | - Bingmei Wang
- Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
17
|
Luangrath MA, Chegondi M, Badheka A. Outcome of extracorporeal membrane oxygenation support among children with methicillin-resistant Staphylococcus aureus infection: A single-center experience. Perfusion 2025; 40:941-946. [PMID: 39097819 DOI: 10.1177/02676591241268706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/05/2024]
Abstract
Introduction: The use of extracorporeal membrane oxygenation (ECMO) in children continues to increase nationally, including patients with methicillin-resistant Staphylococcus aureus (MRSA) infection. Survival of pediatric patients with MRSA sepsis has not improved over the last 20 years. We sought to review our institutional experience and outcomes of ECMO support among children with MRSA infection.Methods: Children aged 0-19 years who received ECMO support from October 2014 to June 2021 were reviewed retrospectively. Patients with laboratory confirmed MRSA infections were identified.Results: Out of 88 unique pediatric patients requiring ECMO support, eight patients had documented MRSA infections. The duration of mechanical ventilation prior to ECMO initiation was an average of seven days (range 0.7 to 21.8 days). The median ECMO duration was 648.1 h (range 15.5 to 1580.5 h). Five patients were successfully decannulated; however, only two patients survived to discharge. The two surviving patients were both cannulated via VV-ECMO. Mechanical ventilation prior to ECMO was 4.5 and 21.8 days in these cases with run durations of 18.9 and 29.9 days, respectively.Conclusions: Our institutional survival of patients with MRSA on ECMO is lower than what has been reported in recent database studies, but notably, 62.5% were successfully decannulated. While both surviving patients were supported with VV-ECMO, there was no other clear trend in factors that contributed to survival. MRSA continues to be a source of significant morbidity and mortality among pediatric patients. On-going investigation of outcomes and factors contributing to survival in patients with MRSA infection on ECMO is warranted.
Collapse
Affiliation(s)
- Mitchell A Luangrath
- Division of Critical Care, Stead Family Department of Pediatrics, University of Iowa Health Care, Iowa City, IA, USA
| | - Madhuradhar Chegondi
- Division of Critical Care, Stead Family Department of Pediatrics, University of Iowa Health Care, Iowa City, IA, USA
| | - Aditya Badheka
- Division of Critical Care, Stead Family Department of Pediatrics, University of Iowa Health Care, Iowa City, IA, USA
| |
Collapse
|
18
|
Souza HCAD, Panzenhagen P, Dos Santos AMP, Portes AB, Fidelis J, Conte-Junior CA. Unravelling the advances of CRISPR-Cas9 as a precise antimicrobial therapy: A systematic review. J Glob Antimicrob Resist 2025; 42:51-60. [PMID: 39954947 DOI: 10.1016/j.jgar.2025.02.002] [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: 10/17/2024] [Revised: 01/21/2025] [Accepted: 02/06/2025] [Indexed: 02/17/2025] Open
Abstract
Antimicrobial resistance is a critical public health threat, compromising treatment effectiveness. The spread of resistant pathogens, facilitated by genetic variability and horizontal gene transfer, primarily through plasmids, poses significant challenges to health systems. OBJECTIVE This review explores the potential of Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) technology and Cas9 nucleases in combating antimicrobial resistance. METHODS The literature review followed the PRISMA guidelines using PubMed, Embase, and Scopus databases until July 2023. RESULTS The Enterobacterales family, particularly Escherichia coli, was the main focus. The resistance genes targeted were mainly associated with β-lactam antibiotics, specifically bla genes, and colistin resistance linked to the mcr-1 gene. Plasmid vectors have been the primary delivery method for the CRISPR-Cas9 system, with conjugative plasmids resensitizing bacterial strains to various antimicrobials. Other delivery methods included electroporation, phage-mediated delivery, and nanoparticles. The efficacy of the CRISPR-Cas9 system in resensitizing bacterial strains ranged from 4.7% to 100%. CONCLUSIONS Despite challenges in delivery strategies and clinical application, studies integrating nanotechnology present promising approaches to overcome these limitations. This review highlights new perspectives for the clinical use of CRISPR-Cas9 as a specific and efficient antimicrobial agent, potentially replacing traditional broad-spectrum antimicrobials in the future.
Collapse
Affiliation(s)
- Hannay Crystynah Almeida de Souza
- Center for Food Analysis (NAL), Technological Development Support Laboratory (LADETEC), Federal University of Rio de Janeiro (UFRJ), Cidade Universitária, Rio de Janeiro, Brazil; Department of Biochemistry, Laboratory of Advanced Analysis in Biochemistry and Molecular Biology (LAABBM), Federal University of Rio de Janeiro (UFRJ), Cidade Universitária, Rio de Janeiro, Brazil; Graduate Program in Biochemistry (PPGBq), Institute of Chemistry (IQ), Federal University of Rio de Janeiro (UFRJ), Cidade Universitária, Rio de Janeiro, RJ, Brazil
| | - Pedro Panzenhagen
- Center for Food Analysis (NAL), Technological Development Support Laboratory (LADETEC), Federal University of Rio de Janeiro (UFRJ), Cidade Universitária, Rio de Janeiro, Brazil; Department of Biochemistry, Laboratory of Advanced Analysis in Biochemistry and Molecular Biology (LAABBM), Federal University of Rio de Janeiro (UFRJ), Cidade Universitária, Rio de Janeiro, Brazil; Graduate Program in Biochemistry (PPGBq), Institute of Chemistry (IQ), Federal University of Rio de Janeiro (UFRJ), Cidade Universitária, Rio de Janeiro, RJ, Brazil; Analytical and Molecular Laboratory Center (CLAn), Institute of Chemistry (IQ), Federal University of Rio de Janeiro (UFRJ), Cidade Universitária, Rio de Janeiro, Brazil.
| | - Anamaria Mota Pereira Dos Santos
- Center for Food Analysis (NAL), Technological Development Support Laboratory (LADETEC), Federal University of Rio de Janeiro (UFRJ), Cidade Universitária, Rio de Janeiro, Brazil; Department of Biochemistry, Laboratory of Advanced Analysis in Biochemistry and Molecular Biology (LAABBM), Federal University of Rio de Janeiro (UFRJ), Cidade Universitária, Rio de Janeiro, Brazil; Graduate Program in Veterinary Hygiene (PGHIGVET), Faculty of Veterinary Medicine, Fluminense Federal University (UFF), Niterói, RJ, Brazil
| | - Ana Beatriz Portes
- Center for Food Analysis (NAL), Technological Development Support Laboratory (LADETEC), Federal University of Rio de Janeiro (UFRJ), Cidade Universitária, Rio de Janeiro, Brazil; Department of Biochemistry, Laboratory of Advanced Analysis in Biochemistry and Molecular Biology (LAABBM), Federal University of Rio de Janeiro (UFRJ), Cidade Universitária, Rio de Janeiro, Brazil; Department of General Microbiology, Laboratory of Microorganism Structure, Institute of Microbiology Paulo de Góes (IMPG), Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Juliana Fidelis
- Center for Food Analysis (NAL), Technological Development Support Laboratory (LADETEC), Federal University of Rio de Janeiro (UFRJ), Cidade Universitária, Rio de Janeiro, Brazil; Department of Biochemistry, Laboratory of Advanced Analysis in Biochemistry and Molecular Biology (LAABBM), Federal University of Rio de Janeiro (UFRJ), Cidade Universitária, Rio de Janeiro, Brazil; Graduate Program in Food Science, Institute of Chemistry, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Carlos Adam Conte-Junior
- Center for Food Analysis (NAL), Technological Development Support Laboratory (LADETEC), Federal University of Rio de Janeiro (UFRJ), Cidade Universitária, Rio de Janeiro, Brazil; Department of Biochemistry, Laboratory of Advanced Analysis in Biochemistry and Molecular Biology (LAABBM), Federal University of Rio de Janeiro (UFRJ), Cidade Universitária, Rio de Janeiro, Brazil; Graduate Program in Biochemistry (PPGBq), Institute of Chemistry (IQ), Federal University of Rio de Janeiro (UFRJ), Cidade Universitária, Rio de Janeiro, RJ, Brazil; Analytical and Molecular Laboratory Center (CLAn), Institute of Chemistry (IQ), Federal University of Rio de Janeiro (UFRJ), Cidade Universitária, Rio de Janeiro, Brazil; Graduate Program in Veterinary Hygiene (PGHIGVET), Faculty of Veterinary Medicine, Fluminense Federal University (UFF), Niterói, RJ, Brazil
| |
Collapse
|
19
|
Chen M, Jiang S, Sun L, Wang H, Di L, Liu Y, Zhang Y, Zhuang H, Hong Y, Wang Z, Zhu F, Chen Y, Ji S, Yu Y, Chen Y, Du X. "Seesaw effect" between daptomycin and ceftobiprole in daptomycin-resistant methicillin-resistant Staphylococcus aureus isolates. Int J Antimicrob Agents 2025; 65:107469. [PMID: 39986399 DOI: 10.1016/j.ijantimicag.2025.107469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 12/28/2024] [Accepted: 02/14/2025] [Indexed: 02/24/2025]
Abstract
OBJECTIVES This study aimed to investigate the "seesaw effect" of daptomycin (DAP) and ceftobiprole (BPR) on DAP-resistant (DAP-R) methicillin-resistant Staphylococcus aureus (MRSA) isolates. METHODS Broth microdilution minimum inhibitory concentrations (MICs) of DAP and BPR were tested for laboratory-derived and clinical DAP-R MRSA isolates to estimate the "seesaw effect." Time-kill curves for seven representative DAP-R isolates were obtained using DAP and BPR to validate their synergistic activity in vitro. Whole genome sequencing as well as deletion and complementation of the mprF gene were performed to investigate the mechanisms of the "seesaw effect." RESULTS The BPR MICs decreased by half-fold in DAP-R MRSA isolates. The synergistic effect of DAP and BPR against representative clinical and community-associated MRSA (CA-MRSA) isolates was demonstrated in time-kill analyses, showing that synergistic activity was preferred in CA-MRSA compared with hospital-associated MRSA. The mprF mutations were identified in isolates exhibiting the "seesaw effect." These mutations increased the DAP MIC while decreasing the BPR MIC. CONCLUSIONS The "seesaw effect" between DAP and BPR was prevalent among DAP-R MRSA isolates. This phenomenon was associated with the mprF mutations of MRSA.
Collapse
Affiliation(s)
- Mengzhen Chen
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China; Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shengnan Jiang
- Center of Laboratory Medicine, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Lu Sun
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China; Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Haiping Wang
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China; Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lingfang Di
- Department of Clinical Laboratory, Tongxiang First people's hospital, Tongxiang, Zhejiang, China
| | - Yeqiong Liu
- Shanghai Jiading Central Hospital, Shanghai, China
| | - Ying Zhang
- Center of Laboratory Medicine, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Hemu Zhuang
- Respiratory Department, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yueqin Hong
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China; Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhengan Wang
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China; Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Feiteng Zhu
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China; Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yiyi Chen
- Center of Laboratory Medicine, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Shujuan Ji
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China; Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yunsong Yu
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China; Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Yan Chen
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China; Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Xiaoxing Du
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China; Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
20
|
Kuriu A, Ishikawa K, Tsuchiya K, Furuta K, Kaito C. Xenopus laevis as an infection model for human pathogenic bacteria. Infect Immun 2025:e0012625. [PMID: 40310291 DOI: 10.1128/iai.00126-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Accepted: 04/03/2025] [Indexed: 05/02/2025] Open
Abstract
Animal infection models are essential for understanding bacterial pathogenicity and corresponding host immune responses. In this study, we investigated whether juvenile Xenopus laevis could be used as an infection model for human pathogenic bacteria. Xenopus frogs succumbed to intraperitoneal injection containing the human pathogenic bacteria Staphylococcus aureus, Pseudomonas aeruginosa, and Listeria monocytogenes. In contrast, non-pathogenic bacteria Bacillus subtilis and Escherichia coli did not induce mortality in Xenopus frogs. The administration of appropriate antibiotics suppressed mortality caused by S. aureus and P. aeruginosa. Strains lacking the agr locus, cvfA (rny) gene, or hemolysin genes in S. aureus, LIPI-1-deleted mutant of L. monocytogenes, which attenuate virulence within mammals, exhibited reduced virulence in Xenopus frogs compared with their respective wild-type counterparts. Bacterial distribution analysis revealed that S. aureus persisted in the blood, liver, heart, and muscles of Xenopus frogs until death. These results suggested that intraperitoneal injection of human pathogenic bacteria induces sepsis-like symptoms in Xenopus frogs, supporting their use as a valuable animal model for evaluating antimicrobial efficacy and identifying virulence genes in various human pathogenic bacteria.
Collapse
Affiliation(s)
- Ayano Kuriu
- Division of Molecular Biology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Kazuya Ishikawa
- Division of Molecular Biology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Kohsuke Tsuchiya
- Division of Immunology and Molecular Biology, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Kazuyuki Furuta
- Division of Molecular Biology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Chikara Kaito
- Division of Molecular Biology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| |
Collapse
|
21
|
Xiong X, Wen X, Zhang Y, Li X, Zhang Y, Long N. Microbial community and immune modulation enable effective treatment of methicillin-resistant Staphylococcus aureus skin infections with linalool. Microb Pathog 2025; 202:107406. [PMID: 39999898 DOI: 10.1016/j.micpath.2025.107406] [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: 08/23/2024] [Revised: 02/14/2025] [Accepted: 02/20/2025] [Indexed: 02/27/2025]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA), as one of the main pathogens causing skin and soft tissue infections, poses challenges in treatment due to its high resistance to antibiotics. As one of the efficacious essential oil components in numerous traditional Chinese medicines, linalool was believed to possess antimicrobial activity against pathogenic microorganisms. Here, we investigated the therapeutic effects of linalool on MRSA-infected mice by examining their post-treatment outcomes. This was done through observations of physiological conditions, pathological sections, inflammatory factors, and changes in the skin microenvironment. We have confirmed the effectiveness of linalool in treating MRSA infections. Mice treated with linalool exhibited more pronounced signs of recovery, such as reduced skin necrosis, increased fibroplasia, greater neovascularization, and resolution of inflammatory infiltration. In addition, there was an improvement in the inflammatory environment, with a decrease in inflammatory factors. The microbial composition on the skin surface also confirmed this improvement. After linalool treatment, mice exhibited better species diversity on the skin, making it easier to maintain the skin's homeostasis. The excellent performance of linalool in combating MRSA infections provides a new direction for the search for new antibiotics against multidrug-resistant bacteria, highlighting the potential of linalool as a promising anti-MRSA drug.
Collapse
Affiliation(s)
- Xingyun Xiong
- Dazhu County People's Hospital, Dazhou, 635100, PR China
| | - Xin Wen
- Dazhu County People's Hospital, Dazhou, 635100, PR China
| | - Yangjing Zhang
- Dazhu County People's Hospital, Dazhou, 635100, PR China
| | - Xiaofang Li
- Dazhu County People's Hospital, Dazhou, 635100, PR China
| | - Yuping Zhang
- Dazhu County People's Hospital, Dazhou, 635100, PR China
| | - Nana Long
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, 610083, PR China; Sichuan Provincial Engineering Laboratory for Prevention and Control Technology of Veterinary Drug Residue in Animal-origin Food, Chengdu Medical College, Chengdu, 610083, PR China.
| |
Collapse
|
22
|
Mathew N, Suresh SA, Pallivalappil L, Suseela KV. Nasal Methicillin Resistant Staphylococcus Aureus Colonisation and the Incidence of Invasive Staphylococcal Infection in Patients Undergoing Hemodialysis. Indian J Nephrol 2025; 35:419-421. [PMID: 40352893 PMCID: PMC12065608 DOI: 10.25259/ijn_418_2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 01/07/2025] [Indexed: 05/14/2025] Open
Affiliation(s)
- Navya Mathew
- Department of Pulmonary Medicine, Amala Institute of Medical Sciences, Thrissur, Kerala, India
| | - Shweta Ann Suresh
- Department of Pulmonary Medicine, Amala Institute of Medical Sciences, Thrissur, Kerala, India
| | - Lisha Pallivalappil
- Department of Pulmonary Medicine and Critical Care, Amala Institute of Medical Sciences, Thrissur, Kerala, India
| | | |
Collapse
|
23
|
Khan ZA, Song SS, Xu H, Ahmad M, Wang A, Abdullah A, Jiang L, Ding X. Elimination of intracellular microbes using drug combination therapy and unveiling survival mechanism of host cells upon microbial invasion. Int J Antimicrob Agents 2025; 65:107471. [PMID: 39986398 DOI: 10.1016/j.ijantimicag.2025.107471] [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: 08/09/2024] [Revised: 01/16/2025] [Accepted: 02/14/2025] [Indexed: 02/24/2025]
Abstract
Intracellular microbes are actively present in various tumor types in low biomass and play a major role in metastasis. Eliminating intracellular microbes on a cellular level with precision remains a challenge. To address this issue, we designed a screening pipeline to characterize intracellular microbes and their interaction with host cells. We used host and microbial in vitro lab-based constant and reproducible model, host as (mammalian cancer HeLa), and microbial strain as (Escherichia coli 25922). To study the pharmacological impact on intracellular bacterial load, we used antibiotics (ampicillin, roxithromycin, and ciprofloxacin) and chemotherapy drugs (doxorubicin and cisplatin) as external stimuli for both host and microbes. We found that increasing pharmacological stress does not increase microbial load inside the host cells. Eliminations of intracellular bacteria was done by using permutation orthogonal arrays (POA), whereby we acquired optimal drug combination in particular sequence of drugs, which reduced 90%-95% of the intracellular microbial load. Proteomic analysis revealed that upon invasion of Escherichia coli 25922, HeLa cells enriched ATP production pathways to activate intermediate filaments, which should be investigated closely via in vivo models.
Collapse
Affiliation(s)
- Zara Ahmad Khan
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China; Department of Pathology, Wenling First People's Hospital, Wenling City, Zhejiang Province, China
| | - Sha-Sha Song
- Pathology Department, Yantai Fushan People's Hospital, Yantai, China
| | - Hongquan Xu
- Department of Statistics and Data Science, University of California, Los Angeles, California, USA
| | - Mashaal Ahmad
- Department of Anatomy, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
| | - Aiting Wang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China; Department of Pathology, Wenling First People's Hospital, Wenling City, Zhejiang Province, China
| | - Aynur Abdullah
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China; Department of Pathology, Wenling First People's Hospital, Wenling City, Zhejiang Province, China
| | - Lai Jiang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Xianting Ding
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China; Department of Pathology, Wenling First People's Hospital, Wenling City, Zhejiang Province, China.
| |
Collapse
|
24
|
Pan X, Zhang J, Pan F, Wang C, Qin H, Yu F, Zhang T, Liu W, Wu Q, Ma Z, Weng W, Chen L, Yu F, Zhang H, Wang B. Longitudinal surveillance of the molecular evolution of methicillin-resistant Staphylococcus aureus isolates from pediatric patients in Shanghai, China, from 2013 to 2022. mSystems 2025:e0037125. [PMID: 40304507 DOI: 10.1128/msystems.00371-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Accepted: 04/01/2025] [Indexed: 05/02/2025] Open
Abstract
The objective of this research was to investigate the genomic epidemiology of methicillin-resistant Staphylococcus aureus (MRSA) in a pediatric population in Shanghai, China. Whole-genome sequencing was conducted for 492 randomly selected MRSA isolates obtained from a pediatric hospital between 2013 and 2022. ST59 (37.4%), ST398 (22.4%), ST88 (5.7%), and ST22 (5.5%) were the predominant lineages among these children. While ST59 maintained a dominant annual proportion before 2017, the proportion of ST398 gradually increased from 2013 to 2016, with ST398 ultimately emerging as a prevalent clone with a proportion comparable to that of ST59 after 2017. Among the prevalent STs, the spa-SCCmec structure also experienced dynamic changes. Within ST59, the t437-IV subtype experienced a decline and has even been replaced by t172-IV in recent years. In ST398, the t011-V subtype appeared in 2014 and rapidly became the leading subtype. The antibiotic resistance profiles and virulence factors exhibited clone-related features. Compared with other prevalent lineages, ST59 presented high resistance to erythromycin and clindamycin, whereas ST398 presented relatively low resistance to common antimicrobial agents and fewer virulence determinants. Panton-Valentine leucocidin was more common in ST338 and ST1232, whereas toxic shock syndrome toxin was closely associated with ST1 and ST5. The MRSA cases could also be classified into community- and hospital-associated cases, with highly significant differences between the two in terms of demographic characteristics, clindamycin susceptibility, and virulence genes. In conclusion, this study revealed high genetic diversity and dynamic changes in the molecular epidemiology of pediatric MRSA isolates from Shanghai collected over a decade. IMPORTANCE Methicillin-resistant Staphylococcus aureus (MRSA) has emerged as a significant global health concern. Previous research on MRSA epidemiology has predominantly focused on adult populations or targeted specific infection sites, while there was limited research on the long-term evolution of MRSA from the pediatric population. This study addresses this knowledge gap by conducting a comprehensive, 10-year surveillance of pediatric MRSA isolates using whole-genome sequencing. We characterized the molecular typing, as well as the phenotypic and genotypic antimicrobial resistance profiles, and virulence factors present in MRSA isolates obtained from children. Our results highlight the imperative for continuous, vigilant monitoring of MRSA within the pediatric demographic to track its evolving genetic landscape.
Collapse
Affiliation(s)
- Xiaozhou Pan
- Department of Clinical Laboratory, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, School of Medicine, Shanghai, China
| | - Jiao Zhang
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Fen Pan
- Department of Clinical Laboratory, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, School of Medicine, Shanghai, China
| | - Chun Wang
- Department of Clinical Laboratory, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Huihong Qin
- Department of Clinical Laboratory, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Fangyuan Yu
- Department of Clinical Laboratory, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Tiandong Zhang
- Department of Clinical Laboratory, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wenxin Liu
- Department of Clinical Laboratory, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, School of Medicine, Shanghai, China
| | - Qianyue Wu
- Department of Clinical Laboratory, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, School of Medicine, Shanghai, China
| | - Zhan Ma
- Department of Clinical Laboratory, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, School of Medicine, Shanghai, China
| | - Wenhao Weng
- Department of Clinical Laboratory, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, School of Medicine, Shanghai, China
| | - Liang Chen
- School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, New York, USA
| | - Fangyou Yu
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Hong Zhang
- Department of Clinical Laboratory, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, School of Medicine, Shanghai, China
| | - Bingjie Wang
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
25
|
Sharma A, Heffernan LM, Hoang K, Jeyaseelan S, Beavers WN, Abuaita BH. Activation of the endoplasmic reticulum stress regulator IRE1α compromises pulmonary host defenses. Cell Rep 2025; 44:115632. [PMID: 40315054 DOI: 10.1016/j.celrep.2025.115632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 01/29/2025] [Accepted: 04/09/2025] [Indexed: 05/04/2025] Open
Abstract
The endoplasmic reticulum (ER) stress sensor inositol-requiring enzyme 1α (IRE1α) is associated with lung infections where innate immune cells are drivers for progression and resolution ammatory cytokinesflammation. Yet, the role of IRE1α in pulmonary innate immune host defense during acute respiratory infection remains unexplored. Here, we found that activation of IRE1α in infected lungs compromises immunity against methicillin-resistant Staphylococcus aureus (MRSA)-induced primary and secondary pneumonia. Moreover, activation of IRE1α in MRSA-infected lungs and alveolar macrophages (AMs) leads to exacerbated production of inflammatory mediators followed by cell death. Ablation of myeloid IRE1α or global IRE1α inhibition confers protection against MRSA-induced pneumonia with improved survival, bacterial clearance, cytokine reduction, and lung injury. In addition, loss of myeloid IRE1α protects mice against MRSA-induced secondary to influenza pneumonia by promoting AM survival. Thus, activation of IRE1α is detrimental to pneumonia, and therefore, it shows potential as a target to control excessive unresolved lung inflammation.
Collapse
Affiliation(s)
- Amit Sharma
- Department of Pathobiological Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA 70803, USA
| | - Linda M Heffernan
- Department of Pathobiological Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA 70803, USA
| | - Ky Hoang
- Department of Pathobiological Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA 70803, USA
| | - Samithamby Jeyaseelan
- Department of Pathobiological Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA 70803, USA
| | - William N Beavers
- Department of Pathobiological Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA 70803, USA; Mass Spectrometry Resource Center, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA 70803, USA
| | - Basel H Abuaita
- Department of Pathobiological Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA 70803, USA.
| |
Collapse
|
26
|
Liu J, Yao K, Sun R, Ma X, Ma C, Chen X, Jiang Y, Wang T, Chen T, Shaw C, Zhou M, Wang L. Discovery and Optimisation of Novel Bombinin-Derived Peptides from Bombina variegata against Staphylococcus aureus. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10542-1. [PMID: 40301231 DOI: 10.1007/s12602-025-10542-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/16/2025] [Indexed: 05/01/2025]
Abstract
Amphibian skin-secreted antimicrobial peptides (AMPs) have garnered significant attention for their excellent biological activity and low propensity for drug resistance over the past 40 years. Bombinins and bombinin H, two classes of AMPs isolated from the skin secretions of Bombina species, demonstrate strong antimicrobial activity against broad-spectrum microorganisms. In this study, two novel peptides, bombinin-like peptide 7S and bombinin-H2L, were identified from the toad, Bombina variegata. While both peptides exhibited broad-spectrum antimicrobial activity, they also showed relatively high cytotoxicity. To explore the structure-activity relationship and enhance therapeutic potential, bombinin-H2L, which displayed stronger average antimicrobial activity, was used as a template. With the aid of bioinformatics analysis, a series of bombinin-H2L analogues were designed by increasing the net positive charges and/or adjusting the amphiphilicity of the parent peptide. Among these analogues, [Arg8, 15]BH2L and [Lys7, 8]BH2L demonstrated high therapeutic efficacy and specificity toward clinically isolated, drug-resistant Staphylococcus aureus strains in both in vitro and ex vivo tests. Their notable biosafety profiles, sensitivity to diverse environments, and ability to disrupt biofilms highlight their potential for further development. Additionally, studies on the mechanism of [Arg8, 15]BH2L and [Lys7, 8]BH2L revealed a membrane-targeted antimicrobial mechanism, with its antibacterial function exerted by disrupting the integrity of bacterial membranes. These findings provide valuable insights into structural modifications of bombinin H peptides for enhanced activity, and [Arg8, 15]BH2L and [Lys7, 8]BH2L have the potential as promising candidates for novel antibacterial agents in treated bacterial skin infections.
Collapse
Affiliation(s)
- Jiachen Liu
- Natural Drug Discovery Group, School of Pharmacy, Queen's University Belfast, Belfast, Northern Ireland, BT9 7BL, UK
| | - Keyi Yao
- Natural Drug Discovery Group, School of Pharmacy, Queen's University Belfast, Belfast, Northern Ireland, BT9 7BL, UK
| | - Ruize Sun
- Natural Drug Discovery Group, School of Pharmacy, Queen's University Belfast, Belfast, Northern Ireland, BT9 7BL, UK
| | - Xiaonan Ma
- Natural Drug Discovery Group, School of Pharmacy, Queen's University Belfast, Belfast, Northern Ireland, BT9 7BL, UK
| | - Chengbang Ma
- Natural Drug Discovery Group, School of Pharmacy, Queen's University Belfast, Belfast, Northern Ireland, BT9 7BL, UK
| | - Xiaoling Chen
- Natural Drug Discovery Group, School of Pharmacy, Queen's University Belfast, Belfast, Northern Ireland, BT9 7BL, UK
| | - Yangyang Jiang
- Natural Drug Discovery Group, School of Pharmacy, Queen's University Belfast, Belfast, Northern Ireland, BT9 7BL, UK.
| | - Tao Wang
- Natural Drug Discovery Group, School of Pharmacy, Queen's University Belfast, Belfast, Northern Ireland, BT9 7BL, UK.
| | - Tianbao Chen
- Natural Drug Discovery Group, School of Pharmacy, Queen's University Belfast, Belfast, Northern Ireland, BT9 7BL, UK
| | - Chris Shaw
- Natural Drug Discovery Group, School of Pharmacy, Queen's University Belfast, Belfast, Northern Ireland, BT9 7BL, UK
| | - Mei Zhou
- Natural Drug Discovery Group, School of Pharmacy, Queen's University Belfast, Belfast, Northern Ireland, BT9 7BL, UK
| | - Lei Wang
- Natural Drug Discovery Group, School of Pharmacy, Queen's University Belfast, Belfast, Northern Ireland, BT9 7BL, UK
| |
Collapse
|
27
|
Gao M, Yu X, Yang C, Mi Z, Bai C, Liu C, Liu H. Characteristics and Antibacterial Activity of Staphylococcus aureus Phage-Derived Endolysin LysP4. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10543-0. [PMID: 40299201 DOI: 10.1007/s12602-025-10543-0] [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] [Accepted: 04/16/2025] [Indexed: 04/30/2025]
Abstract
The rise in multidrug resistance and strong biofilm-forming ability of Staphylococcus aureus has led to significant public health concerns. Phage or phage-derived components, such as depolymerase or endolysin, have been considered as potential alternatives to antibiotics for combating antibiotic-resistant bacterial infections. In this study, we cloned and expressed a Staphylococcus aureus phage endolysin, LysP4, and identified its lytic activity. The bactericidal effect of LysP4 was more pronounced against planktonic cells in the logarithmic phase compared to those in the stationary phase. LysP4 reduces bacterial counts by 3 log CFU/mL in 60 min and about 2 log CFU/mL during the stationary phase. LysP4 exhibited optimal lytic activity at pH 5.0-7.0 and remained stable across a temperature range of 16 to 40 °C, with maximal activity observed at 37 °C. LysP4 effectively targets 31 of 38 Staphylococcus strains and successfully eliminates biofilms, reducing bacterial counts by 4 log CFU/mL when combined with vancomycin. Notably, LysP4 demonstrated no hemolytic effects on human red blood cells and no toxic effects on embryonic kidney cells or lung cancer cells. Based on these findings, we believe that LysP4 holds promise as a biological control agent against Staphylococcus infections.
Collapse
Affiliation(s)
- Mingming Gao
- Chinese PLA General Hospital, Beijing, 100853, China
| | - Xinting Yu
- Mianyang Central Hospital, Mianyang, 621000, Sichuan, China
| | - Cuiping Yang
- College of Pulmonary and Critical Care Medicine, The 8th Medical Centre, Chinese PLA General Hospital, Beijing, 100853, China
| | - Zhiqiang Mi
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Changqing Bai
- Department of Respiratory and Critical Care Diseases, General Hospital of Shenzhen University, Shenzhen, 518055, China
| | - Chuanbin Liu
- Western Medical Branch of PLA General Hospital, Beijing, 100853, China.
| | - Huiying Liu
- College of Pulmonary and Critical Care Medicine, The 8th Medical Centre, Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
28
|
Lovins V, Farias Amorim C, Robles N, Murga-Garrido S, Ting-Chun Pan J, Singh TP, DeNardo E, Carvalho LP, Carvalho EM, Scott P, Grice EA. Staphylococcus aureus promotes strain-dependent immunopathology during cutaneous leishmaniasis through induction of IL-1β. Cell Rep 2025; 44:115624. [PMID: 40293920 DOI: 10.1016/j.celrep.2025.115624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 01/29/2025] [Accepted: 04/07/2025] [Indexed: 04/30/2025] Open
Abstract
Cutaneous leishmaniasis is a parasitic infection that causes a spectrum of pathology ranging from single, self-healing lesions to disfiguring chronic wounds. In severe disease, uncontrolled inflammation exacerbates tissue damage and delays healing, though the contributing factors are unclear. We previously observed that delayed healing was associated with Staphylococcus aureus in the lesional microbiota of patients with cutaneous leishmaniasis. To investigate how S. aureus impacts immunopathology during leishmania infection, we established a murine model of S. aureus colonization with clinical isolates followed by Leishmania infection. S. aureus triggered early production of interleukin (IL)-1β during Leishmania infection, which was critical for neutrophil recruitment and cutaneous inflammation. S. aureus isolates differentially induced IL-1β and neutrophil recruitment, and isolates that induced greater neutrophil recruitment were resistant to neutrophil killing and persisted longer. We reveal a mechanism whereby S. aureus mediates immunopathology during cutaneous leishmaniasis, suggesting IL-1β as a promising immunomodulatory target for non-healing infections.
Collapse
Affiliation(s)
- Victoria Lovins
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Camila Farias Amorim
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nélida Robles
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sofía Murga-Garrido
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jamie Ting-Chun Pan
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Tej P Singh
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Erin DeNardo
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lucas P Carvalho
- Laboratório de Pesquisas Clínicas de Instituto de Pesquisas Gonçalo Muniz, Fiocruz, Bahia, Brazil
| | - Edgar M Carvalho
- Laboratório de Pesquisas Clínicas de Instituto de Pesquisas Gonçalo Muniz, Fiocruz, Bahia, Brazil
| | - Phillip Scott
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Elizabeth A Grice
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
29
|
Xie Y, Wang X, Chang T, Chen Z, Luo Y, Zhang J, Wang H, Dong J, Chen F, Zhang J, Guan D. Novel Aryl Sulfonium Modification on Vancomycin to Tackle MRSA and VRE In Vitro and In Vivo through Dual Enhanced Cell-Wall and Membrane Inhibition. J Med Chem 2025; 68:8310-8329. [PMID: 40219998 DOI: 10.1021/acs.jmedchem.4c03028] [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: 04/14/2025]
Abstract
Gram-positive superbugs resistant to methicillin and vancomycin pose a severe threat to global public health, urgently demanding novel therapeutic strategies. Herein, we rationally designed and synthesized vancomycin derivatives modified with diverse aryl sulfonium moieties to reactivate its antibacterial potency. By optimizing the sulfonium-based SAR, we got derivatives 2-3 orders of magnitude more active in vitro than vancomycin. Subsequently, preliminary toxicity evaluations for the optimal derivative, 7e, indicated a favorable therapeutic index, while pharmacokinetic assays revealed its good properties, suggesting great drug-like potential. Notably, 7e showed extremely potent in vivo protection efficacy by only a single-dose treatment in the challenging methicillin-resistant Staphylococcus aureus and VRE lethal sepsis mice models. Moreover, two independent and synergistic mechanisms of action were uncovered: membrane perturbation and enhanced cell wall biosynthesis inhibition. These findings revealed the unknown role of sulfonium strategy in vitro and in vivo and positioned 7e as a promising candidate for future development.
Collapse
Affiliation(s)
- Yuanyuan Xie
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, no. 555 Zuchongzhi Rd, Pudong, Shanghai 201203, China
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xiaowen Wang
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, no. 555 Zuchongzhi Rd, Pudong, Shanghai 201203, China
| | - Taopeng Chang
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China
| | - Zhifu Chen
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, China
| | - Youhong Luo
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang 261031, China
| | - Jingwen Zhang
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang 261031, China
| | - Hui Wang
- Nanjing Cantech Microbial Technology Co. Ltd. no. 18, Zhilan Rd, Jiangning, Nanjing 211100, China
| | - Jinhua Dong
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Feifei Chen
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, no. 555 Zuchongzhi Rd, Pudong, Shanghai 201203, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Jinyong Zhang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, China
| | - Dongliang Guan
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, no. 555 Zuchongzhi Rd, Pudong, Shanghai 201203, China
- University of Chinese Academy of Sciences, no. 19A Yuquan Road, Beijing 100049, China
| |
Collapse
|
30
|
Berrow M, Brooks A, Kotowska AM, Watts J, Riordan L, Kidger L, Scurr DJ, Nikoi ND, Banzhaf M, Bryant JA, Greenway S, Mendez V, Norton B, de Cogan F. Development and characterisation of antimicrobial epoxy resin. Sci Rep 2025; 15:12463. [PMID: 40263385 PMCID: PMC12015595 DOI: 10.1038/s41598-025-90465-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 02/13/2025] [Indexed: 04/24/2025] Open
Abstract
Surface contamination is an important, if under-discussed, route of infection transmission. In this study, we suspended chlorhexidine digluconate (CHX) in epoxy resin. CHX was found to be stably incorporated into the material, and its addition to epoxy resin was found to have minimal effects on the optical transparency of the material. After application of the epoxy resin to steel surfaces, time-of-flight secondary ion mass spectrometry revealed that CHX was uniformly present over the surface. Surfaces painted with CHX-resin were found to have significant, reproducible antimicrobial efficacy against E. coli, S. aureus, and C. albicans. We have shown that the addition of CHX has minimal effects on the adhesion of the epoxy resin to surfaces, as well as a high durability of the antimicrobial efficacy. We believe that this material has a wide array of applications, and could be utilised to confer significant, low-cost antimicrobial efficacy to existing surfaces, to prevent surface contamination, and to stop the transmission of infectious disease.
Collapse
Affiliation(s)
- Madeline Berrow
- School of Pharmacy, University of Nottingham, Nottingham, UK
| | | | - Anna M Kotowska
- School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Julie Watts
- School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Lily Riordan
- School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Luke Kidger
- School of Pharmacy, University of Nottingham, Nottingham, UK
| | - David J Scurr
- School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Naa Dei Nikoi
- School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Manuel Banzhaf
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, UK
| | - Jack Alfred Bryant
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, UK
| | - Simon Greenway
- Indestructible Paint Ltd, 16-25 Pentos Drive, Birmingham, UK
| | - Violaine Mendez
- Indestructible Paint Ltd, 16-25 Pentos Drive, Birmingham, UK
| | - Brian Norton
- Indestructible Paint Ltd, 16-25 Pentos Drive, Birmingham, UK
| | | |
Collapse
|
31
|
Liu X, Zhao X, Qiu H, Liang W, Liu L, Sun Y, Zhao L, Wang X, Liang H. Antibacterial Activity of GO-Based Composites Enhanced by Phosphonate-Functionalized Ionic Liquids and Silver. MATERIALS (BASEL, SWITZERLAND) 2025; 18:1889. [PMID: 40333535 PMCID: PMC12028358 DOI: 10.3390/ma18081889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2025] [Revised: 04/12/2025] [Accepted: 04/15/2025] [Indexed: 05/09/2025]
Abstract
The development of antibiotic-independent antimicrobial materials is critical for addressing bacterial resistance to conventional antibiotics. Currently, there is a lack of comprehensive understanding of ionic liquid-modified composites in antimicrobial applications. Here, we innovatively prepared GO-based composites modified with phosphonate ionic liquids via a series of surface functionalizations. The resulting antibacterial composites exhibit significant broad-spectrum activity against both Gram-negative and Gram-positive bacteria, including drug-resistant strains, with stronger efficacy against Gram-negative species. Additionally, the material features excellent long-term reusability and the ability to inhibit/destroy biofilms, which is vital for combating persistent infections. Mechanistic studies reveal its antibacterial effects through multiple pathways: disrupting bacterial membranes, inducing ROS, and inactivating intracellular substances-mechanisms less likely to promote resistance. Overall, these phosphonate ionic liquid-modified polycationic materials demonstrate substantial potential in treating bacterial infections, offering a promising strategy to tackle antibiotic resistance challenges.
Collapse
Affiliation(s)
- Xinyu Liu
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo 315211, China; (X.Z.); (H.Q.); (W.L.); (L.L.); (L.Z.)
| | - Xing Zhao
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo 315211, China; (X.Z.); (H.Q.); (W.L.); (L.L.); (L.Z.)
| | - Hongda Qiu
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo 315211, China; (X.Z.); (H.Q.); (W.L.); (L.L.); (L.Z.)
| | - Weida Liang
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo 315211, China; (X.Z.); (H.Q.); (W.L.); (L.L.); (L.Z.)
| | - Linlin Liu
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo 315211, China; (X.Z.); (H.Q.); (W.L.); (L.L.); (L.Z.)
| | - Yunyu Sun
- Anhui Key Laboratory of Spin Electron and Nanomaterials of Anhui Higher Education Institutes, School of Chemistry and Chemical Engineering, Suzhou University, Suzhou 234000, China;
| | - Lingling Zhao
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo 315211, China; (X.Z.); (H.Q.); (W.L.); (L.L.); (L.Z.)
| | - Xiao Wang
- Health Science Center, Ningbo University, Ningbo 315211, China;
| | - Hongze Liang
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo 315211, China; (X.Z.); (H.Q.); (W.L.); (L.L.); (L.Z.)
| |
Collapse
|
32
|
Liang Y, Zhang H, Dai S, Cong Y, Wu W. Inhibiting Staphylococcus aureus Virulence Factors: Advances in Traditional Chinese Medicines and Active Compounds. Curr Microbiol 2025; 82:247. [PMID: 40244366 DOI: 10.1007/s00284-025-04236-8] [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: 12/08/2024] [Accepted: 04/04/2025] [Indexed: 04/18/2025]
Abstract
Staphylococcus aureus is one of the most prevalent antibiotic-resistant bacteria, characterized by high morbidity and mortality. The pathogenicity of S. aureus relies on the production of multiple virulence factors. In recent years, antivirulence strategies have shown promise in developing antiinfective drugs by targeting the inhibition of bacterial virulence factors rather than directly killing pathogens. In Asia, some traditional Chinese medicines have a long history of antiinfective application and have demonstrated therapeutic efficacy. However, their antiinfective mechanism has not been fully elucidated. Recent studies have revealed that numerous extracts of TCM, as well as pure compounds from TCM, significantly inhibited the expression of virulence factors of S. aureus, which might be one of their antiinfective mechanisms with potential for the development of novel antiinfective agents. In this review, we summarized the major virulence factors of S. aureus and recent advances in TCM-derived antivirulence agents, including TCM formulae, single herbs, and isolated bioactive compounds, which showed antivirulence capability against S. aureus. Investigating the antivirulence mechanism of TCM not only enhances our understanding of TCM's antiinfective mechanisms but also facilitates the isolation of active compounds with therapeutic potential against S. aureus infection.
Collapse
Affiliation(s)
- Yuqi Liang
- Zhuhai College of Science and Technology, Zhuhai, 519041, China
| | - Huiyong Zhang
- The Forth Affiliated Hospital of China Medical University, Shenyang, 110084, China
| | - Sisi Dai
- Zhuhai College of Science and Technology, Zhuhai, 519041, China
| | - Yanguang Cong
- Department of Clinical Laboratory, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523710, China.
- Dongguan Key Laboratory for Pathogenesis and Experimental Diagnosis of Infectious Diseases, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523710, China.
| | - Wei Wu
- Zhuhai College of Science and Technology, Zhuhai, 519041, China.
| |
Collapse
|
33
|
Sarac B, Yücer S, Ciftci F. MXenes in microbiology and virology: from pathogen detection to antimicrobial applications. NANOSCALE 2025; 17:9619-9651. [PMID: 40135595 DOI: 10.1039/d5nr00477b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/27/2025]
Abstract
MXenes, a rapidly emerging class of two-dimensional materials, have demonstrated exceptional versatility and functionality across various domains, including microbiology and virology. Recent advancements in MXene synthesis techniques, encompassing both top-down and bottom-up approaches, have expanded their potential applications in pathogen detection, antimicrobial treatments, and biomedical platforms. This review highlights the unique physicochemical properties of MXenes, including their large surface area, tunable surface chemistry, and high biocompatibility, which contribute to their antimicrobial efficacy against bacteria, fungi, and viruses, such as SARS-CoV-2. The antibacterial mechanisms of MXenes, including membrane disruption, reactive oxygen species (ROS) generation, and photothermal inactivation, are discussed alongside hybridization strategies that enhance their bioactivity. Additionally, the challenges and future prospects of MXenes in developing advanced antimicrobial coatings, diagnostic tools, and therapeutic systems are outlined. By addressing current limitations and exploring innovative solutions, this study underscores the transformative potential of MXenes in microbiology, virology, and biomedical applications.
Collapse
Affiliation(s)
- Begüm Sarac
- Faculty of Engineering, Department of Biomedical Engineering, Fatih Sultan Mehmet Vakıf University, Istanbul, Turkey.
| | - Seydanur Yücer
- Faculty of Engineering, Department of Biomedical Engineering, Fatih Sultan Mehmet Vakıf University, Istanbul, Turkey.
| | - Fatih Ciftci
- Faculty of Engineering, Department of Biomedical Engineering, Fatih Sultan Mehmet Vakıf University, Istanbul, Turkey.
- Department of Technology Transfer Office, Fatih Sultan Mehmet Vakıf University, Istanbul, Turkey
| |
Collapse
|
34
|
Kawayanagi T, Kawada-Matsuo M, Kusaka S, Yasutomi Y, Suzuki Y, Nishihama S, Le MNT, Sugawara Y, Hisatsune J, Kutsuno S, Asakawa M, Takeshita T, Nomura R, Shiba H, Sugai M, Komatsuzawa H. Clinical and genetic analysis of oral and nasal staphylococcus aureus isolates in dental patients. Sci Rep 2025; 15:13149. [PMID: 40240397 PMCID: PMC12003906 DOI: 10.1038/s41598-025-93773-0] [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: 10/24/2024] [Accepted: 03/10/2025] [Indexed: 04/18/2025] Open
Abstract
Staphylococcus aureus is a common bacterium that sometimes causes various pyogenic diseases. Methicillin resistant S. aureus (MRSA) infections are particularly difficult to treat. Recently, MRSA has been spreading in the community, so it is important to determine the prevalence of MRSA in the community and to conduct epidemiological studies from genetic and statistical perspectives. In this study, S. aureus/MRSA was isolated from the oral and nasal cavities of 504 dental patients (65 inpatients and 439 outpatients). Sixty-two S. aureus strains and 9 MRSA strains were isolated from the oral cavity, and 112 S. aureus strains and 21 MRSA strains were isolated from the nasal cavity. Multi-locus sequence typing (MLST) analysis showed ST8 was high (18 isolates) among 30 MRSA isolates, whereas among 144 methicillin sensitive isolates, ST15 (25 isolates) and ST8 (20 isolates) were high. Statistical analysis of the patients' clinical status revealed a correlation between oral S. aureus and denture use. Among the 34 patients from whom S. aureus was isolated from both sites, 25 had the same ST, and 23 showed below 40 single-nucleotide polymorphisms which are considered to be identical strains. Our study revealed various properties of S. aureus/MRSA in the oral and nasal cavities as commensals.
Collapse
Affiliation(s)
- Tomoki Kawayanagi
- Department of Biological Endodontics, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- Department of Bacteriology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Kasumi 1-2-3, Hiroshima City, 734-8551, Hiroshima, Japan
| | - Miki Kawada-Matsuo
- Department of Bacteriology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Kasumi 1-2-3, Hiroshima City, 734-8551, Hiroshima, Japan.
- Project Research Center for Nosocomial Infectious Diseases, Hiroshima University, Hiroshima, Japan.
| | - Satoru Kusaka
- Department of Bacteriology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Kasumi 1-2-3, Hiroshima City, 734-8551, Hiroshima, Japan
- Department of Pediatric Dentistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yoshimasa Yasutomi
- Department of Bacteriology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Kasumi 1-2-3, Hiroshima City, 734-8551, Hiroshima, Japan
| | - Yujin Suzuki
- Department of Bacteriology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Kasumi 1-2-3, Hiroshima City, 734-8551, Hiroshima, Japan
| | - Saki Nishihama
- Department of Biological Endodontics, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- Department of Bacteriology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Kasumi 1-2-3, Hiroshima City, 734-8551, Hiroshima, Japan
| | - Mi Nguyen-Tra Le
- Department of Bacteriology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Kasumi 1-2-3, Hiroshima City, 734-8551, Hiroshima, Japan
- Project Research Center for Nosocomial Infectious Diseases, Hiroshima University, Hiroshima, Japan
| | - Yo Sugawara
- Antimicrobial Resistance Research Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Junzo Hisatsune
- Project Research Center for Nosocomial Infectious Diseases, Hiroshima University, Hiroshima, Japan
- Antimicrobial Resistance Research Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Shoko Kutsuno
- Antimicrobial Resistance Research Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Mikari Asakawa
- Section of Preventive and Public Health Dentistry, Division of Oral Health, Growth and Development, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Toru Takeshita
- Section of Preventive and Public Health Dentistry, Division of Oral Health, Growth and Development, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Ryota Nomura
- Department of Pediatric Dentistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Hideki Shiba
- Department of Biological Endodontics, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Motoyuki Sugai
- Project Research Center for Nosocomial Infectious Diseases, Hiroshima University, Hiroshima, Japan
- Antimicrobial Resistance Research Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Hitoshi Komatsuzawa
- Department of Bacteriology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Kasumi 1-2-3, Hiroshima City, 734-8551, Hiroshima, Japan.
- Project Research Center for Nosocomial Infectious Diseases, Hiroshima University, Hiroshima, Japan.
| |
Collapse
|
35
|
Ali L, Karki S, Boorgula GD, Mekakda A, Cagle-White B, Bhattarai S, Beaudoin R, Blakeney A, Singh S, Srivastava S, Abdel Aziz MH. A mechanistic understanding of the effect of Staphylococcus aureus VraS histidine kinase single-point mutation on antibiotic resistance. Microbiol Spectr 2025; 13:e0009525. [PMID: 40233945 PMCID: PMC12054033 DOI: 10.1128/spectrum.00095-25] [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: 01/09/2025] [Accepted: 03/17/2025] [Indexed: 04/17/2025] Open
Abstract
Bacterial genomic mutations in Staphylococcus aureus have been detected in isolated resistant clinical strains, yet their mechanistic effect on the development of antimicrobial resistance remains unclear. Resistance-associated regulatory systems acquire adaptive mutations under stress conditions that may lead to a gain-of-function effect and contribute to the resistance phenotype. Here, we investigate the effect of a single-point mutation (T331I) in VraS histidine kinase, part of the VraSR two-component system in S. aureus. VraSR senses and responds to environmental stress signals by upregulating gene expression for cell wall synthesis. A combination of enzyme kinetics, microbiological, and transcriptomic analyses revealed the mechanistic effect of the mutation on VraS and S. aureus. Michaelis-Menten kinetics show that the VraS mutation caused an increase in the autophosphorylation rate of VraS and enhanced its catalytic efficiency. The introduction of the mutation through recombineering coupled with CRISPR-Cas9 counterselection to the Newman strain wild-type (WT) genome doubled the minimum inhibitory concentration of three cell wall-targeting antibiotics. The mutation caused an enhanced S. aureus growth rate at sub-lethal doses of the antibiotics, confirming the causative effect of the mutation on bacterial persistence. Transcriptomic analysis showed a genome-wide alteration in gene expression levels and protein-protein interaction network of the mutant compared to the WT strain after exposure to vancomycin. The results suggest that the vraS mutation causes several mechanistic changes at the protein and cellular levels that favor bacterial survival under antibiotic stress and cause the mutation-harboring strains to become the dominant population during infection.IMPORTANCERising antimicrobial resistance (AMR) is a global health problem. Mutations in the two-component system have been linked to drug resistance in Staphylococcus aureus, yet the exact mechanism through which these mutations work is understudied. We investigated the T331I mutation in the vraS gene linked to sensing and responding to cell wall stress. The mutation caused changes at the protein level by increasing the catalytic efficiency of VraS kinase activity. Introducing the mutation to the genome of an S. aureus strain resulted in changes in phenotypic antibiotic susceptibility, growth kinetics, and genome-wide transcriptomic alterations. By a combination of enzyme kinetics, microbiological, and transcriptomic approaches, we highlight how small genetic changes can significantly impact bacterial physiology and survival under antibiotic stress. Understanding the mechanistic basis of antibiotic resistance is crucial to guide the development of novel therapeutic agents to combat AMR.
Collapse
Affiliation(s)
- Liaqat Ali
- Fisch College of Pharmacy, University of Texas at Tyler, Tyler, Texas, USA
| | - Salima Karki
- Fisch College of Pharmacy, University of Texas at Tyler, Tyler, Texas, USA
| | - Gunavanthi D. Boorgula
- Division of Infectious Diseases, Department of Medicine, School of Medicine, University of Texas at Tyler, Tyler, Texas, USA
| | - Amir Mekakda
- Fisch College of Pharmacy, University of Texas at Tyler, Tyler, Texas, USA
| | | | - Shrijan Bhattarai
- Fisch College of Pharmacy, University of Texas at Tyler, Tyler, Texas, USA
| | - Robert Beaudoin
- Fisch College of Pharmacy, University of Texas at Tyler, Tyler, Texas, USA
| | - Aryanna Blakeney
- Fisch College of Pharmacy, University of Texas at Tyler, Tyler, Texas, USA
| | - Sanjay Singh
- Division of Infectious Diseases, Department of Medicine, School of Medicine, University of Texas at Tyler, Tyler, Texas, USA
| | - Shashikant Srivastava
- Division of Infectious Diseases, Department of Medicine, School of Medicine, University of Texas at Tyler, Tyler, Texas, USA
- Department of Cellular and Molecular Biology, University of Texas Health Science Center at Tyler, Tyler, Texas, USA
| | - May H. Abdel Aziz
- Fisch College of Pharmacy, University of Texas at Tyler, Tyler, Texas, USA
| |
Collapse
|
36
|
Sun S. Emerging antibiotic resistance by various novel proteins/enzymes. Eur J Clin Microbiol Infect Dis 2025:10.1007/s10096-025-05126-4. [PMID: 40232578 DOI: 10.1007/s10096-025-05126-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 04/02/2025] [Indexed: 04/16/2025]
Abstract
BACKGROUND The emergence and dissemination of antibiotic resistance represents a significant and ever-increasing global threat to human, animal, and environmental health. The explosive proliferation of resistance has ultimately been seen in all clinically used antibiotics. Infections caused by antibiotic-resistant bacteria have been associated with an estimated 4,950,000 deaths annually, with extremely limited therapeutic options and only a few new antibiotics under development. To combat this silent pandemic, a better understanding of the molecular mechanisms of antibiotic resistance is immensely needed, which not only helps to improve the efficacy of current drugs in clinical use but also design new antimicrobial agents that are less susceptible to resistance. RESULTS The past few years have witnessed a number of new advances in revealing the molecular mechanisms of AMR. Following five sophisticated mechanisms (efflux pump, antibiotics inactivation by enzymes, alteration of membrane permeability, target modification, and target protection), the roles of various novel proteins/enzymes in the acquisition of antibiotic resistance are constantly being described. They are widely used by clinical bacterial strains, playing a key role in the emergence of resistance. CONCLUSION While most of these have so far received less attention, expanding our understanding of these emerging resistance mechanisms is of crucial importance to combat the antibiotic resistance crisis in the world. This review summarizes recent advances in our knowledge of emerging resistance mechanisms in bacteria, providing an update on the current antibiotic resistance threats and encouraging researchers to develop critical strategies for overcoming the resistance.
Collapse
Affiliation(s)
- Shengwei Sun
- School of Engineering Sciences in Chemistry, Biotechnology and Health, Department of Fibre and Polymer Technology, KTH Royal Institute of Technology, Stockholm, SE-100 44, Sweden.
- School of Engineering Sciences in Chemistry, Biotechnology and Health, Science for Life Laboratory, Tomtebodavägen 23, Solna, 171 65, Sweden.
| |
Collapse
|
37
|
Zhong Y, Chen F, Chen D, He Q, Zhang X, Lan L, Yang C. Design, synthesis and optimization of TarO inhibitors as multifunctional antibiotics against Methicillin-resistant Staphylococcus aureus. NPJ ANTIMICROBIALS AND RESISTANCE 2025; 3:28. [PMID: 40221595 PMCID: PMC11993615 DOI: 10.1038/s44259-025-00098-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 03/27/2025] [Indexed: 04/14/2025]
Abstract
UDP-N-acetylglucosamine-undecaprenyl-phosphate N-acetylglucosaminephosphotransferase (TarO) has been found to simultaneously contribute to β-lactam resistance and virulence of Methicillin-resistant Staphylococcus aureus (MRSA). However, optimization of hit compounds targeting TarO has been hindered due to their high lipophilicity and the poor correlation between the enzyme activity inhibition and β-lactam sensitization. In this study, 31 analogues of Tarocin A were designed, synthesized and evaluated by a luminescence-based reporter preliminary screening. In the subsequent β-lactams synergy test, a good correlation was observed between the results obtained from these two methods. Finally, analog 18a with more potential against TarO and an improved hydrophilicity (clogP = 3.2) was obtained. Compared with Tarocin A, 18a shows stronger β-lactam sensitizing and anti-biofilm activities in vitro, as well as potent anti-virulence and synergistic potency with imipenem in vivo. These results suggest that TarO is a promising target for combating MRSA, and 18a can serve as a lead molecule.
Collapse
Affiliation(s)
- Yuanchen Zhong
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, 310024, Hangzhou, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, 201203, Shanghai, China
| | - Feifei Chen
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, 310024, Hangzhou, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, 201203, Shanghai, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Dianyan Chen
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, 310024, Hangzhou, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Qian He
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, 201203, Shanghai, China
| | - Xiaofei Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, 201203, Shanghai, China.
- University of Chinese Academy of Sciences, 100049, Beijing, China.
| | - Lefu Lan
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, 310024, Hangzhou, China.
- University of Chinese Academy of Sciences, 100049, Beijing, China.
| | - Chunhao Yang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, 201203, Shanghai, China.
- University of Chinese Academy of Sciences, 100049, Beijing, China.
| |
Collapse
|
38
|
Wu M, Nie Q, Zhang Y, Qin J, Ye L, Zhao R, Dai M, Wu M. METTL3 Plays Regulatory Roles in Acute Pneumonia during Staphylococcus aureus Infection. ACS Infect Dis 2025; 11:905-916. [PMID: 40105125 DOI: 10.1021/acsinfecdis.4c00938] [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: 03/20/2025]
Abstract
Pneumonia caused by Staphylococcus aureus infection has consistently been a significant cause of morbidity and mortality worldwide. Extensive research to date indicates that N6-methyladenosine (m6A) modification plays a crucial role in the development and progression of various diseases. However, it remains unknown whether the m6A modification affects the progression of bacterial pneumonia. To explore this question, we assessed the levels of m6A as well as the expression of methyltransferases (METTL3 and METTL14), demethylase fat mass and obesity-related protein (FTO), and methylation reader proteins YTHDF1 and YTHDF2 in mice and MH-S cells during S. aureus infection. The levels of m6A and METTL3 were significantly upregulated in S. aureus-infected mice and MH-S cells. siMETTL3 knockdown resulted in more severe bacterial colonization, lung damage, increased inflammatory cytokines (IL-6, IL-1β, TNF-α), and mortality rates in mice as well as MH-S cells following the bacterial infection. Regulation of lung inflammation levels by METTL3 was associated with the activation of the MAPK/NF-κB/JAK2-STAT3 signaling pathway. Moreover, siMETTL3 mice exhibited an increased release of superoxides and exacerbated oxidative stress in the lungs following S. aureus infection, which was correlated with impaired mitochondrial autophagy mediated by the Pink1/Parkin pathway. Our findings provide previously unrecognized evidence of the protective role of METTL3 in S. aureus-induced acute pneumonia, indicating a potential therapeutic target for S. aureus infections.
Collapse
Affiliation(s)
- Menghui Wu
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
| | - Qihang Nie
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, College of Life Science, Northwest University, Xi'an 710000, China
| | - Yanyan Zhang
- School of Medicine, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Jiaoxia Qin
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Liumei Ye
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Ruoyang Zhao
- Wenzhou Institute, University of the Chinese Academy of Sciences, Wenzhou 325024, China
| | - Menghong Dai
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
| | - Min Wu
- Wenzhou Institute, University of the Chinese Academy of Sciences, Wenzhou 325024, China
| |
Collapse
|
39
|
Xu T, Xue Z, Li X, Zhang M, Yang R, Qin S, Guo Y. Development of Membrane-Targeting Osthole Derivatives Containing Pyridinium Quaternary Ammonium Moieties with Potent Anti-Methicillin-Resistant Staphylococcus aureus Properties. J Med Chem 2025; 68:7459-7475. [PMID: 40205941 DOI: 10.1021/acs.jmedchem.4c03167] [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: 04/11/2025]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is a leading cause of hospital- and community-acquired infections, necessitating the development of novel antibacterials. Here, we designed and synthesized 30 osthole derivatives with pyridinium quaternary ammonium moieties. In vitro bioassay showed that compounds 8u and 8ac exhibited potent antibacterial activity against S. aureus ATCC 29213 and ten clinical MRSA isolates (MIC = 0.5-1 μg/mL), with low hemolytic activity, rapid bactericidal effects, and minimal resistance induction. In MRSA-infected mouse models of skin abscesses and sepsis, 8u and 8ac also displayed excellent antibacterial effects and safety, which were comparable to vancomycin. Mechanistic studies revealed that 8u and 8ac selectively target bacterial membranes via binding to phosphatidylglycerol (PG), increasing intracellular reactive oxygen species (ROS), inducing content leakage, and ultimately causing bacterial death. These findings suggest 8u and 8ac as promising novel lead candidates for anti-MRSA drug development.
Collapse
Affiliation(s)
- Ting Xu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, Hunan Province, China
| | - Zihan Xue
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| | - Xinhui Li
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| | - Miaomiao Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| | - Ruige Yang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, Hunan Province, China
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| | - Shangshang Qin
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| | - Yong Guo
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, Hunan Province, China
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| |
Collapse
|
40
|
Maraolo AE, Gatti M, Principe L, Marino A, Pipitone G, De Pascale G, Ceccarelli G. Management of methicillin-resistant Staphylococcus aureus bloodstream infections: a comprehensive narrative review of available evidence focusing on current controversies and the challenges ahead. Expert Rev Anti Infect Ther 2025:1-26. [PMID: 40165471 DOI: 10.1080/14787210.2025.2487163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 03/18/2025] [Accepted: 03/21/2025] [Indexed: 04/02/2025]
Abstract
INTRODUCTION Bloodstream infections (BSIs) caused by Staphylococcus aureus are common worldwide, representing one of the most relevant issues in clinical infectious diseases practice. In particular, BSIs by methicillin-resistant S. aureus (MRSA-BSI) are still today a challenge since mortality burden remains elevated although decades of research. AREAS COVERED The following topics regarding MRSA-BSI were reviewed and discussed by resorting to best available evidence retrieved from PubMed/MEDLINE up to October 2024: i) epidemiology; ii) microbiology; iii) classification, with a focus on complicated and not complicated forms; iv) the structured approach to the patient; v) pharmacokinetics and pharmacodynamics of the main antimicrobial options; vi) controversies regarding the best therapeutic approach. EXPERT OPINION Despite ongoing efforts to better stratify and manage MRSA-BSI, there is no universally accepted classification system accurately distinguishing between uncomplicated/low risk and complicated/high risk forms. Biomarkers such as interleukin(IL)-10 hold promise in order to enable a more precise stratification, premise for an appropriate treatment plan. There is a theoretical rationale for implementing a combination therapy including a beta-lactam agent upfront, especially for patients considered at higher risk of unfavorable outcomes, but further data are necessary, and the same applies to newer adjuvants. Novel microbiological techniques may help in guiding antimicrobial duration.
Collapse
Affiliation(s)
- Alberto Enrico Maraolo
- Section of Infectious Diseases, Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | - Milo Gatti
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, Bologna, Italy
- Clinical Pharmacology Unit, Department for Integrated Infectious Risk Management, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Luigi Principe
- Microbiology and Virology Unit, Great Metropolitan Hospital "Bianchi-Melacrino-Morelli", Reggio Calabria, Italy
| | - Andrea Marino
- Department of Clinical and Experimental Medicine, Infectious Diseases Unit, ARNAS Garibaldi Hospital, University of Catania, Catania, Italy
| | | | - Gennaro De Pascale
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Rome, Italy
- Dipartimento di Scienze dell 'Emergenza, Anestesiologiche e della Rianimazione, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Giancarlo Ceccarelli
- Department of Public Health and Infectious Diseases, University Hospital Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
41
|
Enroth TJ, Severn MM, Costa FG, Bovee AR, Wilkening RV, Nguyen DT, Langouët-Astrié C, Horswill AR. Global changes in Staphylococcus aureus virulence and metabolism during colonization of healthy skin. Infect Immun 2025; 93:e0002825. [PMID: 40116525 PMCID: PMC11977313 DOI: 10.1128/iai.00028-25] [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: 01/22/2025] [Accepted: 02/15/2025] [Indexed: 03/23/2025] Open
Abstract
Staphylococcus aureus and its antibiotic-resistant derivative, methicillin-resistant S. aureus (MRSA), are the leading causative agents of skin and soft tissue infections globally. S. aureus transiently colonizes the skin of healthy adults, and this transient colonization likely precedes an active infection. In recent years, there have been efforts to elucidate specific factors that help MRSA transition to an active infection, but the specific genetic determinants required for this transition following skin colonization are largely unknown. To address this question, we developed a model of asymptomatic colonization of mouse skin by MRSA. From this model, we could determine the MRSA and mouse transcriptional profiles by RNA sequencing (RNAseq) at 5- and 24-hour post-colonization. The fadXDEBA locus, required for fatty acid metabolism, was highly upregulated in our data, as were numerous virulence factors. RNAseq data were confirmed via functional in vitro and in vivo promoter-fusion assays using live bioluminescent imaging of the fadXDEBA locus promoter driving fadB transcription. We analyzed the functional capacity of members of the fadXDEBA locus, which encode crucial enzymatic components of the S. aureus β-oxidation pathway. The genes fadD and fadA modulate MRSA resistance to fosfomycin and other oxidative stressors during growth in the presence of the common skin fatty acid, palmitic acid. Overall, our data demonstrate that there are global changes to the MRSA transcriptome, priming the bacteria for survival by upregulation of known virulence factors and metabolic genes implicated in host skin-nutrient utilization.IMPORTANCEStaphylococcus aureus is a major global agent of skin and soft tissue infections. S. aureus colonizes the skin transiently, an important precursor to infection. However, little is known about how S. aureus adapts to the skin at the transcriptional level. This study provides an overview of the S. aureus transcriptome during mouse skin colonization via RNA sequencing. We identified that the most highly upregulated genes during colonization are related to fatty acid metabolism. The disruption of certain genes in the fatty acid degradation pathway altered resistance of S. aureus to the antibiotic fosfomycin. This study provides an important step in understanding the transcriptional changes that occur during S. aureus skin colonization and may reveal novel targets of therapeutic interest for preventing skin infections.
Collapse
Affiliation(s)
- Timothy J. Enroth
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Morgan M. Severn
- Department of Dermatology, Duke University, Durham, North Carolina, USA
| | - Flavia G. Costa
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Alyson R. Bovee
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Reid V. Wilkening
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
- Section of Pediatric Critical Care Medicine, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Dustin T. Nguyen
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | | | - Alexander R. Horswill
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
- Department of Veterans Affairs Eastern, Colorado Healthcare System, Grand Junction, Colorado, USA
| |
Collapse
|
42
|
Toneto Francisco M, de Jesus Barbosa PH, Ahmad F, Fernandes SA, Giambiagi-deMarval M, Diaz MAN, Rossi CC. Hydrocleys nymphoides (water poppy): a cosmopolitan aquatic plant with a phytochemical-rich extract exhibiting antimicrobial and antibiofilm activity against multidrug-resistant Staphylococcus. Lett Appl Microbiol 2025; 78:ovaf046. [PMID: 40175299 DOI: 10.1093/lambio/ovaf046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/18/2025] [Accepted: 04/01/2025] [Indexed: 04/04/2025]
Abstract
Antimicrobial resistance among Staphylococcus species, including multidrug-resistant and biofilm-forming strains, poses a critical threat to global health, demanding innovative therapeutic solutions. In this context, this study explores the antimicrobial and antibiofilm potential of the aquatic plant Hydrocleys nymphoides as a promising alternative. Extracts from the plant's leaves and roots were obtained using solvents of increasing polarity and tested against five key pathogenic Staphylococcus species: S. aureus, S. epidermidis, S. haemolyticus, S. pseudintermedius, and S. coagulans. The hexane extract of H. nymphoides leaves showed the most notable activity, with inhibition zones of 9-17 mm and minimum inhibitory concentrations (MICs) as low as 0.8 mg/ml for certain strains. Subinhibitory concentrations of the extract significantly reduced biofilm formation in most strains, with reductions up to 46.9%. Gas chromatography-mass spectrometry revealed bioactive compounds such as linoleic acid, n-hexadecanoic acid, 9-octadecenal, eicosane, and tetratriacontane, known for their antimicrobial and antibiofilm properties. Although cytotoxicity was observed at concentrations near the MIC, lower concentrations were non-toxic, indicating potential for controlled therapeutic applications. These findings underscore the biotechnological value of H. nymphoides as a sustainable source of antimicrobial agents against multidrug-resistant Staphylococcus. This work emphasizes the critical role of phytotherapy in combating the escalating antimicrobial resistance crisis.
Collapse
Affiliation(s)
- Marcos Toneto Francisco
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal de Viçosa, Viçosa, 36570-000, MG, Brazil
| | | | - Faizan Ahmad
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal de Viçosa, Viçosa, 36570-000, MG, Brazil
| | | | - Marcia Giambiagi-deMarval
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-902, RJ, Brazil
| | - Marisa Alves Nogueira Diaz
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal de Viçosa, Viçosa, 36570-000, MG, Brazil
| | - Ciro César Rossi
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal de Viçosa, Viçosa, 36570-000, MG, Brazil
| |
Collapse
|
43
|
Burillo A, Bouza E. Community-acquired methicillin-resistant Staphylococcus aureus : is it still a significant pathogen for skin and soft tissue infections? A 30-year overview. Curr Opin Infect Dis 2025; 38:78-91. [PMID: 39945402 DOI: 10.1097/qco.0000000000001086] [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: 02/28/2025]
Abstract
PURPOSE OF REVIEW The prevalence of community-acquired methicillin-resistant Staphylococcus aureus (CA-MRSA) in skin and soft tissue infections (SSTI) has significantly changed in recent decades. We conducted a literature review to determine whether this microorganism, which became increasingly common as a cause of SSTI in the 2000s, still plays a significant role in these infections today. RECENT FINDINGS Over the past 30 years, there has been a pattern of increase and then decrease in these infections. The highest frequency was observed in the United States, to the extent that guidelines recommended empirical antibiotic treatment for this pathogen in SSTI. Clone USA300 is the primary causative agent in the United States. In Europe, SSTI are much less common than in the United States, and the presence of this clone has been significantly lower. A decrease in the frequency of SSTI and CA-MRSA has been observed in developed countries. However, the spread of specific clones in Latin America, Asia and Africa highlights the need for rigorous global surveillance. SUMMARY In recent years, the prevalence of CA-MRSA SSTI has decreased in developed countries. However, globalisation, immigration and intercontinental travel have favoured the spread of some clones with epidemic potential. It remains to be seen whether the current lower frequency will be maintained or whether these clones will give rise to a new wave.
Collapse
Affiliation(s)
- Almudena Burillo
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón
- Medicine Department, School of Medicine, Universidad Complutense de Madrid
- Gregorio Marañón Health Research Institute (IiSGM)
| | - Emilio Bouza
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón
- Medicine Department, School of Medicine, Universidad Complutense de Madrid
- Gregorio Marañón Health Research Institute (IiSGM)
- CIBER of Respiratory Diseases (CIBERES CB06/06/0058), Madrid, Spain
| |
Collapse
|
44
|
Barman P, Sharma C, Joshi S, Sharma S, Maan M, Rishi P, Singla N, Saini A. In Vivo Acute Toxicity and Therapeutic Potential of a Synthetic Peptide, DP1 in a Staphylococcus aureus Infected Murine Wound Excision Model. Probiotics Antimicrob Proteins 2025; 17:843-856. [PMID: 37910332 DOI: 10.1007/s12602-023-10176-1] [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] [Accepted: 10/10/2023] [Indexed: 11/03/2023]
Abstract
Bacterial infections at the surgical sites are one of the most prevalent skin infections that impair the healing mechanism. They account for about 20% of all types of infections and lead to approximately 75% of surgical-site infection-associated mortality. Several antibiotics, such as cephalosporins, fluoroquinolones, quinolones, penicillin, sulfonamides, etc., that are used to treat such wound infections not only counter infections but also disrupt the normal flora. Moreover, antibiotics, when used for a prolonged duration, may impair the formation of new blood vessels, delay collagen production, or inhibit the migration of certain cells involved in wound repair, leading to an impaired healing process. Therefore, there is a dire need for alternate therapeutic approaches against such infections. Antimicrobial peptides have gained considerable attention as a promising strategy to counter these pathogens and prevent the spread of infection. Recently, we have reported a designed peptide, DP1, and its broad-spectrum in vitro antimicrobial activity against Gram-positive and Gram-negative bacteria. In the present study, in vivo acute toxicity of DP1 was evaluated and even at a high dose (20 mg/kg body weight) of DP1, a 100% survival of mice was observed. Subsequently, a Staphylococcus aureus-infected murine wound excision model was established to assess the wound healing efficacy of DP1. The study revealed significant wound healing vis-a-vis attenuated S. aureus bioburden at the wound site and also controlled the oxidative stress depicting anti-oxidant activity as well. Healing of the infected wounds was also verified by histopathological examination. Based on the results of this study, it can be concluded that DP1 improves wound resolution despite infections and promotes the healing mechanism. Hence, DP1 holds compelling potential as a novel antimicrobial drug that requires further explorations in clinical platforms.
Collapse
Affiliation(s)
- Panchali Barman
- Institute of Forensic Science and Criminology (UIEAST), Panjab University, Chandigarh, 160014, India
| | - Chakshu Sharma
- Department of Biophysics, Panjab University, Chandigarh, U.T, 160014, India
| | - Shubhi Joshi
- Department of Biophysics, Panjab University, Chandigarh, U.T, 160014, India
| | - Sheetal Sharma
- Department of Biophysics, Panjab University, Chandigarh, U.T, 160014, India
| | - Mayank Maan
- Department of Biophysics, Panjab University, Chandigarh, U.T, 160014, India
| | - Praveen Rishi
- Department of Microbiology, Panjab University, Chandigarh, U.T, 160014, India
| | - Neha Singla
- Department of Biophysics, Panjab University, Chandigarh, U.T, 160014, India
| | - Avneet Saini
- Department of Biophysics, Panjab University, Chandigarh, U.T, 160014, India.
| |
Collapse
|
45
|
Cashman-Kadri S, Fliss I, Beaulieu L, Lagüe P. Ergosterol depletion by fish AMP analogs likely enhances fungal membrane permeability. Biophys J 2025; 124:1105-1116. [PMID: 40007119 PMCID: PMC11993923 DOI: 10.1016/j.bpj.2025.02.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 01/30/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025] Open
Abstract
The molecular interactions between a fungal membrane model and SJGAP, a 32-amino-acid antimicrobial peptide (AMP) derived from skipjack tuna GAPDH, as well as four analogs, were investigated using molecular dynamics simulations and Fourier transform infrared (FTIR) spectroscopy. In a previous study, Analog 7, modified by replacing three alanine residues with leucine residues, exhibited unique antifungal activity without any antibacterial effect. This contrasts with other analogs, which showed both antifungal and antibacterial effects. In this study, Analog 7 displayed the strongest interactions with the membrane's hydrophobic core, inserting more deeply and causing significantly greater membrane deformation and thinning compared with the other analogs. Its presence caused significant membrane deformation, evident from the displacement of both the phosphate groups and terminal methyls of the lipids. Notably, Analog 7 was the only analog to induce a marked depletion of ergosterol around the peptide insertion site. FTIR spectroscopy experiments further confirmed the distinctive impact of Analog 7 on a fungal membrane model. The combined results from molecular dynamics simulations and spectroscopy emphasize the critical role of leucine substitutions in Analog 7, particularly at residues 18 and 19 within the central α helix, in promoting membrane thinning and inducing ergosterol depletion, suggesting increased membrane permeabilization, which could explain its previously reported antifungal specificity. This study provides the first insights into the molecular interactions between a GAPDH-derived AMP and a fungal membrane model, offering valuable information about its antifungal mechanism of action.
Collapse
Affiliation(s)
- Samuel Cashman-Kadri
- Institute of Nutrition and Functional Foods (INAF), Université Laval, Québec, Quebec, Canada; Department of Food Science, Faculty of Agricultural and Food Sciences, Université Laval, Québec, Quebec, Canada; Québec-Océan, Université Laval, Québec, Quebec, Canada
| | - Ismail Fliss
- Institute of Nutrition and Functional Foods (INAF), Université Laval, Québec, Quebec, Canada; Department of Food Science, Faculty of Agricultural and Food Sciences, Université Laval, Québec, Quebec, Canada
| | - Lucie Beaulieu
- Institute of Nutrition and Functional Foods (INAF), Université Laval, Québec, Quebec, Canada; Department of Food Science, Faculty of Agricultural and Food Sciences, Université Laval, Québec, Quebec, Canada; Québec-Océan, Université Laval, Québec, Quebec, Canada
| | - Patrick Lagüe
- Department of Biochemistry, Microbiology and Bioinformatics, Pavillon Alexandre-Vachon, Université Laval, Québec, Quebec, Canada; Institute for Integrative Systems Biology, Université Laval, Québec, Quebec, Canada; The Quebec Network for Research on Protein Function, Engineering, and Applications (PROTEO), Montréal, Quebec, Canada.
| |
Collapse
|
46
|
Iszatt JJ, Larcombe AN, Garratt LW, Stick SM, Kicic A. Lytic activity, stability, biofilm disruption capabilities, and genomic characterization of two bacteriophages active against respiratory MRSA. J Appl Microbiol 2025; 136:lxaf081. [PMID: 40180588 DOI: 10.1093/jambio/lxaf081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 02/20/2025] [Accepted: 04/10/2025] [Indexed: 04/05/2025]
Abstract
AIMS This study aimed to characterize bacteriophages for potential therapeutic use against Staphylococcus aureus, focusing on clinical respiratory isolates of methicillin-sensitive (MSSA) and methicillin-resistant (MRSA) strains. Specifically, it sought to evaluate phage lytic activity, host range, stability, biofilm disruption capabilities, and overall safety for therapeutic use. METHODS AND RESULTS Novel phages, Koomba kaat 1 and Biyabeda mokiny 1, were identified and characterized using microbiological assays and bioinformatics. They exhibited lytic activity against clinical MSSA and MRSA isolates, disrupted biofilms from airway isolates, remained stable for at least one year in storage, and could be aerosolized without significant reductions in activity. Bioinformatic tools were used to assess safety, lifecycle, virulence, and prophage contamination when grown using their original isolation host. Receptor binding proteins within their genomes were also predicted, providing insight into their mechanisms of action. Both phages demonstrated strong efficacy against the clinical isolates tested and demonstrated robust stability under storage and delivery conditions. CONCLUSIONS Koomba kaat 1 and Biyabeda mokiny 1 are promising candidates for phage therapy. Their efficacy against clinical S. aureus isolates, ability to break down biofilm, and stability for airway implementation, positions them as valuable tools for addressing persistent airway infections caused by S. aureus.
Collapse
Affiliation(s)
- Joshua J Iszatt
- Occupation, Environment and Safety, School of Population Health, Curtin University, Perth 6102, Australia
- Wal-yan Respiratory Research Centre, The Kids Research Institute Australia, Perth 6009, Australia
| | - Alexander N Larcombe
- Occupation, Environment and Safety, School of Population Health, Curtin University, Perth 6102, Australia
- Wal-yan Respiratory Research Centre, The Kids Research Institute Australia, Perth 6009, Australia
| | - Luke W Garratt
- Occupation, Environment and Safety, School of Population Health, Curtin University, Perth 6102, Australia
- Wal-yan Respiratory Research Centre, The Kids Research Institute Australia, Perth 6009, Australia
- Centre for Child Health Research, University of Western Australia, Perth 6009, Australia
| | - Stephen M Stick
- Centre for Child Health Research, University of Western Australia, Perth 6009, Australia
- Department of Respiratory and Sleep Medicine, Perth Children's Hospital, Perth, 6009, Australia
| | - Anthony Kicic
- Occupation, Environment and Safety, School of Population Health, Curtin University, Perth 6102, Australia
- Wal-yan Respiratory Research Centre, The Kids Research Institute Australia, Perth 6009, Australia
- Centre for Child Health Research, University of Western Australia, Perth 6009, Australia
| |
Collapse
|
47
|
Singh S, Kaul G, Shukla M, Akhir A, Tripathi S, Gupta A, Bormon R, Nair NN, Chopra S, Verma S. Linear Antimicrobial Peptide, Containing a Diindolyl Methane Unnatural Amino Acid, Potentiates Gentamicin Against Methicillin-Resistant Staphylococcus aureus. Drug Dev Res 2025; 86:e70070. [PMID: 40025838 DOI: 10.1002/ddr.70070] [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: 08/10/2024] [Revised: 11/22/2024] [Accepted: 02/17/2025] [Indexed: 03/04/2025]
Abstract
The headway for the management of emerging resistant microbial strains has become a demanding task. Over the years, antimicrobial peptides (AMP), have been recognized and explored for their highly systematized SAR and antibacterial properties. With this background, we have reported a new class of AMPs. These peptides incorporate an unnatural amino acid, with a motivation from cruciferous bioactive phytochemical bisindoles methane derivatives with highly selective antimicrobial action. These peptides may also be considered as linear derivatives of hirsutide isolated from entomopathogenic fungus. The synthesized peptides were tested for their antimicrobial activity against an ESKAPE pathogen panel, where peptide 3 exhibited equipotent MIC and potent synergistic action along with gentamicin against Staphylococcus aureus and Enterococcus clinical isolates. This combination was also able to repotentiate gentamicin against NRS119, a gentamicin-resistant MRSA. Molecular dynamics study and free energy calculations provided insights to membrane disruptive properties of AMP action, which assisted gentamicin pass through the lipid-water interface.
Collapse
Affiliation(s)
- Shalini Singh
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur, India
| | - Grace Kaul
- Department of Microbiology, CSIR-Central Drug Research Institute, Lucknow, India
- AcSIR: Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Manjulika Shukla
- Department of Microbiology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Abdul Akhir
- Department of Microbiology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Shubhandra Tripathi
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur, India
| | - Abhinav Gupta
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur, India
| | - Rakhi Bormon
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur, India
| | - Nisanth N Nair
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur, India
| | - Sidharth Chopra
- Department of Microbiology, CSIR-Central Drug Research Institute, Lucknow, India
- AcSIR: Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Sandeep Verma
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur, India
- Center for Nanosciences, Centre for Environmental Science and Engineering, Mehta Center for Engineering in Medicine, Gangwal School of Medical Sciences and Technology, Centre for Environmental Science and Engineering, Indian Institute of Technology Kanpur, Kanpur, India
| |
Collapse
|
48
|
Post V, Pascoe B, Hitchings MD, Erichsen C, Fischer J, Morgenstern M, Richards RG, Sheppard SK, Moriarty TF. Methicillin-sensitive Staphylococcus aureus lineages contribute towards poor patient outcomes in orthopaedic device-related infections. Microb Genom 2025; 11. [PMID: 40238650 DOI: 10.1099/mgen.0.001390] [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: 04/18/2025] Open
Abstract
Staphylococci are the most common cause of orthopaedic device-related infections (ODRIs), with Staphylococcus aureus responsible for a third or more of cases. This prospective clinical and laboratory study investigated the association of genomic and phenotypic variation with treatment outcomes in ODRI isolates. Eighty-six invasive S. aureus isolates were collected from patients with ODRI, and clinical outcome was assessed after a follow-up examination of 24 months. Each patient was then considered to have been 'cured' or 'not cured' based on predefined clinical criteria. Whole-genome sequencing and molecular characterization identified isolates belonging to globally circulating community- and hospital-acquired lineages. Most isolates were phenotypically susceptible to methicillin and lacked the staphylococcal cassette chromosome mec cassette [methicillin-susceptible S. aureus (MSSA); 94%] but contained several virulence genes, including toxins and biofilm genes. Whilst recognizing the role of the host immune response, we identified genetic variance, which could be associated with the infection severity or clinical outcome. Whilst this and several other studies reinforce the role antibiotic resistance [e.g. methicillin-resistant S. aureus (MRSA) infection] has on treatment failure, it is important not to overlook MSSA that can cause equally destructive infections and lead to poor patient outcomes.
Collapse
Affiliation(s)
| | - Ben Pascoe
- Ineos Oxford Institute for Antimicrobial Research, Department of Biology, University of Oxford, Oxford, UK
- Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, Arizona, USA
| | | | | | - Julian Fischer
- Centrum of Orthopedic Isartal, Pullach im Isartal, Germany
| | - Mario Morgenstern
- Department of Orthopedic and Trauma Surgery, University Hospital, Basel, Switzerland
| | | | - Samuel K Sheppard
- Ineos Oxford Institute for Antimicrobial Research, Department of Biology, University of Oxford, Oxford, UK
| | - T Fintan Moriarty
- AO Research Institute Davos, Davos, Switzerland
- Department of Orthopedic and Trauma Surgery, University Hospital, Basel, Switzerland
| |
Collapse
|
49
|
Rodriguez A, Rich RL, Semanco M. Utility of MRSA nares PCR for non-respiratory cultures in critically ill patients: an observational evaluation. Infect Dis (Lond) 2025; 57:361-365. [PMID: 39655512 DOI: 10.1080/23744235.2024.2438822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 11/27/2024] [Accepted: 12/02/2024] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND The overuse of antibiotics may lead to complications such as increased resistance, adverse events, and toxicities. Literature demonstrates a negative Methicillin-resistant Staphylococcus aureus (MRSA) nares polymerase chain reaction (PCR) may be used to streamline antibiotic therapy prior to respiratory culture results based on a negative predictive value (NPV) of 95-99%. Additional literature supports a high NPV when MRSA nares PCR is evaluated in non-respiratory cultures; however, this use in critically ill patients has not been studied. OBJECTIVES The purpose of this study was to evaluate the clinical utility of MRSA nares PCR in non-respiratory cultures in critically ill patients. METHODS This was a single centre, retrospective, cohort evaluation. Outcomes evaluated were NPV, positive predictive value (PPV), sensitivity, and specificity of MRSA nares PCR in critically ill patients. A sub-group analysis based on the site of culture (blood, urine, and wound) was also conducted. RESULTS Of the 325 patients screened, 200 critically ill patients were included for analysis. A total of 259 cultures were evaluated with blood being the most common source (n = 124). The MRSA nares PCR was positive in 34 (17%) patients and thirteen (5%) of the 259 cultures were positive for MRSA. For all cultures, the MRSA nares PCR demonstrated an NPV 99%, PPV 28%, sensitivity 77%, and specificity 85%. The subgroup analysis for the individual culture types reflected similar findings. CONCLUSIONS A negative MRSA nares PCR may be used to withhold initiation or allow for timely de-escalation of anti-MRSA antibiotics in critically ill patients if clinically applicable.
Collapse
Affiliation(s)
- Alexa Rodriguez
- Department of Pharmacy, Lakeland Regional Health, Lakeland, FL, USA
| | - Rebecca L Rich
- Department of Pharmacy, Lakeland Regional Health, Lakeland, FL, USA
| | - Michael Semanco
- Department of Pharmacy, Lakeland Regional Health, Lakeland, FL, USA
| |
Collapse
|
50
|
Zhang Q, Leng X, Peng L, Lin H, Xuan G, Zhang W, Mitomo H, Ijiro K, Wang G. Streamlining Bacterial Gene Regulation via Nucleic Acid Delivery with Gold Nanoclusters. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2411723. [PMID: 39989200 DOI: 10.1002/smll.202411723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 02/06/2025] [Indexed: 02/25/2025]
Abstract
Delivery of exogenous nucleic acids (NAs) for gene regulation in bacteria, bypassing the barrier of the cell wall, is essential for advancing fundamental microbiology and genetic engineering, and the treatment of bacterial diseases. However, current methods that rely on electrical or chemical interventions are limited by their complexity, specialized expertise, and laboratory-specific instrumentation. This study explores the capability of gold nanoclusters (AuNCs) as carriers for delivering small-interfering RNA and antisense oligonucleotides into bacteria for targeted gene regulation while shielding them from degradation during transport. By enhancing the cytoplasmic membrane permeability, the AuNCs enable efficient internalization of NAs into both Gram-positive and Gram-negative bacteria while exerting negligible influence on bacterial activity. It is demonstrated that the rationally designed NAs can be released from the AuNCs within bacteria, enabling ~70% knockdown of mecA in Methicillin-resistant Staphylococcus aureus (MRSA). This significantly reduces MRSA's antibiotic resistance and enhances oxacillin treatment efficacy. Furthermore, the successful silencing of ligA in Escherichia coli and pilQ in Pseudomonas aeruginosa highlights the broad adaptability of the approach across diverse bacterial species. The AuNCs-based next-generation NA delivery system has the potential to transform bacterial gene regulation-previously restricted to laboratory settings-into a versatile and scalable solution for real-world application.
Collapse
Affiliation(s)
- Qingsong Zhang
- MOE Key Laboratory of Evolution and Marine Biodiversity and Institute of Evolution and Marine Biodiversity, Ocean University of China, 5 Yushan Road, Qingdao, 266003, China
- SKL of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, 1299 Sansha Road, Qingdao, 266404, China
| | - Xinyi Leng
- MOE Key Laboratory of Evolution and Marine Biodiversity and Institute of Evolution and Marine Biodiversity, Ocean University of China, 5 Yushan Road, Qingdao, 266003, China
- SKL of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, 1299 Sansha Road, Qingdao, 266404, China
| | - Lin Peng
- MOE Key Laboratory of Evolution and Marine Biodiversity and Institute of Evolution and Marine Biodiversity, Ocean University of China, 5 Yushan Road, Qingdao, 266003, China
- SKL of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, 1299 Sansha Road, Qingdao, 266404, China
| | - Hong Lin
- SKL of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, 1299 Sansha Road, Qingdao, 266404, China
| | - Guanhua Xuan
- SKL of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, 1299 Sansha Road, Qingdao, 266404, China
| | - Weiwei Zhang
- School of Marine Sciences, Ningbo University, 169 Qixingnan Road, Ningbo, 315832, China
| | - Hideyuki Mitomo
- Research Institute for Electronic Science, Hokkaido University, Sapporo, 001-0021, Japan
| | - Kuniharu Ijiro
- Research Institute for Electronic Science, Hokkaido University, Sapporo, 001-0021, Japan
| | - Guoqing Wang
- MOE Key Laboratory of Evolution and Marine Biodiversity and Institute of Evolution and Marine Biodiversity, Ocean University of China, 5 Yushan Road, Qingdao, 266003, China
- SKL of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, 1299 Sansha Road, Qingdao, 266404, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Centre, Qingdao, 266237, China
| |
Collapse
|