1
|
Li R, Sun M, Li ZH, Qu Y, Li Y, Ampomah-Wireko M, Li D, Kong H, Wu Y, Hossain AA, Zhang E. Important Role of Triphenylamine in Modulating the Antibacterial Performance Relationships of Antimicrobial Peptide Mimics by Alkyl Chain Engineering. J Med Chem 2025; 68:10299-10313. [PMID: 40270226 DOI: 10.1021/acs.jmedchem.5c00454] [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: 04/25/2025]
Abstract
Multidrug resistance (MDR) bacteria pose a serious threat to human health, and the development of effective antimicrobial drugs is urgent. Herein, we used alkyl chain engineering to design and synthesize two series of antimicrobial peptide mimics with distinct cores: triphenylamine quaternary ammonium derivatives (TPQs) and diphenylethene quaternary ammonium derivatives (BPQs), and we investigated the effect of varying the alkyl chain lengths on antibacterial activity. We found that the introduction of a triphenylamine group significantly enhances the antibacterial activity of short-chain dimethyl quaternary ammonium derivatives while maintaining their excellent biocompatibility. Most notably, TPQ-1 exhibited negligible invasiveness toward living cells and possesses good antimicrobial activities, with good efficacy against biofilms and persisters. Moreover, TPQ-1 exhibited good antimicrobial effects in vivo and significantly accelerated the healing process of methicillin-resistant Staphylococcus aureus-infected wounds. This work promotes the practical application of antimicrobial peptide mimics and triphenylamine derivatives.
Collapse
Affiliation(s)
- Ruirui Li
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, China
| | - Moran Sun
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, China
| | - Zhi-Hao Li
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, PR China
| | - Ye Qu
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, China
| | - Yuanbo Li
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, China
| | - Maxwell Ampomah-Wireko
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, China
| | - Daran Li
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, China
| | - Hongtao Kong
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, China
| | - Yuequan Wu
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, China
| | - Adib Azwad Hossain
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, China
| | - En Zhang
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, China
- Pingyuan Laboratory (Zhengzhou University), Zhengzhou 450001, China
| |
Collapse
|
2
|
Agrahari B, Chaudhary K, Dewan S, Sonker H, S V A, Chandra A, Awasthi N, Makari H, Sinharay S, Singh RG. Theragnostic Dual-Action Platform: Ruthenium p-Cymene-Derived Metalloantibiotics With NIR-II Photoacoustic Spectral Signal. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2503986. [PMID: 40370269 DOI: 10.1002/smll.202503986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2025] [Revised: 04/10/2025] [Indexed: 05/16/2025]
Abstract
This study initiates the development of Ru1, a metalloantibiotic derived from p-cymene, which delivers dual functionality. Our approach begins with the design and synthesis of half-sandwich ruthenium complexes, Ru1-Ru11. By varying the structure of the ancillary ligand, the biological activities of eleven ruthenium complexes against five bacterial strains were explored. Most active, Ru1 (4 µg mL-1) and Ru 11 (1 µg mL-1) was solicited for further study. To assess the safety of these compounds, hemolytic activity and cytotoxicity were evaluated against human red blood cells (RBCs) and the HEK cell line, respectively. Based on these results, Ru1 was selected for further investigation. Ru1 exhibited significant efficacy against MRSA (methicillin-resistant S. aureus) and VRSA (vancomycin-resistant S. aureus). Additionally, Ru1 exhibited strong DNA-binding affinity (Kb = 1.73*105 M-1 and 1.6 *105 M-1). Molecular docking studies further highlighted Ru1's potent interaction with the active sites of key bacterial proteins, including S. aureus MurB, topoisomerase II DNA gyrase, and 1BNA, with binding energies of -12.05 kcal/mol, -6.30 kcal/mol and -7.77 kcal/mol, respectively. Furthermore, Ru1 showed high photoacoustic signal intensity at 902 nm in MSOT imaging, underscoring its potential as a dual-function therapeutic and diagnostic (theragnostic) agent.
Collapse
Affiliation(s)
| | | | - Sayari Dewan
- Department of Chemistry, IIT Kanpur, Kanpur, 208016, India
| | | | - Arjun S V
- Department of Bioengineering, IISc, Bengaluru, 560012, India
| | - Anshuman Chandra
- School of Physical Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Nidhi Awasthi
- Department of Chemistry, IIT Kanpur, Kanpur, 208016, India
| | - Hanumanthappa Makari
- Maharani's Science College for Women, University of Mysore, Mysore, 570005, India
| | | | | |
Collapse
|
3
|
Phat VV, Lim AST, De Cozar-Gallardo C, Alvaro MIC, Alvarez DM, Alvaro EF, Ballell-Pages L, Lozano-Arias S, Baker S. A three-dimensional high throughput assay identifies novel antibacterial molecules with activity against intracellular Shigella. NPJ ANTIMICROBIALS AND RESISTANCE 2025; 3:40. [PMID: 40374850 DOI: 10.1038/s44259-025-00110-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 04/27/2025] [Indexed: 05/18/2025]
Abstract
The Gram-negative bacterial species Shigella is the second leading cause of diarrhea among children in low and middle-income countries (LMICs) and is a World Health Organization (WHO) priority pathogen. Shigella infections are becoming increasing difficult to treat due to antimicrobial resistance (AMR), leading to an urgent need for new antimicrobial agents with novel modes of action. Shigella pathogenesis is largely intracellular and antibacterial chemicals that preferentially work inside cells may be desirable to limit collateral AMR and block key components of the Shigella infection cycle. Aiming to facilitate the process of identifying antibacterial chemicals that kill intracellular Shigella, we developed a high-throughput screening (HTS) cell-based chemical screening assay. The three-dimensional (3-D) assay, incorporating Shigella invasion into Caco-2 cells on Cytodex 3 beads, was scaled into a 384-well platform for screening chemical compound libraries. Using this assay, we evaluated >500,000 compounds, identifying 12 chemical hits that inhibit Shigella replication inside cells. This simple, efficient and HTS-compatible assays circumvents many of the limitations of traditional screening methods with cell monolayers and may be deployed for antibacterial compound screening for other intracellular pathogens.
Collapse
Affiliation(s)
- Voong Vinh Phat
- The Hospital for Tropical Diseases, Wellcome Trust Major Overseas Programme, Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
| | - Andrew Shih Teong Lim
- The ALBORADA Drug Discovery Institute, University of Cambridge, Cambridge, United Kingdom
| | | | | | | | | | | | | | - Stephen Baker
- A*STAR Infectious Diseases Labs (A*STAR IDL), Agency for Science, Technology and Research (A*STAR), Singapore, 138648, Singapore.
| |
Collapse
|
4
|
Xie W, Luo D, Wu M, Sun Y, Wang Z. The evaluation of Phenylalanine-tRNA ligase beta unit (PheT), as a potential target in Mycobacterium abscessus. Tuberculosis (Edinb) 2025; 152:102626. [PMID: 40043507 DOI: 10.1016/j.tube.2025.102626] [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: 10/24/2024] [Revised: 02/22/2025] [Accepted: 02/28/2025] [Indexed: 05/04/2025]
Abstract
Mycobacterium abscessus (M. abscessus) is an emerging pathogenic mycobacterium that mainly causes pulmonary infections, especially in immunocompromised patients. This bacterium shows exhibits intrinsic resistance to many anti-tuberculosis drugs, posing significant challenges for both patients and clinicians, thereby raising the need for innovative drug discovery. In this study, we selected phenylalanine-tRNA ligase beta unit (PheT) as a model target and used CRISPR interference to evaluate its essentiality as a therapeutic target against M. abscessus. The results show that genetically disruption of PheT leads to clear growth inhibitory phenotypes both in vitro and in vivo. Further transcriptome analysis revealed differential expression of host genes in response to PheT gene silencing, including genes involved in the cell cycle, apoptotic signaling, and inflammatory responses. Overall, PheT gene plays a crucial role in M. abscessus infection, and its silencing may represent a druggable therapeutic strategy for treating this infection.
Collapse
Affiliation(s)
- Weile Xie
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, China; Collaborative Innovation Center of Agri-Seeds / School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Dan Luo
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, China; Collaborative Innovation Center of Agri-Seeds / School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Mingqing Wu
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, China; Collaborative Innovation Center of Agri-Seeds / School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yicheng Sun
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, and State Key Laboratory of Respiratory Health and Multimorbidity, Center for Tuberculosis Research, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 102629, China
| | - Zhe Wang
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, China; Collaborative Innovation Center of Agri-Seeds / School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
5
|
Ge M, Xiong W, Zeng H, Su H, Wang X, Zhao D, Du X, Wen L. Oriented Channel Functionalization in Covalent Organic Framework Fibers for Boosting the Antibiotics Removal from Environmental Water. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2500711. [PMID: 40183985 DOI: 10.1002/smll.202500711] [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: 01/19/2025] [Revised: 03/06/2025] [Indexed: 04/05/2025]
Abstract
The excessive presence of antibiotics in water is a significant social concern, as it poses serious health risks to humans, necessitating the urgent development of effective removal methods. Herein, an interfacial polymerization method is presented to fabricate a caterpillar-like covalent organic frameworks (COF) platform with branch buds (Tp-Bpy) and utilize a post-modified method to modulate the environment of channels. The Tp-Bpy channels grafted with Cu ions and ether-oxygen chains (Mae) afforded more recognition sites and inner hindrance, thereby enhancing antibiotic removal capacity and efficiency through synergistic interactions and controlled analyte diffusion. The Cu@Tp-Bpy-Mae exhibited significantly higher removal capacities (412.79-435.49 mg g-1) for four antibiotics, far surpassing those of Tp-Bpy, Cu@Tp-Bpy, and other documented material, due to synergistic interactions of electrostatic forces, π-π interactions, coordination bonding, and hydrogen bonding. More importantly, Cu@Tp-Bpy-Mae is capable of treating real wastewater to antibiotic concentrations below 0.02 mg L-1 under continuous flow conditions, effectively mitigating drinking water risks caused by high antibiotic levels. This study offers a facile method for tailoring material properties to optimize antibiotic removal performance and exhibits great potential in environmental pollutant removal.
Collapse
Affiliation(s)
- Miaoxiu Ge
- The Research Institute of Advanced Technology, College of Food Science and Engineering, Ningbo University, Ningbo, 315211, P. R. China
| | - Wei Xiong
- The Research Institute of Advanced Technology, College of Food Science and Engineering, Ningbo University, Ningbo, 315211, P. R. China
- China Innovation Instrument Co., Ningbo, 315100, P. R. China
| | - Hongping Zeng
- The Research Institute of Advanced Technology, College of Food Science and Engineering, Ningbo University, Ningbo, 315211, P. R. China
| | - Hang Su
- The Research Institute of Advanced Technology, College of Food Science and Engineering, Ningbo University, Ningbo, 315211, P. R. China
| | - Xiangyu Wang
- The Research Institute of Advanced Technology, College of Food Science and Engineering, Ningbo University, Ningbo, 315211, P. R. China
| | - Dan Zhao
- China Innovation Instrument Co., Ningbo, 315100, P. R. China
| | - Xiaoming Du
- College of Pharmaceutical Engineering and Biotechnology, College of Cosmetics, Zhejiang Pharmaceutical University, Ningbo, 315100, P. R. China
| | - Luhong Wen
- The Research Institute of Advanced Technology, College of Food Science and Engineering, Ningbo University, Ningbo, 315211, P. R. China
- China Innovation Instrument Co., Ningbo, 315100, P. R. China
| |
Collapse
|
6
|
Chen Y, Li D, Chen X, Wang D, Huang Y, Gao Y, Liu F, Lin X, Zhao D, Ji J, Wang D, Jin Q. Side-Chain Engineering of NIR-II-Emissive Aggregation-Induced Emission Luminogens to Boost Photodynamic and Photothermal Antimicrobial Therapy. ACS NANO 2025; 19:16147-16162. [PMID: 40247726 DOI: 10.1021/acsnano.5c04175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
The development of antibiotic resistance has made multidrug-resistant bacterial and fungal infections one of the most serious health problems worldwide. Photothermal therapy (PTT) and photodynamic therapy (PDT) have received increasing attention in antimicrobial fields due to their precision treatment and less susceptibility to inducing resistance. In particular, developing second near-infrared (NIR-II, 1000-1700 nm)-emissive semiconducting polymers for phototheranostics is highly desirable but remains challenging due to the lack of rational molecular design guidelines. Herein, a precise side-chain engineering strategy based on donor-acceptor (D-A)-type semiconductor polymers is developed for antimicrobial phototherapy. By subtle regulation of the side-chain flexibility, a series of NIR-II-emissive polymer aggregation-induced-emission (AIE) luminogens (AIEgens) are constructed. The optimal polymer PIDT(He)TBT bearing flexible side chains shows optimal physicochemical properties, including the highest mass extinction coefficient, the best AIE property, red-shifted absorption/emission spectra, and desirable photodynamic and photothermal effects. PIDT(He)TBT is then encapsulated into nanoparticles to endow them with water solubility, excellent photostability, and enhanced type-I photodynamic and photothermal effects. The excellent performance of PIDT(He)TBT nanoparticles in terms of fluorescence-guided type-I PDT and PTT of bacterial and fungal infections has been demonstrated both in vitro and in vivo. This work brings useful insights into designing NIR-II-emissive semiconducting polymer AIEgens for highly efficient phototheranostics.
Collapse
Affiliation(s)
- Yongcheng Chen
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310058, P. R. China
| | - Dan Li
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, College of Materials Science and Engineering, Shenzhen University, Shenzhen 518060, China
| | - Xiaohui Chen
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China
| | - Deliang Wang
- Department of Materials Chemistry, Huzhou University, Huzhou 313000, China
| | - Yue Huang
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310058, P. R. China
| | - Yifan Gao
- Suzhou Sanegene Bio Inc., Room 301, Building 2, Zone B, Phase III of BioBAY, No. 99 Jingu Road, Suzhou Industrial Park, Suzhou 215028, China
| | - Fang Liu
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310058, P. R. China
| | - Xintong Lin
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, College of Materials Science and Engineering, Shenzhen University, Shenzhen 518060, China
| | - Daiguo Zhao
- Sichuan Institute for Drug Control, Chengdu 611731, China
| | - Jian Ji
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310058, P. R. China
| | - Dong Wang
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, College of Materials Science and Engineering, Shenzhen University, Shenzhen 518060, China
| | - Qiao Jin
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310058, P. R. China
| |
Collapse
|
7
|
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
|
8
|
Montúfar-Romero M, Valenzuela-Miranda D, Valenzuela-Muñoz V, Morales-Rivera MF, Gallardo-Escárate C. Microbiota Dysbiosis in Mytilus chilensis Is Induced by Hypoxia, Leading to Molecular and Functional Consequences. Microorganisms 2025; 13:825. [PMID: 40284661 PMCID: PMC12029581 DOI: 10.3390/microorganisms13040825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 03/22/2025] [Accepted: 03/29/2025] [Indexed: 04/29/2025] Open
Abstract
Bivalve microbiota play a vital role in host health, supporting nutrient processing, immunity, and disease resistance. However, the increasing hypoxia in Chilean coastal waters, caused by climate change and eutrophication, threatens to disrupt this microbial balance, potentially promoting pathogens and impairing essential functions. Mytilus chilensis is vulnerable to hypoxia-reoxygenation cycles, yet the effects on its microbiota remain poorly understood. This study investigates the impact of hypoxia on the structure and functional potential of the microbial communities residing in the gills and digestive glands of M. chilensis. Employing full-length 16S rRNA gene sequencing, we explored hypoxia's effects on microbial diversity and functional capacity. Our results revealed significant alterations in the microbial composition, with a shift towards facultative anaerobes thriving in low oxygen environments. Notably, there was a decrease in dominant bacterial taxa such as Rhodobacterales, while opportunistic pathogens such as Vibrio and Aeromonas exhibited increased abundance. Functional analysis indicated a decline in critical microbial functions associated with nutrient metabolism and immune support, potentially jeopardizing the health and survival of the host. This study sheds light on the intricate interactions between host-associated microbiota and environmental stressors, underlining the importance of managing the microbiota in the face of climate change and aquaculture practices.
Collapse
Affiliation(s)
- Milton Montúfar-Romero
- Interdisciplinary Center for Aquaculture Research (INCAR), Universidad de Concepción, P.O. Box 160-C, Concepción 4030000, Chile; (M.M.-R.); (V.V.-M.); (M.F.M.-R.)
- Instituto Público de Investigación de Acuicultura y Pesca (IPIAP), Guayaquil 090314, Ecuador
| | - Diego Valenzuela-Miranda
- Interdisciplinary Center for Aquaculture Research (INCAR), Universidad de Concepción, P.O. Box 160-C, Concepción 4030000, Chile; (M.M.-R.); (V.V.-M.); (M.F.M.-R.)
- Centro de Biotecnología, Universidad de Concepción, P.O. Box 160-C, Concepción 4030000, Chile
| | - Valentina Valenzuela-Muñoz
- Interdisciplinary Center for Aquaculture Research (INCAR), Universidad de Concepción, P.O. Box 160-C, Concepción 4030000, Chile; (M.M.-R.); (V.V.-M.); (M.F.M.-R.)
- Center for Oceanographic Research COPAS COASTAL, Universidad de Concepción, Concepción 4070409, Chile
| | - María F. Morales-Rivera
- Interdisciplinary Center for Aquaculture Research (INCAR), Universidad de Concepción, P.O. Box 160-C, Concepción 4030000, Chile; (M.M.-R.); (V.V.-M.); (M.F.M.-R.)
| | - Cristian Gallardo-Escárate
- Interdisciplinary Center for Aquaculture Research (INCAR), Universidad de Concepción, P.O. Box 160-C, Concepción 4030000, Chile; (M.M.-R.); (V.V.-M.); (M.F.M.-R.)
- Center for Oceanographic Research COPAS COASTAL, Universidad de Concepción, Concepción 4070409, Chile
| |
Collapse
|
9
|
Miao ZY, Lin J, Chen WM. Natural sideromycins and siderophore-conjugated natural products as inspiration for novel antimicrobial agents. Eur J Med Chem 2025; 287:117333. [PMID: 39892091 DOI: 10.1016/j.ejmech.2025.117333] [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: 12/10/2024] [Revised: 01/25/2025] [Accepted: 01/25/2025] [Indexed: 02/03/2025]
Abstract
The widespread emergence of multidrug-resistant (MDR) Gram-negative pathogens has posed a major challenge to clinical anti-infective therapy, and new effective treatments are urgently needed. A promising "Trojan horse" strategy involves conjugating antibiotics to siderophore molecules; the resulting siderophore-antibiotic conjugates (SACs) deliver antibiotics directly into cells by hijacking the sophisticated iron transport systems of Gram-negative bacteria, bypassing the outer membrane permeability barrier to enhance uptake and antibacterial efficacy. The clinical release of the first siderophore-antibiotic conjugate, cefiderocol, has aroused tremendous interest in the field among researchers and pharmaceutical companies. To date, most of the reported SACs have focused on the conjugation of siderophores to traditional antibacterial drugs. However, these antibacterial agents designed on the basis of the traditional antibiotic skeleton theoretically bear the risk of cross-resistance caused by shared molecular scaffolds. In this case, exploring novel natural product antibacterial conjugate scaffolds to circumvent the risk of early cross-resistance represents a presumably more sustainable approach for the development of SACs. In this review, we systematically summarize the research progress on siderophore-natural product conjugates as novel antimicrobial agents reported since 2010. Additionally, we propose challenges to be overcome and prospects for future development in this field.
Collapse
Affiliation(s)
- Zhi-Ying Miao
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou, 511400, China
| | - Jing Lin
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou, 511400, China.
| | - Wei-Min Chen
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou, 511400, China.
| |
Collapse
|
10
|
Liu W, Chen Z, Li T, Wen X. Geographical distribution and risk of antibiotic resistance genes in sludge anaerobic digestion process across China. JOURNAL OF HAZARDOUS MATERIALS 2025; 487:137290. [PMID: 39837034 DOI: 10.1016/j.jhazmat.2025.137290] [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: 10/21/2024] [Revised: 01/09/2025] [Accepted: 01/18/2025] [Indexed: 01/23/2025]
Abstract
Anaerobic digestion (AD) is gaining increasing attention as the central reservoir of antibiotic resistance genes (ARGs), while the geographical distribution of ARGs in AD is neglected. Accordingly, a sampling scheme on full-scale AD plants across China was implemented, and the resistome therein was excavated. The abundance of ARGs in AD sludge ranged from 0.198 to 0.574 copies/cell. Some of the frequently reported and emergent ARGs were detected in our AD system. Both the abundance and composition of ARGs presented significant differences between the south and north regions of China, hinting the physical/economic factors may function in the formation of ARG profiles. The risk scores of AD samples were in middle of domestic and hospital wastewater. Risk scores were significantly higher in the north. Besides, the proportion of Rank I and Rank II ARGs was also higher in north, which explained the regional difference of ARG composition in a micro-perspective. This study provides a fundamental survey on the of ARG level and profile in AD process across China, reveals the biogeography of ARGs and inspires the control strategies of antibiotic resistance.
Collapse
Affiliation(s)
- Wei Liu
- School of Environment, Tsinghua University, Beijing 100084, PR China
| | - Zhan Chen
- School of Environment, Tsinghua University, Beijing 100084, PR China; State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 100012, PR China
| | - Tianle Li
- School of Environment, Tsinghua University, Beijing 100084, PR China
| | - Xianghua Wen
- School of Environment, Tsinghua University, Beijing 100084, PR China.
| |
Collapse
|
11
|
Chen X, Gao L, Kou Y, Wang X, Li X, He H, Wang M. Composition, Distribution and Mobility Potential of the Antibiotic Resistome in Sediments from the East China Sea Revealed by Metagenomic Analysis. Microorganisms 2025; 13:697. [PMID: 40142589 PMCID: PMC11944410 DOI: 10.3390/microorganisms13030697] [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: 03/01/2025] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 03/28/2025] Open
Abstract
Marine sediments are recognized as crucial reservoirs of antibiotic resistance genes (ARGs). However, the antibiotic resistome in sediments of the East China Sea, an area heavily impacted by human activities, has not been thoroughly studied. Here, we conducted a systematic investigation into the antibiotic resistome in these sediments using metagenomic analysis. Overall, we detected eighty ARG subtypes and nineteen ARG types. Beta-lactams were the dominant ARG type, and Gammaproteobacteria was the main ARG host in this study. Mobile genetic elements (MGEs) were not major drivers of ARG profiles. Although the ARG host communities significantly differed between the spring and autumn (p < 0.05), the antibiotic resistome remained stable across the two seasons. The assembly of ARGs and their hosts was governed by stochastic processes, and a high ratio of stochastic processes implied its crucial role in the assembly and stabilization of the antibiotic resistome. Co-occurrence network analysis revealed an important role of Deltaproteobacteria in the stabilization of ARG profiles across seasons. Environmental parameters (e.g., temperature and density) played certain roles in the stabilization of the antibiotic resistome between spring and autumn. Moreover, nine human pathogen bacteria (HPB) were detected in this study. We also found that the health risks caused by ARGs were relatively higher in the spring. Our results will provide a strong foundation for the development of targeted management strategies to mitigate the further dissemination and spread of ARGs in marine sediments.
Collapse
Affiliation(s)
- Xiaozhong Chen
- College of Marine Life Sciences, Key Laboratory of Evolution & Marine Biodiversity (Ministry of Education) and Institute of Evolution & Marine Biodiversity, Frontiers Science for Deep Ocean Multispheres and Earth System, Ocean University of China, Qingdao 266003, China; (X.C.); (L.G.); (Y.K.); (X.W.); (X.L.); (M.W.)
| | - Long Gao
- College of Marine Life Sciences, Key Laboratory of Evolution & Marine Biodiversity (Ministry of Education) and Institute of Evolution & Marine Biodiversity, Frontiers Science for Deep Ocean Multispheres and Earth System, Ocean University of China, Qingdao 266003, China; (X.C.); (L.G.); (Y.K.); (X.W.); (X.L.); (M.W.)
| | - Yanxue Kou
- College of Marine Life Sciences, Key Laboratory of Evolution & Marine Biodiversity (Ministry of Education) and Institute of Evolution & Marine Biodiversity, Frontiers Science for Deep Ocean Multispheres and Earth System, Ocean University of China, Qingdao 266003, China; (X.C.); (L.G.); (Y.K.); (X.W.); (X.L.); (M.W.)
| | - Xiaoxuan Wang
- College of Marine Life Sciences, Key Laboratory of Evolution & Marine Biodiversity (Ministry of Education) and Institute of Evolution & Marine Biodiversity, Frontiers Science for Deep Ocean Multispheres and Earth System, Ocean University of China, Qingdao 266003, China; (X.C.); (L.G.); (Y.K.); (X.W.); (X.L.); (M.W.)
| | - Xintong Li
- College of Marine Life Sciences, Key Laboratory of Evolution & Marine Biodiversity (Ministry of Education) and Institute of Evolution & Marine Biodiversity, Frontiers Science for Deep Ocean Multispheres and Earth System, Ocean University of China, Qingdao 266003, China; (X.C.); (L.G.); (Y.K.); (X.W.); (X.L.); (M.W.)
| | - Hui He
- College of Marine Life Sciences, Key Laboratory of Evolution & Marine Biodiversity (Ministry of Education) and Institute of Evolution & Marine Biodiversity, Frontiers Science for Deep Ocean Multispheres and Earth System, Ocean University of China, Qingdao 266003, China; (X.C.); (L.G.); (Y.K.); (X.W.); (X.L.); (M.W.)
| | - Min Wang
- College of Marine Life Sciences, Key Laboratory of Evolution & Marine Biodiversity (Ministry of Education) and Institute of Evolution & Marine Biodiversity, Frontiers Science for Deep Ocean Multispheres and Earth System, Ocean University of China, Qingdao 266003, China; (X.C.); (L.G.); (Y.K.); (X.W.); (X.L.); (M.W.)
- Haide College, Ocean University of China, Qingdao 266100, China
| |
Collapse
|
12
|
Shiralizadeh S, Farmani A, Shokoohizadeh L, Pourhajibagher M, Alikhani MY, Bahador A. The effect of photodynamic therapy using gentamicin and imipenem-derived carbon dots on Pseudomonas aeruginosa isolates. Lasers Med Sci 2025; 40:150. [PMID: 40105985 DOI: 10.1007/s10103-025-04402-1] [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] [Received: 03/13/2024] [Accepted: 02/26/2025] [Indexed: 03/22/2025]
Abstract
The increasing prevalence of antibiotic resistance poses a significant threat to the ability of healthcare systems to manage Pseudomonas aeruginosa infections, highlighting the need for innovative therapeutic strategies. The aim of this study was to evaluate the effects of photodynamic therapy (PDT) using carbon dots (CDs) derived from gentamicin and imipenem against P. aeruginosa isolates. A hydrothermal method was used to synthesise amine-functionalised CDs (CDsGEN-NH2, CDsIMP-NH2) from gentamicin and imipenem precursors, respectively. Conjugation of imipenem to CDsGEN-NH2 (CDsGEN-IMP) and gentamicin to CDsIMP-NH2 (CDsIMP-GEN) was achieved using N-(3-dimethylaminopropyl)-N'-ethylcarbodiimide hydrochloride and N-hydroxysuccinimide. The synthesised CDs were analysed by HRTEM, zeta potential, UV-Vis, FTIR and fluorescence. The efficacy of photodynamic therapy (PDT) against P. aeruginosa ATCC27853 and two clinical P. aeruginosa isolates was evaluated using a combination of sub-minimum inhibitory concentrations (sub-MIC) and sub-lethal doses of UVA light (sDL) irradiation. The nanoscale size, electrical charge and photophysical properties of the synthesised CDs were confirmed by HRTEM, zeta potential, UV-Vis, FTIR and fluorescence techniques. Treatment with CDsGEN-NH2, CDsIMP-NH2 and CDsGEN-IMP at concentrations of 3 mg/mL (sub-MIC) and sDL irradiation for 10 s (3.12 J/cm2) resulted in significantly reduced viability in P. aeruginosa ATCC27853 compared to the control (P ≤ 0.05), achieving viable cell reductions of 3.44, 3.11 and 2.65 log, respectively. Treatment with CDsIMP-GEN at a concentration of 125 µg/mL (sub-MIC) and sDL irradiation for 10 s (3.12 J/cm2) also significantly reduced viability in P. aeruginosa ATCC27853 compared to the control (P ≤ 0.05), achieving a viable cell reduction of 3.75 log. These results demonstrate that CDs derived from imipenem and gentamicin can serve as effective antimicrobial agents in combination with UVA light irradiation for antimicrobial photodynamic therapy.
Collapse
Affiliation(s)
- Somaye Shiralizadeh
- Department of Microbiology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Abbas Farmani
- Dental Research Center, Avicenna Institute of Clinical Sciences, Avicenna Health Research Institute, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Leili Shokoohizadeh
- Infectious Disease Research Center, Avicenna Institute of Clinical Sciences, Avicenna Health Research Institute, Hamadan University of Medical Sciences, Hamadan, Iran
| | | | - Mohammad Yousef Alikhani
- Department of Microbiology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
- Infectious Disease Research Center, Avicenna Institute of Clinical Sciences, Avicenna Health Research Institute, Hamadan University of Medical Sciences, Hamadan, Iran.
| | - Abbas Bahador
- Department of Microbiology, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
13
|
Isozaki Y, Makikawa T, Kimura K, Nishihara D, Fujino M, Tanaka Y, Hayashi C, Ishizaki Y, Igarashi M, Yokoyama T, Toshima K, Takahashi D. Creation of a macrolide antibiotic against non-tuberculous Mycobacterium using late-stage boron-mediated aglycon delivery. SCIENCE ADVANCES 2025; 11:eadt2352. [PMID: 40043128 PMCID: PMC11881915 DOI: 10.1126/sciadv.adt2352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 01/30/2025] [Indexed: 05/13/2025]
Abstract
Non-tuberculous mycobacteria (NTM) is gaining clinical recognition as a recently emerging pulmonary pathogen. Mycobacterium avium complex (MAC), the most common NTM, is the cause of pulmonary MAC disease. Currently, the macrolide azithromycin (AZM) is the standard first-line antibiotic for treatment of the disease. However, the rise of drug-resistant MAC necessitates the development of alternative therapeutics. Here, we present a late-stage boron-mediated aglycon delivery strategy for selective modification of AZM, generating a library of potential anti-MAC drugs designated KU01 to KU13. Screening of KU01 to KU13 revealed that KU13 exhibited enhanced antimicrobial activity against wild-type and macrolide-resistant MAC compared to AZM. Cryo-electron microscopy analysis indicated that the inserted tercyclic moiety of KU13 formed a robust anchor on the bacterial ribosome, creating a binding pocket with base flipping of U2847, potentially bypassing the standard mechanism of macrolide resistance. These results position KU13 as a promising lead for therapeutics against macrolide-resistant MAC.
Collapse
Affiliation(s)
- Yuka Isozaki
- Department of Applied Chemistry, Faculty of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama 223-8522, Japan
| | - Takumi Makikawa
- Department of Applied Chemistry, Faculty of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama 223-8522, Japan
| | - Kosuke Kimura
- Department of Applied Chemistry, Faculty of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama 223-8522, Japan
| | - Daiki Nishihara
- Graduate School of Life Sciences, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai, Miyagi 980-8577, Japan
| | - Maho Fujino
- Graduate School of Life Sciences, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai, Miyagi 980-8577, Japan
| | - Yoshikazu Tanaka
- Graduate School of Life Sciences, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai, Miyagi 980-8577, Japan
- The Advanced Center for Innovations in Next-Generation Medicine (INGEM), Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8573, Japan
| | - Chigusa Hayashi
- Institute of Microbial Chemistry (BIKAKEN), 3-14-23 Kamiosaki, Shinagawa-ku, Tokyo 141-0021, Japan
| | - Yoshimasa Ishizaki
- Institute of Microbial Chemistry (BIKAKEN), 3-14-23 Kamiosaki, Shinagawa-ku, Tokyo 141-0021, Japan
| | - Masayuki Igarashi
- Institute of Microbial Chemistry (BIKAKEN), 3-14-23 Kamiosaki, Shinagawa-ku, Tokyo 141-0021, Japan
| | - Takeshi Yokoyama
- Graduate School of Life Sciences, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai, Miyagi 980-8577, Japan
- The Advanced Center for Innovations in Next-Generation Medicine (INGEM), Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8573, Japan
| | - Kazunobu Toshima
- Department of Applied Chemistry, Faculty of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama 223-8522, Japan
| | - Daisuke Takahashi
- Department of Applied Chemistry, Faculty of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama 223-8522, Japan
| |
Collapse
|
14
|
Liao M, Gong H, Ge T, Shen K, Campana M, McBain AJ, Wu C, Hu X, Lu JR. Probing antimicrobial synergy by novel lipopeptides paired with antibiotics. J Colloid Interface Sci 2025; 681:82-94. [PMID: 39591858 DOI: 10.1016/j.jcis.2024.11.169] [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: 10/27/2024] [Accepted: 11/21/2024] [Indexed: 11/28/2024]
Abstract
Antimicrobial resistance (AMR) is fast becoming a major global challenge in both hospital and community settings as many current antibiotics and treatment processes are under the threat of being rendered less effective or ineffective. Synergistic combination of an antibiotic and an aiding agent with a different set of properties provides an important but largely unexploited option to 'repurpose' existing biomaterial's space while addressing issues of potency, spectrum, toxicity and resistance in early stages of antimicrobial drug discovery. This work explores how to combine tetracycline/minocycline (TC/MC) with a broad-spectrum antimicrobial lipopeptide that has been designed to improve the efficiency of membrane targeting and intramembrane accumulation, thereby enhancing antimicrobial efficacy. Experimental measurements of fractional inhibition concentration index (FICI) were undertaken from binary antibiotic-lipopeptide combinations. Most FICI values were found to be lower than 0.5 against both Gram-positive and Gram-negative bacterial strains studied including 3 AMR strains, revealing strong synergetic effects via favorable membrane-lytic interactions. The antimicrobial actions of this type of binary combinations are featured by the fast time-killing and high TC/MC uptake, benefited from effective membrane-lytic disruptions by the lipopeptide. This study thus provides an important mechanistic understanding of the combined antibiotic-lipopeptide approach to improve the therapeutic potential of conventional antibiotics by illustrating how amphiphilic lipopeptide-antibiotic combinations interact with biological membranes, providing a promising alternative to combat AMR through rational design of lipopeptide as an aiding agent.
Collapse
Affiliation(s)
- Mingrui Liao
- Biological Physics Laboratory, Department of Physics and Astronomy, School of Natural Science, The University of Manchester, Oxford Road, Manchester M13 9PL UK
| | - Haoning Gong
- Biological Physics Laboratory, Department of Physics and Astronomy, School of Natural Science, The University of Manchester, Oxford Road, Manchester M13 9PL UK
| | - Tianhao Ge
- Biological Physics Laboratory, Department of Physics and Astronomy, School of Natural Science, The University of Manchester, Oxford Road, Manchester M13 9PL UK
| | - Kangcheng Shen
- Biological Physics Laboratory, Department of Physics and Astronomy, School of Natural Science, The University of Manchester, Oxford Road, Manchester M13 9PL UK
| | - Mario Campana
- ISIS Pulsed Neutron & Muon Source, STFC Rutherford Appleton Laboratory, Didcot OX11 0QX, UK
| | - Andrew J McBain
- Division of Pharmacy and Optometry, Faculty of Biology, Medicine and Health, The University of Manchester, Oxford Road, Manchester M13 9PL, UK
| | - Chunxian Wu
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Xuzhi Hu
- State Key Laboratory of Solid Lubrication, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou 730000, PR China.
| | - Jian R Lu
- Biological Physics Laboratory, Department of Physics and Astronomy, School of Natural Science, The University of Manchester, Oxford Road, Manchester M13 9PL UK.
| |
Collapse
|
15
|
He X, Zou C, Zhang L, Wu P, Yao Y, Dong K, Ren Y, Hu WW, Li Y, Luo H, Ying B, Luo F, Sun X. Advances in Electrochemical Nitrite Reduction toward Nitric Oxide Synthesis for Biomedical Applications. Adv Healthc Mater 2025; 14:e2403468. [PMID: 39865954 DOI: 10.1002/adhm.202403468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 12/03/2024] [Indexed: 01/28/2025]
Abstract
Nitric oxide (NO) is an essential molecule in biomedicine, recognized for its antibacterial properties, neuronal modulation, and use in inhalation therapies. The effectiveness of NO-based treatments relies on precise control of NO concentrations tailored to specific therapeutic needs. Electrochemical generation of NO (E-NOgen) via nitrite (NO2 -) reduction offers a scalable and efficient route for controlled NO production, while also addressing environmental concerns by reducing NO2 - pollution and maintaining nitrogen cycle balance. Recent developments in catalysts and E-NOgen devices have propelled NO2 - conversion, enabling on-demand NO production. This review provides an overview of NO2 - reduction pathways, with a focus on cutting-edge Fe/Cu-based E-NOgen catalysts, and explores the development of E-NOgen devices for biomedical use. Challenges and future directions for advancing E-NOgen technologies are also discussed.
Collapse
Affiliation(s)
- Xun He
- Center for High Altitude Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610054, China
| | - Chang Zou
- Center for High Altitude Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Limei Zhang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Peilin Wu
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yongchao Yao
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Kai Dong
- College of Chemistry, Chemical Engineering and Materials Science, Shandong Normal University, Jinan, Sichuan, 250014, China
| | - Yuchun Ren
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610054, China
| | - Wenchuang Walter Hu
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yi Li
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Han Luo
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Binwu Ying
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Fengming Luo
- Center for High Altitude Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xuping Sun
- Center for High Altitude Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- College of Chemistry, Chemical Engineering and Materials Science, Shandong Normal University, Jinan, Sichuan, 250014, China
| |
Collapse
|
16
|
Kanwal S, Aziz UBA, Quaas E, Achazi K, Klinger D. Sulfonium-based polymethacrylamides for antimicrobial use: influence of the structure and composition. Biomater Sci 2025; 13:993-1009. [PMID: 39801426 DOI: 10.1039/d4bm01247j] [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: 02/12/2025]
Abstract
We are facing a shortage of new antibiotics to fight against increasingly resistant bacteria. As an alternative to conventional small molecule antibiotics, antimicrobial polymers (AMPs) have great potential. These polymers contain cationic and hydrophobic groups and disrupt bacterial cell membranes through a combination of electrostatic and hydrophobic interactions. While most examples focus on ammonium-based cations, sulfonium groups are recently emerging to broaden the scope of polymeric therapeutics. Here, main-chain sulfonium polymers exhibit good antimicrobial activity. In contrast, the potential of side-chain sulfonium polymers remains less explored with structure-activity relationships still being limited. To address this limitation, we thoroughly investigated key factors influencing antimicrobial activity in side-chain sulfonium-based AMPs. For this, we combined sulfonium cations with different hydrophobic (aliphatic/aromatic) and hydrophilic polyethylene glycol (PEG) groups to create a library of polymers with comparable chain lengths. For all compositions, we additionally examined the position of cationic and hydrophobic groups on the polymer backbone, i.e., we systematically compared same center and different center structures. Bactericidal tests against Gram-positive and Gram-negative bacteria suggest that same center polymers are more active than different center polymers of similar clog P. Ultimately, sulfonium-based AMPs show superior bactericidal activity and selectivity when compared to their quaternary ammonium cationic analogues.
Collapse
Affiliation(s)
- Sidra Kanwal
- Institute of Pharmacy, Freie Universität Berlin, 14195 Berlin, Germany.
| | | | - Elisa Quaas
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Katharina Achazi
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Daniel Klinger
- Institute of Pharmacy, Freie Universität Berlin, 14195 Berlin, Germany.
| |
Collapse
|
17
|
Tijani NA, Hokello J, Eilu E, Akinola SA, Afolabi AO, Makeri D, Lukwago TW, Mutuku IM, Mwesigwa A, Baguma A, Adebayo IA. Metallic nanoparticles: a promising novel therapeutic tool against antimicrobial resistance and spread of superbugs. Biometals 2025; 38:55-88. [PMID: 39446237 DOI: 10.1007/s10534-024-00647-5] [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: 08/03/2024] [Accepted: 10/12/2024] [Indexed: 10/25/2024]
Abstract
In recent years, antimicrobial resistance (AMR) has become an alarming threat to global health as notable increase in morbidity and mortality has been ascribed to the emergence of superbugs. The increase in microbial resistance because of harboured or inherited resistomes has been complicated by the lack of new and effective antimicrobial agents, as well as misuse and failure of existing ones. These problems have generated severe and growing public health concern, due to high burden of bacterial infections resulting from scarce financial resources and poor functioning health systems, among others. It is therefore, highly pressing to search for novel and more efficacious alternatives for combating the action of these super bacteria and their infection. The application of metallic nanoparticles (MNPs) with their distinctive physical and chemical attributes appears as promising tools in fighting off these deadly superbugs. The simple, inexpensive and eco-friendly model for enhanced biologically inspired MNPs with exceptional antimicrobial effect and diverse mechanisms of action againsts multiple cell components seems to offer the most promising option and said to have enticed many researchers who now show tremendous interest. This synopsis offers critical discussion on application of MNPs as the foremost intervening strategy to curb the menace posed by the spread of superbugs. As such, this review explores how antimicrobial properties of the metallic nanoparticles which demonstrated considerable efficacy against several multi-drugs resistant bacteria, could be adopted as promising approach in subduing the threat of AMR and harvoc resulting from the spread of superbugs.
Collapse
Affiliation(s)
- Naheem Adekilekun Tijani
- Department of Microbiology and Immunology, Kampala International University, Western Campus, Bushenyi, Uganda
| | - Joseph Hokello
- Department of Biology, Faculty of Science and Education, Busitema University, Tororo, Uganda
| | - Emmanuel Eilu
- Department of Microbiology and Immunology, Kampala International University, Western Campus, Bushenyi, Uganda
| | - Saheed Adekunle Akinola
- Food Security and Safety Focus Area, Faculty of Natural and Agricultural Sciences, North-West University, Mmabatho, South Africa
| | - Abdullateef Opeyemi Afolabi
- Department of Microbiology and Immunology, Kampala International University, Western Campus, Bushenyi, Uganda
| | - Danladi Makeri
- Department of Microbiology and Immunology, Kampala International University, Western Campus, Bushenyi, Uganda
| | - Tonny Wotoyitide Lukwago
- Department of Pharmacology and Toxicology, Kampala International University, Western Campus, Bushenyi, Uganda
| | - Irene M Mutuku
- Department of Microbiology, School of Medicine, Kabale University, Kabale, Uganda
| | - Alex Mwesigwa
- Department of Microbiology, School of Medicine, Kabale University, Kabale, Uganda
| | - Andrew Baguma
- Department of Microbiology, School of Medicine, Kabale University, Kabale, Uganda
| | | |
Collapse
|
18
|
Huang X, Cai J, Xiao J, Zhou X, Liu Y, Li Y, Yang B. Antimicrobial Isochroman-Derived Atropo-Diastereomeric Dimers from Penicillium Steckii SCISO 41228. Chem Biodivers 2025; 22:e202402152. [PMID: 39385043 DOI: 10.1002/cbdv.202402152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 10/11/2024]
Abstract
A pair of unidentified atropo-diastereomeric dimers, penicisteckins G (1) and H (2), and twelve known compounds (3-16) were isolated from the marine coral-derived fungus Penicillium steckii SCISO 41228. Their structures including the absolute configuration were determined by HR-ESI-MS, ECD, 1D-, and 2D-NMR spectra. Compounds 1 and 2 exhibited moderate antibacterial activity against most tested pathogenic strains, especially for methicillin-resistant Staphyloccocus aureus (MASA) and Micrococcus luteus, with MIC values of 4.0 μg mL-1. In addition, compounds 2 and 3 exhibit potent radical scavenging activity against 2,2-diphenyl-1-picrylhydrazylhydrate (DPPH), with IC50 values of 10.76 and 8.66 μg mL-1, respectively.
Collapse
Affiliation(s)
- Xiaowen Huang
- Guilin Medical University, Guilin, Guangxi, China
- South China Sea Institute of Oceanography, Chinese Academy of Sciences, Key Laboratory of Tropical Marine Biological Resources and Ecology, Chinese Academy of Sciences, Guangzhou, Guangdong, China
| | - Jian Cai
- South China Sea Institute of Oceanography, Chinese Academy of Sciences, Key Laboratory of Tropical Marine Biological Resources and Ecology, Chinese Academy of Sciences, Guangzhou, Guangdong, China
| | - Jiao Xiao
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, P. R. China
| | - Xuefeng Zhou
- South China Sea Institute of Oceanography, Chinese Academy of Sciences, Key Laboratory of Tropical Marine Biological Resources and Ecology, Chinese Academy of Sciences, Guangzhou, Guangdong, China
| | - Yonghong Liu
- South China Sea Institute of Oceanography, Chinese Academy of Sciences, Key Laboratory of Tropical Marine Biological Resources and Ecology, Chinese Academy of Sciences, Guangzhou, Guangdong, China
| | - Yunqiu Li
- Guilin Medical University, Guilin, Guangxi, China
| | - Bin Yang
- South China Sea Institute of Oceanography, Chinese Academy of Sciences, Key Laboratory of Tropical Marine Biological Resources and Ecology, Chinese Academy of Sciences, Guangzhou, Guangdong, China
| |
Collapse
|
19
|
Hsiao WWW, Selvi SV, Alagumalai K. Fabrication of MnSnO 2 intercalated TA-rGO modified sensor for selective electrochemical detection of chloramphenicol in real samples. Food Chem 2025; 464:141474. [PMID: 39427617 DOI: 10.1016/j.foodchem.2024.141474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 10/22/2024]
Abstract
Chloramphenicol (CAP), a potent antibiotic capable of inhibiting protein synthesis, presents significant challenges related to long-term dosing and its persistent leaching into the environment, raising concerns about environmental contamination and resistance development. To address this issue, we developed a reliable, low-cost, and biocompatible nanocomposite material comprising tannic acid (TA)-reduced graphene oxide (rGO) intercalated into manganese-doped tin oxide nanoparticles (MnSnO₂ NPs). The structural formation and catalytic activity of the MnSnO₂ NPs/TA-rGO nanocomposite were characterized using field emission-scanning electron microscopy (FE-SEM), high-resolution transmission electron microscopy (HR-TEM), X-ray diffraction (XRD), Fourier-transform infrared spectroscopy (FT-IR), Raman spectroscopy, X-ray photoelectron spectroscopy (XPS), and electrochemical techniques. This material exhibits robust interfacial interactions and synergistic effects, resulting in an admirable electrocatalytic reduction response for CAP sensing. The presence of co-interference molecules improved the selectivity performance of the MnSnO2 NPs/TA-rGO-modified glassy carbon electrode. The fabricated exhibited a two linear determination range (0.011-103.43 μmol L-1 and 103.43-1924.16 μmol L-1), with a detection limit (LOD) is 6.7 nmol L-1 and limit of quantification (LOQ) is 12.3 nmol L-1. Furthermore, this sensor demonstrated good sensitivity, admirable reproducibility, repeatability, and storage stability. Finally, the practicability of the fabricated MnSnO2 NPs/TA-rGO glassy carbon electrode sensor was evaluated by analyzing the CAP content in milk, honey, eye drops, biofluids (human serum and urine), and river water, and satisfactory recovery rates of 95.4 %-100.3 % were noted.
Collapse
Affiliation(s)
- Wesley Wei-Wen Hsiao
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei 106335, Taiwan.
| | - Subash Vetri Selvi
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei 106335, Taiwan; Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei 106319, Taiwan
| | | |
Collapse
|
20
|
Chen Y, Xu M, Pan J, Liao Y, Na J, Li P, Sun Y, Yu S, Zhao Y, Hu H. Moxifloxacin-Loaded Polymeric Nanoparticles for Overcoming Multidrug Resistance in Chronic Pulmonary Infections Caused by Pseudomonas aeruginosa. ACS APPLIED MATERIALS & INTERFACES 2025; 17:5695-5709. [PMID: 39804842 DOI: 10.1021/acsami.4c14991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Pseudomonas aeruginosa (P. aeruginosa) infections are increasingly challenging due to their propensity to form biofilms and low outer membrane permeability, especially in chronically infected patients with thick mucus. P. aeruginosa exhibits multiple drug resistance mechanisms, making it one of the most significant global public health threats. In this study, we found that moxifloxacin (MXC) and antibacterial peptides (ε-poly-l-lysine, ε-PLL) exhibited a synergistic effect against multidrug-resistant P. aeruginosa (MDR-P. aeruginosa). MXC was combined with ε-PLL to prepare lipase-responsive nanoparticles (MCIP/(PEG-PCL)/PLL NPs) with a weakly negative charge. The weakly negatively charged MCIP/(PEG-PCL)/PLL NPs demonstrated remarkable mucus and biofilm penetration capabilities, thereby overcoming one of the adaptive drug resistance mechanisms. MCIP/(PEG-PCL)/PLL NPs improved the outer and inner membrane permeability and inhibited the expression of the efflux pump MexAB-OprM gene in MDR-P. aeruginosa, thereby overcoming mechanisms of both intrinsic and acquired drug resistance. Meanwhile, the nanoparticles demonstrated an ability to reduce repeated infections with MDR-P. aeruginosa. Additionally, the bacterial burden in the lungs of mice treated with MCIP/(PEG-PCL)/PLL NPs was significantly lower than that in the MXC group, resulting in a 99% clearance rate. Notably, MCIP/(PEG-PCL)/PLL NPs showed no toxicity toward BEAS-2B cells or RAW 267.4 cells, nor did they adversely affect pulmonary function or major organs. This study demonstrated the potential of the nanodrug delivery system composed of the antibiotic moxifloxacin and the antibacterial peptide ε-PLL in addressing the clinical challenges of treating chronic pulmonary infections caused by MDR-P. aeruginosa.
Collapse
Affiliation(s)
- Yujun Chen
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Mao Xu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Jieyi Pan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Yuan Liao
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Jintong Na
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Pengyu Li
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Yingying Sun
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Shihui Yu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Yongxiang Zhao
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Haiyan Hu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| |
Collapse
|
21
|
Furlan B, Sobrinos-Sanguino M, Sammartino M, Monterroso B, Zorrilla S, Lanzini A, Suigo L, Valoti E, Massidda O, Straniero V. Targeting Bacterial Cell Division with Benzodioxane-Benzamide FtsZ Inhibitors as a Novel Strategy to Fight Gram-Positive Ovococcal Pathogens. Int J Mol Sci 2025; 26:714. [PMID: 39859428 PMCID: PMC11765573 DOI: 10.3390/ijms26020714] [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: 12/12/2024] [Revised: 01/10/2025] [Accepted: 01/11/2025] [Indexed: 01/27/2025] Open
Abstract
The widespread emergence of antimicrobial resistance (AMR) is a serious threat to global public health and among Gram-positive cocci, Streptococcus pneumoniae constitutes a priority in the list of AMR-threatening pathogens. To counteract this fundamental problem, the bacterial cell division cycle and the crucial proteins involved in this process emerged as novel attractive targets. FtsZ is an essential cell division protein, and FtsZ inhibitors, especially the benzamide derivatives, have been exploited in the last decade. In this work, we identified, for the first time, some benzodioxane-benzamide inhibitors capable of targeting FtsZ in Streptococcus pneumoniae, in addition to their previously demonstrated activity against other bacteria. These promising benzamides, with minimal inhibitory concentrations (MICs) ranging from 25 to 80 µg/mL, demonstrated bactericidal activity against S. pneumoniae. This was evidenced by their ability to dramatically affect growth and viability, further supported by the morphological changes observed through microscopy. Moreover, the compounds were characterized in vitro, combining turbidity measurements and confocal imaging, and significant alteration of a GTP-induced FtsZ assembly was found, in line with our previous data from other microorganisms.
Collapse
Affiliation(s)
- Berenice Furlan
- Dipartimento di Biologia Cellulare e Integrata, Università degli Studi di Trento, Via Sommarive, 9, 38123 Trento, Italy;
| | - Marta Sobrinos-Sanguino
- Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas, Ramiro de Maeztu 9, 28040 Madrid, Spain; (M.S.-S.); (M.S.); (S.Z.)
| | - Marcella Sammartino
- Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas, Ramiro de Maeztu 9, 28040 Madrid, Spain; (M.S.-S.); (M.S.); (S.Z.)
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Via Luigi Mangiagalli, 25, 20133 Milan, Italy; (A.L.); (L.S.); (E.V.)
| | - Begoña Monterroso
- Instituto de Química Física Blas Cabrera, Consejo Superior de Investigaciones Científicas, Serrano 119, 28006 Madrid, Spain;
| | - Silvia Zorrilla
- Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas, Ramiro de Maeztu 9, 28040 Madrid, Spain; (M.S.-S.); (M.S.); (S.Z.)
| | - Alessia Lanzini
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Via Luigi Mangiagalli, 25, 20133 Milan, Italy; (A.L.); (L.S.); (E.V.)
| | - Lorenzo Suigo
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Via Luigi Mangiagalli, 25, 20133 Milan, Italy; (A.L.); (L.S.); (E.V.)
- Department of Microbiology and Molecular Genetics, McGovern Medical School, University of Texas, Houston, TX 77030, USA
| | - Ermanno Valoti
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Via Luigi Mangiagalli, 25, 20133 Milan, Italy; (A.L.); (L.S.); (E.V.)
| | - Orietta Massidda
- Dipartimento di Biologia Cellulare e Integrata, Università degli Studi di Trento, Via Sommarive, 9, 38123 Trento, Italy;
- Centro Interdipartmentale di Scienze Mediche, Via Santa M. Maddalena, 1, 38122 Trento, Italy
| | - Valentina Straniero
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Via Luigi Mangiagalli, 25, 20133 Milan, Italy; (A.L.); (L.S.); (E.V.)
| |
Collapse
|
22
|
Koller TO, Berger MJ, Morici M, Paternoga H, Bulatov T, Di Stasi A, Dang T, Mainz A, Raulf K, Crowe-McAuliffe C, Scocchi M, Mardirossian M, Beckert B, Vázquez-Laslop N, Mankin AS, Süssmuth RD, Wilson DN. Paenilamicins are context-specific translocation inhibitors of protein synthesis. Nat Chem Biol 2024; 20:1691-1700. [PMID: 39420228 PMCID: PMC11581978 DOI: 10.1038/s41589-024-01752-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 09/13/2024] [Indexed: 10/19/2024]
Abstract
The paenilamicins are a group of hybrid nonribosomal peptide-polyketide compounds produced by the honey bee pathogen Paenibacillus larvae that display activity against Gram-positive pathogens, such as Staphylococcus aureus. While paenilamicins have been shown to inhibit protein synthesis, their mechanism of action has remained unclear. Here we determine structures of paenilamicin PamB2-stalled ribosomes, revealing a unique binding site on the small 30S subunit located between the A- and P-site transfer RNAs (tRNAs). In addition to providing a precise description of interactions of PamB2 with the ribosome, the structures also rationalize the resistance mechanisms used by P. larvae. We further demonstrate that PamB2 interferes with the translocation of messenger RNA and tRNAs through the ribosome during translation elongation, and that this inhibitory activity is influenced by the presence of modifications at position 37 of the A-site tRNA. Collectively, our study defines the paenilamicins as a class of context-specific translocation inhibitors.
Collapse
Affiliation(s)
- Timm O Koller
- Institute for Biochemistry and Molecular Biology, University of Hamburg, Hamburg, Germany
| | - Max J Berger
- Institute for Biochemistry and Molecular Biology, University of Hamburg, Hamburg, Germany
| | - Martino Morici
- Institute for Biochemistry and Molecular Biology, University of Hamburg, Hamburg, Germany
| | - Helge Paternoga
- Institute for Biochemistry and Molecular Biology, University of Hamburg, Hamburg, Germany
| | - Timur Bulatov
- Institut für Chemie, Technische Universität Berlin, Berlin, Germany
| | - Adriana Di Stasi
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Tam Dang
- Institut für Chemie, Technische Universität Berlin, Berlin, Germany
| | - Andi Mainz
- Institut für Chemie, Technische Universität Berlin, Berlin, Germany
| | - Karoline Raulf
- Institute for Biochemistry and Molecular Biology, University of Hamburg, Hamburg, Germany
| | | | - Marco Scocchi
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | | | - Bertrand Beckert
- Dubochet Center for Imaging (DCI) at EPFL, EPFL SB IPHYS DCI, Lausanne, Switzerland
| | - Nora Vázquez-Laslop
- Center for Biomolecular Sciences, University of Illinois at Chicago, Chicago, IL, USA
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Alexander S Mankin
- Center for Biomolecular Sciences, University of Illinois at Chicago, Chicago, IL, USA
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | | | - Daniel N Wilson
- Institute for Biochemistry and Molecular Biology, University of Hamburg, Hamburg, Germany.
| |
Collapse
|
23
|
Singh A, Tanwar M, Singh TP, Sharma S, Sharma P. An escape from ESKAPE pathogens: A comprehensive review on current and emerging therapeutics against antibiotic resistance. Int J Biol Macromol 2024; 279:135253. [PMID: 39244118 DOI: 10.1016/j.ijbiomac.2024.135253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/09/2024]
Abstract
The rise of antimicrobial resistance has positioned ESKAPE pathogens as a serious global health threat, primarily due to the limitations and frequent failures of current treatment options. This growing risk has spurred the scientific community to seek innovative antibiotic therapies and improved oversight strategies. This review aims to provide a comprehensive overview of the origins and resistance mechanisms of ESKAPE pathogens, while also exploring next-generation treatment strategies for these infections. In addition, it will address both traditional and novel approaches to combating antibiotic resistance, offering insights into potential new therapeutic avenues. Emerging research underscores the urgency of developing new antimicrobial agents and strategies to overcome resistance, highlighting the need for novel drug classes and combination therapies. Advances in genomic technologies and a deeper understanding of microbial pathogenesis are crucial in identifying effective treatments. Integrating precision medicine and personalized approaches could enhance therapeutic efficacy. The review also emphasizes the importance of global collaboration in surveillance and stewardship, as well as policy reforms, enhanced diagnostic tools, and public awareness initiatives, to address resistance on a worldwide scale.
Collapse
Affiliation(s)
- Anamika Singh
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Mansi Tanwar
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India
| | - T P Singh
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Sujata Sharma
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India.
| | - Pradeep Sharma
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India.
| |
Collapse
|
24
|
Dou L, Wang X, Bai Y, Li Q, Luo L, Yu W, Wang Z, Wen K, Shen J. Mussel-like polydopamine-assisted aggregation-induced emission nanodot for enhanced broad-spectrum antimicrobial activity: In vitro and in vivo validation. Int J Biol Macromol 2024; 282:136762. [PMID: 39486741 DOI: 10.1016/j.ijbiomac.2024.136762] [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: 06/11/2024] [Revised: 09/22/2024] [Accepted: 10/19/2024] [Indexed: 11/04/2024]
Abstract
Emerging luminogens with aggregation-induced emission properties, namely AIEgens, demonstrated excellent anti-bacteria activity potential. However, their application still limited by the low antibacterial activity caused by the poor binding with bacteria. Polydopamine (PDA), an important biological macromolecule, possesses superior adhesion ability toward various material surface, including bacteria. In this study, the novel mussel-like PDA-assisted AIE Nanodot was proposed, achieving with robust bacterial binding ability and enhanced broad-spectrum antibacterial activity. Binding ability inherited from the PDA enables the constructed PDA-assisted AIE Nanodot to adhere efficiently to the bacterial membrane surface. Meanwhile, the AIE properties endowed them with monitoring capability, allowing for tracking their interaction with bacteria through facile fluorescence imaging in real time. More importantly, excellent killing of both Gram-positive and Gram-negative bacteria were successfully achieved in vitro antibacterial tests with excellent biocompatibility. Furthermore, in the treatment of Methicillin-resistant S. aureus (MRSA)-infected mouse-wound model, the mice exhibited accelerated wound healing with low bacterial load. This novel integrated strategy introduced a simple but effectively design to enhance the binding and antibacterial ability of AIEgens and would diversify the existing pool of antibacterial agents.
Collapse
Affiliation(s)
- Leina Dou
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, Beijing Laboratory for Food Quality and Safety, Beijing 100193, China; College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shanxi, China
| | - Xiaonan Wang
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, Beijing Laboratory for Food Quality and Safety, Beijing 100193, China
| | - Yuchen Bai
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, Beijing Laboratory for Food Quality and Safety, Beijing 100193, China
| | - Qing Li
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, Beijing Laboratory for Food Quality and Safety, Beijing 100193, China
| | - Liang Luo
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, Beijing Laboratory for Food Quality and Safety, Beijing 100193, China
| | - Wenbo Yu
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, Beijing Laboratory for Food Quality and Safety, Beijing 100193, China
| | - Zhanhui Wang
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, Beijing Laboratory for Food Quality and Safety, Beijing 100193, China
| | - Kai Wen
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, Beijing Laboratory for Food Quality and Safety, Beijing 100193, China.
| | - Jianzhong Shen
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, Beijing Laboratory for Food Quality and Safety, Beijing 100193, China.
| |
Collapse
|
25
|
Panigrahi SD, Klebba KC, Rodriguez EN, Mayhan CM, Kotagiri N, Kumari H. Enhancing antibacterial efficacy through macrocyclic host complexation of fluoroquinolone antibiotics for overcoming resistance. Sci Rep 2024; 14:24637. [PMID: 39428392 PMCID: PMC11491488 DOI: 10.1038/s41598-024-73568-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 09/18/2024] [Indexed: 10/22/2024] Open
Abstract
The use of supramolecular assemblies in pharmaceuticals has garnered significant interest. Recent studies have shown that the activities of antibacterial agents can be enhanced through complexation with cyclic oligomers and metal ions. Notably, these complexes sometimes possess greater therapeutic properties than the parent drugs. To develop microbiologically potent supramolecular drugs, the complexation of macrocyclic hosts with fluoroquinolone (FQ) antibiotics was investigated. FQs are a successful family of antibiotics that target the bacterial enzymes DNA gyrase and DNA topoisomerase IV, leading to bacterial cell death through the inhibition of DNA synthesis. However, antibiotic resistance resulting from the repeated use of FQs over time has limited their effectiveness against resistant pathogens. To overcome this issue, the encapsulation of FQs in polyphenolic macrocycles was investigated. This study highlights resorcinarene, a polyphenolic host with antibacterial properties, and its ability to chemically interact with FQs. The inclusion complexation process was analyzed using NMR and FTIR techniques. The binding constants determined by 1H-NMR titration revealed that levofloxacin forms more stable complexes with resorcinarene than with β-cyclodextrin, which aligned with MD simulations. Assessment of the geometric characteristics of the inclusion complexes using 2D NMR analysis confirmed that different moieties of various FQs can fit into a single host cavity and improve activity against gram-negative bacteria. Overall, these findings suggest that encapsulation in polyphenolic macrocycles is a promising strategy for utilizing FQs against antibiotic-resistant bacteria.
Collapse
Affiliation(s)
- Suchitra D Panigrahi
- James L. Winkle College of Pharmacy University of Cincinnati, 231 Albert Sabin Way, Medical Science Building 3109C, Cincinnati, OH, 45267-0514, USA
| | - Karoline C Klebba
- Helias Catholic High School, 1305 Swifts Hwy, Jefferson City, MO, 65109, USA
| | - Emily N Rodriguez
- Helias Catholic High School, 1305 Swifts Hwy, Jefferson City, MO, 65109, USA
| | - Collin M Mayhan
- James L. Winkle College of Pharmacy University of Cincinnati, 231 Albert Sabin Way, Medical Science Building 3109C, Cincinnati, OH, 45267-0514, USA
- Helias Catholic High School, 1305 Swifts Hwy, Jefferson City, MO, 65109, USA
| | - Nalinikanth Kotagiri
- James L. Winkle College of Pharmacy University of Cincinnati, 231 Albert Sabin Way, Medical Science Building 3109C, Cincinnati, OH, 45267-0514, USA
| | - Harshita Kumari
- James L. Winkle College of Pharmacy University of Cincinnati, 231 Albert Sabin Way, Medical Science Building 3109C, Cincinnati, OH, 45267-0514, USA.
| |
Collapse
|
26
|
Tian P, Li QQ, Guo MJ, Zhu YZ, Zhu RQ, Guo YQ, Yang Y, Liu YY, Yu L, Li YS, Li JB. Zidovudine in synergistic combination with nitrofurantoin or omadacycline: in vitro and in murine urinary tract or lung infection evaluation against multidrug-resistant Klebsiella pneumoniae. Antimicrob Agents Chemother 2024; 68:e0034424. [PMID: 39194261 PMCID: PMC11459972 DOI: 10.1128/aac.00344-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 08/05/2024] [Indexed: 08/29/2024] Open
Abstract
Limited treatment options and multidrug-resistant (MDR) Klebsiella pneumoniae present a significant therapeutic challenge, underscoring the need for novel approaches. Drug repurposing is a promising tool for augmenting the activity of many antibiotics. This study aimed to identify novel synergistic drug combinations against K. pneumoniae based on drug repurposing. We used the clinically isolated GN 172867 MDR strain of K. pneumoniae to determine the reversal resistance activity of zidovudine (AZT). The combined effects of AZT and various antibiotics, including nitrofurantoin (NIT) and omadacycline (OMC), were examined using the checkerboard method, growth curves, and crystal violet assays to assess biofilms. An in vitro combination activity testing was carried out in 12 isolates of K. pneumoniae. In vivo murine urinary tract and lung infection models were used to evaluate the therapeutic effects of AZT + NIT and AZT + OMC, respectively. The fractional inhibitory concentration index and growth curve demonstrated that AZT synergized with NIT or OMC against K. pneumoniae strains. In addition, AZT + NIT inhibited biofilm formation and cleared mature biofilms. In vivo, compared with untreated GN 172867-infected mice, AZT + NIT and AZT + OMC treatment decreased colony counts in multiple tissues (P < 0.05) and pathological scores in the bladder and kidneys (P < 0.05) and increased the survival rate by 60% (P < 0.05). This study evaluated the combination of AZT and antibiotics to treat drug-resistant K. pneumoniae infections and found novel drug combinations for the treatment of acute urinary tract infections. These findings suggest that AZT may exert significant anti-resistance activity.
Collapse
Affiliation(s)
- Ping Tian
- Department of Infectious Diseases & Anhui Center for Surveillance of Bacterial Resistance, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Infectious Diseases and Institute of Bacterial Resistance, Anhui Medical University, Hefei, China
| | - Qing-Qing Li
- Department of Infectious Diseases & Anhui Center for Surveillance of Bacterial Resistance, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Infectious Diseases and Institute of Bacterial Resistance, Anhui Medical University, Hefei, China
| | - Ming-Juan Guo
- Department of Hepatology, The First Affiliated Hospital of Jilin University, Changchun, China
| | - Yun-Zhu Zhu
- Department of Infectious Diseases & Anhui Center for Surveillance of Bacterial Resistance, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Infectious Diseases and Institute of Bacterial Resistance, Anhui Medical University, Hefei, China
| | - Rong-Qing Zhu
- Department of Infectious Diseases & Anhui Center for Surveillance of Bacterial Resistance, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Infectious Diseases and Institute of Bacterial Resistance, Anhui Medical University, Hefei, China
| | - Ya-Qin Guo
- Department of Infectious Diseases & Anhui Center for Surveillance of Bacterial Resistance, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Infectious Diseases and Institute of Bacterial Resistance, Anhui Medical University, Hefei, China
| | - Yi Yang
- Department of Infectious Diseases & Anhui Center for Surveillance of Bacterial Resistance, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Infectious Diseases and Institute of Bacterial Resistance, Anhui Medical University, Hefei, China
| | - Yan-Yan Liu
- Department of Infectious Diseases & Anhui Center for Surveillance of Bacterial Resistance, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Infectious Diseases and Institute of Bacterial Resistance, Anhui Medical University, Hefei, China
| | - Liang Yu
- Department of Infectious Diseases & Anhui Center for Surveillance of Bacterial Resistance, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Infectious Diseases and Institute of Bacterial Resistance, Anhui Medical University, Hefei, China
| | - Ya-Sheng Li
- Department of Infectious Diseases & Anhui Center for Surveillance of Bacterial Resistance, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Infectious Diseases and Institute of Bacterial Resistance, Anhui Medical University, Hefei, China
| | - Jia-Bin Li
- Department of Infectious Diseases & Anhui Center for Surveillance of Bacterial Resistance, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Infectious Diseases and Institute of Bacterial Resistance, Anhui Medical University, Hefei, China
| |
Collapse
|
27
|
Ma C, Wei R, Yu R, Lei L, Pan X, Hu HY, Feng B, Liu Z. Design, synthesis of griseofamine A derivatives and development of potent antibacterial agents against MRSA. Eur J Med Chem 2024; 276:116703. [PMID: 39059183 DOI: 10.1016/j.ejmech.2024.116703] [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: 05/06/2024] [Revised: 07/19/2024] [Accepted: 07/19/2024] [Indexed: 07/28/2024]
Abstract
The prevalence of methicillin-resistant Staphylococcus aureus (MRSA), one of the most important multidrug-resistant bacteria in clinic, has become a serious global health issue. In this study, we designed and synthesized a series of griseofamine A derivatives and evaluated their antibacterial profiles. In vitro assays found that compound 9o10 showed a remarkable improvement of antibacterial activity toward MRSA (MIC = 0.0625 μg/mL), compared with griseofamine A (MIC = 8 μg/mL) and vancomycin (MIC = 0.5 μg/mL) with low hemolysis and cytotoxicity. Its rapid bactericidal property was also confirmed by time-kill curve assay. Furthermore, compound 9o10 displayed weak drug resistance frequency. In in vivo experiment, compound 9o10 exhibited more potent antibacterial efficacy than vancomycin and excellent biosafety (LD50 > 2 g/kg). Preliminary mechanism study revealed compound 9o10 might involve antibacterial mechanisms contributing to membrane damage. Taken together, compound 9o10 possessed excellent inhibitory activity against MRSA in vitro and in vivo with low toxicity and drug resistance frequency, making it a promising hit compound for further development against MRSA infections.
Collapse
Affiliation(s)
- Caiyun Ma
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100050, PR China
| | - Rao Wei
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100050, PR China
| | - Rui Yu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100050, PR China
| | - Ling Lei
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100050, PR China
| | - Xuan Pan
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100050, PR China.
| | - Hai-Yu Hu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100050, PR China.
| | - Bo Feng
- Department of Pharmacy, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, PR China.
| | - Zhanzhu Liu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100050, PR China.
| |
Collapse
|
28
|
Chen B, Wang Y, Shen S, Zhong W, Lu H, Pan Y. Lattice Defects and Electronic Modulation of Flower-Like Zn 3In 2S 6 Promote Photocatalytic Degradation of Multiple Antibiotics. SMALL METHODS 2024; 8:e2301598. [PMID: 38168900 DOI: 10.1002/smtd.202301598] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 12/12/2023] [Indexed: 01/05/2024]
Abstract
Photocatalysis is an effective technique to remove antibiotic residues from aquatic environments. Typical metal sulfides like Zn3In2S6 have been applied to a wide range of photocatalytic applications. However, there are currently no readily accessible methods to increase its antibiotic-degrading activity. Here, a facile hydrothermal approach is developed for the preparation of flower-like Zn3In2S6 with tunable sulfur lattice defects. Photogenerated carriers can be separated and transferred more easily when there is an adequate amount of lattice defects. Moreover, lattice defect-induced electronic modulation enhances light utilization and adsorption properties. The modified Zn3In2S6 demonstrates outstanding photocatalytic degradation activity for levofloxacin, ofloxacin, and tetracycline. This work sheds light on exploring metal sulfides with sulfur lattice defects for enhancing photocatalytic activity.
Collapse
Affiliation(s)
- Baofu Chen
- Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Zhejiang, 318000, China
| | - Yichao Wang
- Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Zhejiang, 318000, China
| | - Shijie Shen
- Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Zhejiang, 318000, China
| | - Wenwu Zhong
- Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Zhejiang, 318000, China
| | - Hongsheng Lu
- Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Zhejiang, 318000, China
| | - Yin Pan
- Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Zhejiang, 318000, China
| |
Collapse
|
29
|
Zhou X, Zhang H, Zhou Y, Yi Y, Yuan R, Pu W, Wang S, Shang R. Antimicrobial activity, safety and pharmacokinetics evaluation of PMTM: A novel pleuromutilin candidate. Biomed Pharmacother 2024; 179:117378. [PMID: 39241564 DOI: 10.1016/j.biopha.2024.117378] [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: 07/03/2024] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 09/09/2024] Open
Abstract
The prevalence of infections by methicillin-resistant Staphylococcus aureus (MRSA) has led to dramatically increased mortality and threated the public health worldwide. Pleuromutilin compound 14-O-[(4-(pyrrolidine-1-yl)-6-methylpyrimidine-2-yl) thioacetyl] mutilin (PMTM) is a new antibacterial agent with excellent antibacterial efficacy against Gram positive bacteria. For further developing PMTM as a potential drug against MRSA infections, the in vitro antibacterial efficacy and preclinical safety were explored in this study. The results revealed that PMTM presented the higher anti-MRSA activity, increasing post-antibiotic effect (PAE) and limited potential to develop resistance. In safety evaluation, PMTM demonstrated low cytotoxicity, poor hemolytic activity, tolerable oral acute toxic effects in rats, devoid of mutagenic response and weak inhibitory potential on CYP3A4, but displayed moderate potential hERG K+ channel inhibition. Furthermore, two salts of PMTM with sulfuric acid and hydrochloric acid were prepared and confirmed. The sulfate salt of PMTM exhibited the highest solubility based on powder dissolution experiments and was chosen to evaluate pharmacokinetics properties, in which it displayed improved mouse pharmacokinetics parameters and oral bioavailability. The present study successfully provides a good foundation of PMTM for new antibacterial drug development.
Collapse
Affiliation(s)
- Xingqian Zhou
- Key Laboratory of New Animal Drug Project, Gansu Province/Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs/Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou 730050, China.
| | - Hongjuan Zhang
- Key Laboratory of New Animal Drug Project, Gansu Province/Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs/Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou 730050, China.
| | - Yuhang Zhou
- Key Laboratory of New Animal Drug Project, Gansu Province/Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs/Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou 730050, China
| | - Yunpeng Yi
- Shandong Provincial Animal and Poultry Green Health Products Creation Engineering Laboratory, Institute of Poultry Science, Shandong Academy of Agricultural Science, Jinan, Shandong 250023, China
| | - Ruili Yuan
- Key Laboratory of New Animal Drug Project, Gansu Province/Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs/Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou 730050, China
| | - Wanxia Pu
- Key Laboratory of New Animal Drug Project, Gansu Province/Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs/Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou 730050, China
| | - Shengyi Wang
- Key Laboratory of New Animal Drug Project, Gansu Province/Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs/Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou 730050, China.
| | - Ruofeng Shang
- Key Laboratory of New Animal Drug Project, Gansu Province/Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs/Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou 730050, China.
| |
Collapse
|
30
|
Khan AH, Bilal M, Mahmood A, Rasool N, Qamar MU, Imran M, Toma SI, Andreescu O. Facile Synthesis of N-(4-Bromo-3-methylphenyl)pyrazine-2-carboxamide Derivatives, Their Antibacterial Activities against Clinically Isolated XDR S. Typhi, Alkaline Phosphatase Inhibitor Activities, and Docking Studies. Pharmaceuticals (Basel) 2024; 17:1241. [PMID: 39338403 PMCID: PMC11434897 DOI: 10.3390/ph17091241] [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: 07/23/2024] [Revised: 09/16/2024] [Accepted: 09/17/2024] [Indexed: 09/30/2024] Open
Abstract
The emergence of extensively drug-resistant Salmonella Typhi (XDR-S. Typhi) poses a grave public health threat due to its resistance to fluoroquinolones and third-generation cephalosporins. This resistance significantly complicates treatment options, underscoring the urgent need for new therapeutic strategies. In this study, we synthesized pyrazine carboxamides (3, 5a-5d) in good yields through the Suzuki reaction. Afterward, we evaluate their antibacterial activities against XDR-S. Typhi via the agar well diffusion method; 5d has the strongest antibacterial activity with MIC 6.25 (mg/mL). Moreover, in vitro Alkaline Phosphatase inhibitor activity was also determined; 5d is the most potent compound, with an IC50 of 1.469 ± 0.02 µM. Further, in silico studies were performed to find the type of interactions between synthesized compounds and target proteins.
Collapse
Affiliation(s)
- Abdul Hannan Khan
- Department of Chemistry, Government College University Faisalabad, Faisalabad 38000, Pakistan
| | - Muhammad Bilal
- Department of Chemistry, Government College University Faisalabad, Faisalabad 38000, Pakistan
| | - Abid Mahmood
- Department of Pharmaceutical Chemistry, Government College University Faisalabad, Faisalabad 38000, Pakistan
| | - Nasir Rasool
- Department of Chemistry, Government College University Faisalabad, Faisalabad 38000, Pakistan
| | - Muhammad Usman Qamar
- Institute of Microbiology, Faculty of Life Sciences, Government College University Faisalabad, Faisalabad 38000, Pakistan
| | - Muhammad Imran
- Chemistry Department, Faculty of Science, King Khalid University, P.O. Box 9004, Abha 61413, Saudi Arabia
| | | | - Oana Andreescu
- Faculty of Medicine, Transilvania University of Brasov, 500036 Brasov, Romania
| |
Collapse
|
31
|
Li L, Zhang Y, Liu Y, Wu Y, Wang X, Cao L, Feng X. Synthesis of Pt-MoS 2 with enhanced photothermal and peroxidase-like properties and its antibacterial application. RSC Adv 2024; 14:29428-29438. [PMID: 39297038 PMCID: PMC11409452 DOI: 10.1039/d4ra05487c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 09/05/2024] [Indexed: 09/21/2024] Open
Abstract
Despite tremendous efforts, bacterial infection and contamination remain a major clinical challenge to modern humans. Nanozyme materials with stimuli-responsive properties are expected to be powerful tools for the next generation of antibacterial therapy. Here, MoS2 nanosheet was firstly prepared by liquid phase exfoliation method, and Pt-MoS2 hybrid biomaterial was then successfully synthesized by a simple self-reduction method. The Pt decoration significantly improves the photothermal effect of MoS2 nanosheet under 808 nm NIR laser irradiation. Besides, benefiting from the formation of heterogeneous structure, the Pt-MoS2 has significantly enhanced peroxidase mimetic catalytic activity, which can kill bacteria through catalysis of H2O2 to generate antimicrobial hydroxyl radicals. Moreover, the temperature rise brought about by NIR laser stimulation further amplifies the nanozyme activity of the composites. After treatment by the synergistic platform, both Staphylococcus aureus and Escherichia coli can be effectively inhibited, demonstrating its broad-spectrum antibacterial properties. In addition, the developed antibacterial Pt-MoS2 nanozyme have the excellent biocompatibility, which makes them well suited for infection elimination in biological systems. Overall, this work shows great potential for rationally combining the multiple functions of MoS2-based nanomaterials for synergistic antibacterial therapy. In the future, the Pt-MoS2 nanozyme may find wider applications in areas such as personal healthcare or surface disinfection treatment of medical devices.
Collapse
Affiliation(s)
- Liangyu Li
- Department of Nursing, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College Hangzhou China
| | - Yueqin Zhang
- School of Public Health, Hangzhou Medical College Hangzhou China
| | - Yumeng Liu
- School of Public Health, Hangzhou Medical College Hangzhou China
| | - Yaojuan Wu
- Department of Nursing, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College Hangzhou China
| | - Xiao Wang
- School of Public Health, Hangzhou Medical College Hangzhou China
| | - Lidong Cao
- Department of Hepatobiliary & Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College Hangzhou China
- College of Mechanical Engineering, Zhejiang University Hangzhou China
| | - Xia Feng
- Department of Nursing, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College Hangzhou China
- Department of Hepatobiliary & Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College Hangzhou China
| |
Collapse
|
32
|
Banti CN, Hadjikakou SK. Silver and Gold Compounds as Antibiotics. Antibiotics (Basel) 2024; 13:850. [PMID: 39335023 PMCID: PMC11428958 DOI: 10.3390/antibiotics13090850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 08/29/2024] [Indexed: 09/30/2024] Open
Abstract
This Special Issue entitled "Silver and Gold Compounds as Antibiotics" covers a selection of recent research and review articles focused on biological inorganic chemistry [...].
Collapse
Affiliation(s)
| | - Sotiris K. Hadjikakou
- Laboratory of Biological Inorganic Chemistry, University of Ioannina, 45110 Ioannina, Greece;
| |
Collapse
|
33
|
Cheng K, Sun Y, Yu H, Hu Y, He Y, Shen Y. Staphylococcus aureus SOS response: Activation, impact, and drug targets. MLIFE 2024; 3:343-366. [PMID: 39359682 PMCID: PMC11442139 DOI: 10.1002/mlf2.12137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 03/17/2024] [Accepted: 04/10/2024] [Indexed: 10/04/2024]
Abstract
Staphylococcus aureus is a common cause of diverse infections, ranging from superficial to invasive, affecting both humans and animals. The widespread use of antibiotics in clinical treatments has led to the emergence of antibiotic-resistant strains and small colony variants. This surge presents a significant challenge in eliminating infections and undermines the efficacy of available treatments. The bacterial Save Our Souls (SOS) response, triggered by genotoxic stressors, encompasses host immune defenses and antibiotics, playing a crucial role in bacterial survival, invasiveness, virulence, and drug resistance. Accumulating evidence underscores the pivotal role of the SOS response system in the pathogenicity of S. aureus. Inhibiting this system offers a promising approach for effective bactericidal treatments and curbing the evolution of antimicrobial resistance. Here, we provide a comprehensive review of the activation, impact, and key proteins associated with the SOS response in S. aureus. Additionally, perspectives on therapeutic strategies targeting the SOS response for S. aureus, both individually and in combination with traditional antibiotics are proposed.
Collapse
Affiliation(s)
- Kaiying Cheng
- Zhejiang Key Laboratory of Medical Epigenetics, Department of Immunology and Pathogen Biology, School of Basic Medical Sciences, Affiliated Hospital of Hangzhou Normal UniversityHangzhou Normal UniversityHangzhouChina
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of MedicineZhejiang UniversityHangzhouChina
| | - Yukang Sun
- Zhejiang Key Laboratory of Medical Epigenetics, Department of Immunology and Pathogen Biology, School of Basic Medical Sciences, Affiliated Hospital of Hangzhou Normal UniversityHangzhou Normal UniversityHangzhouChina
| | - Huan Yu
- Zhejiang Key Laboratory of Medical Epigenetics, Department of Immunology and Pathogen Biology, School of Basic Medical Sciences, Affiliated Hospital of Hangzhou Normal UniversityHangzhou Normal UniversityHangzhouChina
| | - Yingxuan Hu
- Zhejiang Key Laboratory of Medical Epigenetics, Department of Immunology and Pathogen Biology, School of Basic Medical Sciences, Affiliated Hospital of Hangzhou Normal UniversityHangzhou Normal UniversityHangzhouChina
| | - Yini He
- Zhejiang Key Laboratory of Medical Epigenetics, Department of Immunology and Pathogen Biology, School of Basic Medical Sciences, Affiliated Hospital of Hangzhou Normal UniversityHangzhou Normal UniversityHangzhouChina
| | - Yuanyuan Shen
- Zhejiang Key Laboratory of Medical Epigenetics, Department of Immunology and Pathogen Biology, School of Basic Medical Sciences, Affiliated Hospital of Hangzhou Normal UniversityHangzhou Normal UniversityHangzhouChina
| |
Collapse
|
34
|
Chandrika KVSM, V P. An in silico molecular docking, ADMET and molecular dynamics simulations studies of azolyl-2H-chroman-4-ones as potential inhibitors against pathogenic fungi and bacteria. J Biomol Struct Dyn 2024; 42:7667-7685. [PMID: 37526222 DOI: 10.1080/07391102.2023.2241102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 07/20/2023] [Indexed: 08/02/2023]
Abstract
Antimicrobial resistance is a major global threat. In an attempt to discover new compounds with improved efficiency and to overcome drug resistance, a library of 3960 compounds was designed as conformationally rigid analogues of oxiconazole with 2H-chroman-4-one, azole and substituted phenyl fragments. The antifungal and antibacterial activity of the compounds was evaluated using molecular docking studies in the active site of six fungal and four bacterial proteins to establish the binding affinity of the designed ligands. In-silico ADME and Lipinski's rule were used to establish the drug-likeness properties of the compounds. This study revealed that all the designed compounds had a high binding affinity with the target proteins and formed H-bond and π-π interactions. The identified hits have been subjected to molecular dynamics simulations to study protein-ligand complex stability. This study has led to the identification of important compounds that can be developed further as therapeutic agents against pathogenic fungi and bacteria.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- K V S Mani Chandrika
- Department of Chemistry, Sri Sathya Sai Institute of Higher Learning, Anantapur Campus, Anantapur, Andhra Pradesh, India
| | - Prathyusha V
- Department of Chemistry, Sri Sathya Sai Institute of Higher Learning, Anantapur Campus, Anantapur, Andhra Pradesh, India
| |
Collapse
|
35
|
Zhang Y, Li R, Zou G, Guo Y, Wu R, Zhou Y, Chen H, Zhou R, Lavigne R, Bergen PJ, Li J, Li J. Discovery of Antimicrobial Lysins from the "Dark Matter" of Uncharacterized Phages Using Artificial Intelligence. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404049. [PMID: 38899839 PMCID: PMC11348152 DOI: 10.1002/advs.202404049] [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: 04/17/2024] [Revised: 05/29/2024] [Indexed: 06/21/2024]
Abstract
The rapid rise of antibiotic resistance and slow discovery of new antibiotics have threatened global health. While novel phage lysins have emerged as potential antibacterial agents, experimental screening methods for novel lysins pose significant challenges due to the enormous workload. Here, the first unified software package, namely DeepLysin, is developed to employ artificial intelligence for mining the vast genome reservoirs ("dark matter") for novel antibacterial phage lysins. Putative lysins are computationally screened from uncharacterized Staphylococcus aureus phages and 17 novel lysins are randomly selected for experimental validation. Seven candidates exhibit excellent in vitro antibacterial activity, with LLysSA9 exceeding that of the best-in-class alternative. The efficacy of LLysSA9 is further demonstrated in mouse bloodstream and wound infection models. Therefore, this study demonstrates the potential of integrating computational and experimental approaches to expedite the discovery of new antibacterial proteins for combating increasing antimicrobial resistance.
Collapse
Affiliation(s)
- Yue Zhang
- National Key Laboratory of Agricultural MicrobiologyKey Laboratory of Environment Correlative DietologyCollege of Biomedicine and HealthShenzhen Institute of Nutrition and HealthHuazhong Agricultural UniversityWuhan430070China
- Hubei Hongshan LaboratoryCollege of Food Science and TechnologyHuazhong Agricultural UniversityWuhan430070China
| | - Runze Li
- National Key Laboratory of Agricultural MicrobiologyKey Laboratory of Environment Correlative DietologyCollege of Biomedicine and HealthShenzhen Institute of Nutrition and HealthHuazhong Agricultural UniversityWuhan430070China
- Hubei Hongshan LaboratoryCollege of Food Science and TechnologyHuazhong Agricultural UniversityWuhan430070China
| | - Geng Zou
- National Key Laboratory of Agricultural MicrobiologyKey Laboratory of Environment Correlative DietologyCollege of Biomedicine and HealthShenzhen Institute of Nutrition and HealthHuazhong Agricultural UniversityWuhan430070China
- Hubei Hongshan LaboratoryCollege of Food Science and TechnologyHuazhong Agricultural UniversityWuhan430070China
| | - Yating Guo
- National Key Laboratory of Agricultural MicrobiologyKey Laboratory of Environment Correlative DietologyCollege of Biomedicine and HealthShenzhen Institute of Nutrition and HealthHuazhong Agricultural UniversityWuhan430070China
- College of Veterinary MedicineHuazhong Agricultural UniversityWuhan430070China
| | - Renwei Wu
- National Key Laboratory of Agricultural MicrobiologyKey Laboratory of Environment Correlative DietologyCollege of Biomedicine and HealthShenzhen Institute of Nutrition and HealthHuazhong Agricultural UniversityWuhan430070China
- College of Veterinary MedicineHuazhong Agricultural UniversityWuhan430070China
| | - Yang Zhou
- National Key Laboratory of Agricultural MicrobiologyKey Laboratory of Environment Correlative DietologyCollege of Biomedicine and HealthShenzhen Institute of Nutrition and HealthHuazhong Agricultural UniversityWuhan430070China
| | - Huanchun Chen
- National Key Laboratory of Agricultural MicrobiologyKey Laboratory of Environment Correlative DietologyCollege of Biomedicine and HealthShenzhen Institute of Nutrition and HealthHuazhong Agricultural UniversityWuhan430070China
- College of Veterinary MedicineHuazhong Agricultural UniversityWuhan430070China
| | - Rui Zhou
- National Key Laboratory of Agricultural MicrobiologyKey Laboratory of Environment Correlative DietologyCollege of Biomedicine and HealthShenzhen Institute of Nutrition and HealthHuazhong Agricultural UniversityWuhan430070China
- College of Veterinary MedicineHuazhong Agricultural UniversityWuhan430070China
| | - Rob Lavigne
- Department of BiosystemsLaboratory of Gene TechnologyKU LeuvenLeuven3001Belgium
| | - Phillip J. Bergen
- Monash Biomedicine Discovery InstituteDepartment of MicrobiologyFaculty of MedicineNursing and Health SciencesMonash UniversityMelbourne3800Australia
| | - Jian Li
- Monash Biomedicine Discovery InstituteDepartment of MicrobiologyFaculty of MedicineNursing and Health SciencesMonash UniversityMelbourne3800Australia
| | - Jinquan Li
- National Key Laboratory of Agricultural MicrobiologyKey Laboratory of Environment Correlative DietologyCollege of Biomedicine and HealthShenzhen Institute of Nutrition and HealthHuazhong Agricultural UniversityWuhan430070China
- Hubei Hongshan LaboratoryCollege of Food Science and TechnologyHuazhong Agricultural UniversityWuhan430070China
- College of Veterinary MedicineHuazhong Agricultural UniversityWuhan430070China
- Shenzhen BranchGuangdong Laboratory for Lingnan Modern AgricultureGenome Analysis Laboratory of the Ministry of Agriculture and Rural AffairsAgricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhen518000China
| |
Collapse
|
36
|
Matos RG, Simmons KJ, Fishwick CWG, McDowall KJ, Arraiano CM. Identification of Ribonuclease Inhibitors for the Control of Pathogenic Bacteria. Int J Mol Sci 2024; 25:8048. [PMID: 39125622 PMCID: PMC11311990 DOI: 10.3390/ijms25158048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/19/2024] [Accepted: 07/21/2024] [Indexed: 08/12/2024] Open
Abstract
Bacteria are known to be constantly adapting to become resistant to antibiotics. Currently, efficient antibacterial compounds are still available; however, it is only a matter of time until these compounds also become inefficient. Ribonucleases are the enzymes responsible for the maturation and degradation of RNA molecules, and many of them are essential for microbial survival. Members of the PNPase and RNase II families of exoribonucleases have been implicated in virulence in many pathogens and, as such, are valid targets for the development of new antibacterials. In this paper, we describe the use of virtual high-throughput screening (vHTS) to identify chemical compounds predicted to bind to the active sites within the known structures of RNase II and PNPase from Escherichia coli. The subsequent in vitro screening identified compounds that inhibited the activity of these exoribonucleases, with some also affecting cell viability, thereby providing proof of principle for utilizing the known structures of these enzymes in the pursuit of new antibacterials.
Collapse
Affiliation(s)
- Rute G. Matos
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República, 2780-157 Oeiras, Portugal
| | - Katie J. Simmons
- Astbury Centre for Structural Molecular Biology, School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK;
| | - Colin W. G. Fishwick
- Astbury Centre for Structural Molecular Biology, School of Chemistry, Faculty of Engineering and Physical Sciences, University of Leeds, Leeds LS2 9JT, UK;
| | - Kenneth J. McDowall
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK;
| | - Cecília M. Arraiano
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República, 2780-157 Oeiras, Portugal
| |
Collapse
|
37
|
Parkhill SL, Johnson EO. Integrating bacterial molecular genetics with chemical biology for renewed antibacterial drug discovery. Biochem J 2024; 481:839-864. [PMID: 38958473 PMCID: PMC11346456 DOI: 10.1042/bcj20220062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/20/2024] [Accepted: 06/24/2024] [Indexed: 07/04/2024]
Abstract
The application of dyes to understanding the aetiology of infection inspired antimicrobial chemotherapy and the first wave of antibacterial drugs. The second wave of antibacterial drug discovery was driven by rapid discovery of natural products, now making up 69% of current antibacterial drugs. But now with the most prevalent natural products already discovered, ∼107 new soil-dwelling bacterial species must be screened to discover one new class of natural product. Therefore, instead of a third wave of antibacterial drug discovery, there is now a discovery bottleneck. Unlike natural products which are curated by billions of years of microbial antagonism, the vast synthetic chemical space still requires artificial curation through the therapeutics science of antibacterial drugs - a systematic understanding of how small molecules interact with bacterial physiology, effect desired phenotypes, and benefit the host. Bacterial molecular genetics can elucidate pathogen biology relevant to therapeutics development, but it can also be applied directly to understanding mechanisms and liabilities of new chemical agents with new mechanisms of action. Therefore, the next phase of antibacterial drug discovery could be enabled by integrating chemical expertise with systematic dissection of bacterial infection biology. Facing the ambitious endeavour to find new molecules from nature or new-to-nature which cure bacterial infections, the capabilities furnished by modern chemical biology and molecular genetics can be applied to prospecting for chemical modulators of new targets which circumvent prevalent resistance mechanisms.
Collapse
Affiliation(s)
- Susannah L. Parkhill
- Systems Chemical Biology of Infection and Resistance Laboratory, The Francis Crick Institute, London, U.K
- Faculty of Life Sciences, University College London, London, U.K
| | - Eachan O. Johnson
- Systems Chemical Biology of Infection and Resistance Laboratory, The Francis Crick Institute, London, U.K
- Faculty of Life Sciences, University College London, London, U.K
- Department of Chemistry, Imperial College, London, U.K
- Department of Chemistry, King's College London, London, U.K
| |
Collapse
|
38
|
Qi XX, Wang PP, Cui LT, Jin M, Zhao LX, Li G. Synthesis, characterization and in vitro antiproliferative effects of isosteviol derivatives. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2024; 26:812-823. [PMID: 38477295 DOI: 10.1080/10286020.2024.2324082] [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: 04/25/2023] [Accepted: 12/01/2023] [Indexed: 03/14/2024]
Abstract
Nineteen isosteviol derivatives were designed and synthesized by C-16, C-19 and D-ring modifications of isosteviol. These compounds were screened for their cytotoxic activities against Hela and A549 cells in vitro. Among them, the inhibitory effect of compounds 3b and 16 on Hela cells was comparable to that of the positive control gefitinib, and the compounds 3b (IC50=7.84 ± 0.84 μM) and 7a (IC50=6.89 ± 0.33 μM) exhibited significant cytotoxicity superior to gefitinib (IC50=11.02 ± 3.27 μM) against A549 cells.
Collapse
Affiliation(s)
- Xin-Xin Qi
- School of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian 116029, China
| | - Pan-Pan Wang
- School of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian 116029, China
| | - Lan-Tian Cui
- Key Laboratory of Natural Resources of Changbai Mountain and Functional Molecules, Ministry of Education, College of Pharmacy, Yanbian University, Yanji 133002, China
| | - Mei Jin
- Key Laboratory of Natural Resources of Changbai Mountain and Functional Molecules, Ministry of Education, College of Pharmacy, Yanbian University, Yanji 133002, China
| | - Long-Xuan Zhao
- School of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian 116029, China
| | - Gao Li
- Key Laboratory of Natural Resources of Changbai Mountain and Functional Molecules, Ministry of Education, College of Pharmacy, Yanbian University, Yanji 133002, China
| |
Collapse
|
39
|
Amin A, Naim MD, Islam N, Mollah MNH. Genome-wide identification and characterization of DTX family genes highlighting their locations, functions, and regulatory factors in banana (Musa acuminata). PLoS One 2024; 19:e0303065. [PMID: 38843276 PMCID: PMC11156367 DOI: 10.1371/journal.pone.0303065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Accepted: 04/19/2024] [Indexed: 06/09/2024] Open
Abstract
The detoxification efflux carriers (DTX) are a significant group of multidrug efflux transporter family members that play diverse functions in all kingdoms of living organisms. However, genome-wide identification and characterization of DTX family transporters have not yet been performed in banana, despite its importance as an economic fruit plant. Therefore, a detailed genome-wide analysis of DTX family transporters in banana (Musa acuminata) was conducted using integrated bioinformatics and systems biology approaches. In this study, a total of 37 DTX transporters were identified in the banana genome and divided into four groups (I, II, III, and IV) based on phylogenetic analysis. The gene structures, as well as their proteins' domains and motifs, were found to be significantly conserved. Gene ontology (GO) annotation revealed that the predicted DTX genes might play a vital role in protecting cells and membrane-bound organelles through detoxification mechanisms and the removal of drug molecules from banana cells. Gene regulatory analyses identified key transcription factors (TFs), cis-acting elements, and post-transcriptional regulators (miRNAs) of DTX genes, suggesting their potential roles in banana. Furthermore, the changes in gene expression levels due to pathogenic infections and non-living factor indicate that banana DTX genes play a role in responses to both biotic and abiotic stresses. The results of this study could serve as valuable tools to improve banana quality by protecting them from a range of environmental stresses.
Collapse
Affiliation(s)
- Al Amin
- Department of Statistics, Bioinformatics Laboratory, Faculty of Science, University of Rajshahi, Rajshahi, Bangladesh
- Department of Zoology, Faculty of Biological Sciences, University of Rajshahi, Rajshahi, Bangladesh
| | - Md. Darun Naim
- Department of Botany, Faculty of Biological Sciences, University of Rajshahi, Rajshahi, Bangladesh
| | - Nurul Islam
- Department of Zoology, Faculty of Biological Sciences, University of Rajshahi, Rajshahi, Bangladesh
| | - Md. Nurul Haque Mollah
- Department of Statistics, Bioinformatics Laboratory, Faculty of Science, University of Rajshahi, Rajshahi, Bangladesh
| |
Collapse
|
40
|
Bahatheg G, Kuppusamy R, Yasir M, Bridge S, Mishra SK, Cranfield CG, StC Black D, Willcox M, Kumar N. Dimeric peptoids as antibacterial agents. Bioorg Chem 2024; 147:107334. [PMID: 38583251 DOI: 10.1016/j.bioorg.2024.107334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 03/31/2024] [Accepted: 04/02/2024] [Indexed: 04/09/2024]
Abstract
Building upon our previous study on peptoid-based antibacterials which showed good activity against Gram-positive bacteria only, herein we report the synthesis of 34 dimeric peptoid compounds and the investigation of their activity against Gram-positive and Gram-negative pathogens. The newly designed peptoids feature a di-hydrophobic moiety incorporating phenyl, bromo-phenyl, and naphthyl groups, combined with variable lengths of cationic units such as amino and guanidine groups. The study also underscores the pivotal interplay between hydrophobicity and cationicity in optimizing efficacy against specific bacteria. The bromophenyl dimeric guanidinium peptoid compound 10j showed excellent activity against S. aureus 38 and E. coli K12 with MIC of 0.8 μg mL-1 and 6.2 μg mL-1, respectively. Further investigation into the mechanism of action revealed that the antibacterial effect might be attributed to the disruption of bacterial cell membranes, as suggested by tethered bilayer lipid membranes (tBLMs) and cytoplasmic membrane permeability studies. Notably, these promising antibacterial agents exhibited negligible toxicity against mammalian red blood cells. Additionally, the study explored the potential of 12 active compounds to disrupt established biofilms of S. aureus 38. The most effective biofilm disruptors were ethyl and octyl-naphthyl guanidinium peptoids (10c and 10 k). These compounds 10c and 10 k disrupted the established biofilms of S. aureus 38 with 51 % at 4x MIC (MIC = 17.6 μg mL-1 and 11.2 μg mL-1) and 56 %-58 % at 8x MIC (MIC = 35.2 μg mL-1 and 22.4 μg mL-1) respectively. Overall, this research contributes insights into the design principles of cationic dimeric peptoids and their antibacterial activity, with implications for the development of new antibacterial compounds.
Collapse
Affiliation(s)
- Ghayah Bahatheg
- School of Chemistry, The University of New South Wales (UNSW), Sydney, NSW 2052, Australia; Department of Chemistry, Faculty of Science, University of Jeddah, Jeddah 21589, Saudi Arabia
| | - Rajesh Kuppusamy
- School of Chemistry, The University of New South Wales (UNSW), Sydney, NSW 2052, Australia; School of Optometry and Vision Science, The University of New South Wales (UNSW), Sydney, NSW 2052, Australia.
| | - Muhammad Yasir
- School of Optometry and Vision Science, The University of New South Wales (UNSW), Sydney, NSW 2052, Australia
| | - Samara Bridge
- School of Life Sciences, University of Technology Sydney, PO Box 123, Ultimo 2007, Australia
| | - Shyam K Mishra
- School of Optometry and Vision Science, The University of New South Wales (UNSW), Sydney, NSW 2052, Australia
| | - Charles G Cranfield
- School of Life Sciences, University of Technology Sydney, PO Box 123, Ultimo 2007, Australia
| | - David StC Black
- School of Chemistry, The University of New South Wales (UNSW), Sydney, NSW 2052, Australia
| | - Mark Willcox
- School of Optometry and Vision Science, The University of New South Wales (UNSW), Sydney, NSW 2052, Australia
| | - Naresh Kumar
- School of Chemistry, The University of New South Wales (UNSW), Sydney, NSW 2052, Australia.
| |
Collapse
|
41
|
He C, Bi S, Zhang R, Chen C, Liu R, Zhao X, Gu J, Yan B. A hyaluronic acid hydrogel as a mild photothermal antibacterial, antioxidant, and nitric oxide release platform for diabetic wound healing. J Control Release 2024; 370:543-555. [PMID: 38729434 DOI: 10.1016/j.jconrel.2024.05.011] [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: 02/17/2024] [Revised: 05/04/2024] [Accepted: 05/06/2024] [Indexed: 05/12/2024]
Abstract
Hyaluronic acid (HA)-based biopolymer hydrogels are promising therapeutic dressings for various wounds but still underperform in treating diabetic wounds. These wounds are extremely difficult to heal and undergo a prolonged and severe inflammatory process due to bacterial infection, overexpression of reactive oxygen species (ROS), and insufficient synthesis of NO. In this study, a dynamic crosslinked hyaluronic acid (HA) hydrogel dressing (Gel-HAB) loaded with allomelanin (AMNP)-N, N'-dis-sec-butyl-N, N'-dinitroso-1, 4-phenylenediamine (BNN6) nanoparticles (AMNP-BNN6) was developed for healing diabetic wounds. The dynamic acylhydrazone bond formed between hydrazide-modified HA (HA-ADH) and oxidized HA (OHA) makes the hydrogel injectable, self-healing, and biocompatible. The hydrogel, loaded with AMNP-BNN6 nanoparticles, exhibits promising ROS scavenging ability and on-demand release of nitric oxide (NO) under near-infrared (NIR) laser irradiation to achieve mild photothermal antibacterial therapy (PTAT) (∼ 48 °C). Notably, the Gel-HAB hydrogel effectively reduced the oxidative stress level, controlled infections, accelerated vascular regeneration, and promoted angiogenesis, thereby achieving rapid healing of diabetic wounds. The injectable self-healing nanocomposite hydrogel could serve as a mild photothermal-enhanced antibacterial, antioxidant, and nitric oxide release platform for the treatment of diabetic wounds.
Collapse
Affiliation(s)
- Changyuan He
- College of Biomass Science and Engineering, Sichuan University, Chengdu 610000, China
| | - Siwei Bi
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Rongya Zhang
- Technology Center, China Tobacco Sichuan Industrial Co. Ltd., Chengdu 610066, China
| | - Chong Chen
- College of Biomass Science and Engineering, Sichuan University, Chengdu 610000, China
| | - Ruiqi Liu
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Xueshan Zhao
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Jun Gu
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu 610000, China.
| | - Bin Yan
- College of Biomass Science and Engineering, Sichuan University, Chengdu 610000, China.
| |
Collapse
|
42
|
Yolmeh M, Khomeiri M, Ghaemi E, Şilbir MS. Acute toxicity and anti-enterotoxigenic activity of pigment extracted from Micrococcus roseus. Braz J Microbiol 2024; 55:1753-1758. [PMID: 38532186 PMCID: PMC11153400 DOI: 10.1007/s42770-024-01316-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 03/21/2024] [Indexed: 03/28/2024] Open
Abstract
Microbial pigments are considered as one of the main sources of natural types, and the attention to them is increasing in the food and pharmaceutical industries. This study aimed to investigate the effects of pigments extracted from Micrococcus roseus (PEM) on the gene expression of a and b staphylococcal enterotoxins (sea and seb) and their acute toxicity. Real-time PCR was used to study the anti-enterotoxigenic activity of PEM against Staphylococcus aureus at sub-inhibitory concentrations. In addition, the acute toxicity of PEM was evaluated on albino mice through alkaline phosphatase (ALP), aspartate aminotransferas (AST), and alanine aminotransferase (ALT) of liver and its histopathological changes. Based on the results, the expression of sea and seb was decreased in the presence of PEM at sub-inhibitory concentrations. The 2-∆∆CT was measured 0.02 and 0.01 for the expression of sea and seb of S. aureus grown in the MHB containing 16 mg/ml PEM. The results showed that the expression of seb is more sensitive to PEM compared to the expression of sea. After treatment of mice with PEM for two weeks, the condition of mice was normal, and the results of liver enzymatic activities and histopathological changes showed insignificant difference compared to the control sample.
Collapse
Affiliation(s)
- Mahmoud Yolmeh
- Department of Food Science and Nutrition, Faculty of Food Engineering, University of Campinas, Rua Monteiro Lobato, 80, Cidade Universitária Zeferino Vaz, Campinas, SP, CEP: 13083-862, Brazil.
| | - Morteza Khomeiri
- Department of Food Science and Technology, Gorgan University of Agricultural Sciences and Natural Resources, Gorgan, Iran
| | - Ezzatollah Ghaemi
- Infectious Research Centre and Microbiology Department, Golestan University of Medical Sciences and Health Services, Gorgan, Iran
| | - Mehmet Selim Şilbir
- Department of Food Engineering, Faculty of Engineering, Iğdır University, Iğdır, Turkey
| |
Collapse
|
43
|
Sil M, Mukherjee D, Goswami A, Nag M, Lahiri D, Bhattacharya D. Antibiofilm activity of mesoporous silica nanoparticles against the biofilm associated infections. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:3617-3633. [PMID: 38051365 DOI: 10.1007/s00210-023-02872-0] [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: 09/21/2023] [Accepted: 11/21/2023] [Indexed: 12/07/2023]
Abstract
In pharmaceutical industries, various chemical carriers are present which are used for drug delivery to the correct target sites. The most popular and upcoming drug delivery carriers are mesoporous silica nanoparticles (MSN). The main reason for its popularity is its ability to be specific and optimize the drug delivery process in a controlled manner. Nowadays, MSNs are widely used to eradicate various microbial infections, especially the ones related to biofilms. Biofilms are sessile groups of cells that live by forming a consortium and exhibit antibacterial resistance (AMR). They exhibit AMR by extracellular polymeric substances (EPS) and various quorum sensing (QS) signaling molecules. Usually, bacterial and fungal cells are capable of forming biofilms. These biofilms are pathogenic. In the majority of the cases, biofilms cause nosocomial diseases. This review will focus on the antibiofilm activities of MSN, its mechanism of target-specific drug delivery, and its ability to disrupt the bacterial biofilms inhibiting the infection. The review will also discuss various mechanisms for the delivery of pharmaceutical molecules by the MSNs to inhibit the bacterial biofilms, and lastly, we will talk about the different types of MSNs and their antibiofilm activities.
Collapse
Affiliation(s)
- Moumita Sil
- Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, India
| | - Dipro Mukherjee
- Agricultural and Ecological Research Unit, Biological Sciences Division, Indian Statistical Institute, Kolkata, India
| | - Arunava Goswami
- Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, India
| | - Moupriya Nag
- Department of Biotechnology, Institute of Engineering and Management, Kolkata, New Town, University of Engineering and Management, Kolkata, India
| | - Dibyajit Lahiri
- Department of Biotechnology, Institute of Engineering and Management, Kolkata, New Town, University of Engineering and Management, Kolkata, India.
| | - Debasmita Bhattacharya
- Department of Basic Science and Humanities, Institute of Engineering and Management, Kolkata, Salt Lake, University of Engineering and Management, Kolkata, India
| |
Collapse
|
44
|
Abaszadeh N, Afzali D, Sargazi G, Golpayegani A. Sonochemical-assisted method for efficient synthesis of Cu-MOF and evaluating its antibacterial properties. Heliyon 2024; 10:e31024. [PMID: 38779023 PMCID: PMC11109762 DOI: 10.1016/j.heliyon.2024.e31024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 05/08/2024] [Accepted: 05/09/2024] [Indexed: 05/25/2024] Open
Abstract
Sonochemical-assisted method was used to synthesize copper metal-organic frameworks (Cu-MOF) nanostructures. The final products were examined by related techniques such as XRD patterns, SEM image, BET N2 adsorption/desorption technique and FTIR spectrum. Microtiter plates microbiological assay were used to investigate antibacterial properties and the results were analyzed using ANOVA and Tukey HSD tests. The results showed that Cu-MOF nanostructures have a mesoporous nature with an average particle size distribution around 60 nm. The final product had the property of preventing the growth of all tested bacteria in certain concentrations. Minimum Inhibitory Concentration (MIC) values were observed in the range of 30-100 ppm. It was also discovered that this nanostructure can not kill bacteria completely. In addition, the minimal inhibitory concentration for biofilm growth (MIC-B) of the nanostructure was investigated. The MIC-B analyzes demonstrated that the growth of bacterial biofilm decreased with increasing Cu-MOF concentration.
Collapse
Affiliation(s)
- Nafiseh Abaszadeh
- Department of Nanotechnology, Graduate University of Advanced Technology, Kerman, Iran
| | - Daryoush Afzali
- Department of Environment, Institute of Science and High Technology and Environmental Sciences, Graduate University of Advanced Technology, Kerman, Iran
| | - Ghasem Sargazi
- Non-Communicable Diseases Research Center, Bam University of Medical Sciences, Bam, Iran
| | - Abdolali Golpayegani
- Department of Environmental Health Engineering, School of Public Health, Bam University of Medical Sciences, Bam, Iran
| |
Collapse
|
45
|
Li J, Her AS, Besch A, Ramirez-Cordero B, Crames M, Banigan JR, Mueller C, Marsiglia WM, Zhang Y, Traaseth NJ. Dynamics underlie the drug recognition mechanism by the efflux transporter EmrE. Nat Commun 2024; 15:4537. [PMID: 38806470 PMCID: PMC11133458 DOI: 10.1038/s41467-024-48803-2] [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: 12/12/2023] [Accepted: 05/14/2024] [Indexed: 05/30/2024] Open
Abstract
The multidrug efflux transporter EmrE from Escherichia coli requires anionic residues in the substrate binding pocket for coupling drug transport with the proton motive force. Here, we show how protonation of a single membrane embedded glutamate residue (Glu14) within the homodimer of EmrE modulates the structure and dynamics in an allosteric manner using NMR spectroscopy. The structure of EmrE in the Glu14 protonated state displays a partially occluded conformation that is inaccessible for drug binding by the presence of aromatic residues in the binding pocket. Deprotonation of a single Glu14 residue in one monomer induces an equilibrium shift toward the open state by altering its side chain position and that of a nearby tryptophan residue. This structural change promotes an open conformation that facilitates drug binding through a conformational selection mechanism and increases the binding affinity by approximately 2000-fold. The prevalence of proton-coupled exchange in efflux systems suggests a mechanism that may be shared in other antiporters where acid/base chemistry modulates access of drugs to the substrate binding pocket.
Collapse
Affiliation(s)
- Jianping Li
- Department of Chemistry, New York University, New York, NY, USA
| | - Ampon Sae Her
- Department of Chemistry, New York University, New York, NY, USA
| | - Alida Besch
- Department of Chemistry, New York University, New York, NY, USA
| | | | - Maureen Crames
- Department of Chemistry, New York University, New York, NY, USA
| | - James R Banigan
- Department of Chemistry, New York University, New York, NY, USA
| | - Casey Mueller
- Department of Chemistry, New York University, New York, NY, USA
| | | | - Yingkai Zhang
- Department of Chemistry, New York University, New York, NY, USA
- Simons Center for Computational Physical Chemistry, New York University, New York, NY, USA
| | | |
Collapse
|
46
|
Koller TO, Berger MJ, Morici M, Paternoga H, Bulatov T, Di Stasi A, Dang T, Mainz A, Raulf K, Crowe-McAuliffe C, Scocchi M, Mardirossian M, Beckert B, Vázquez-Laslop N, Mankin A, Süssmuth RD, Wilson DN. Paenilamicins from the honey bee pathogen Paenibacillus larvae are context-specific translocation inhibitors of protein synthesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.21.595107. [PMID: 38826346 PMCID: PMC11142091 DOI: 10.1101/2024.05.21.595107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
The paenilamicins are a group of hybrid non-ribosomal peptide-polyketide compounds produced by the honey bee pathogen Paenibacillus larvae that display activity against Gram-positive pathogens, such as Staphylococcus aureus. While paenilamicins have been shown to inhibit protein synthesis, their mechanism of action has remained unclear. Here, we have determined structures of the paenilamicin PamB2 stalled ribosomes, revealing a unique binding site on the small 30S subunit located between the A- and P-site tRNAs. In addition to providing a precise description of interactions of PamB2 with the ribosome, the structures also rationalize the resistance mechanisms utilized by P. larvae. We could further demonstrate that PamB2 interferes with the translocation of mRNA and tRNAs through the ribosome during translation elongation, and that this inhibitory activity is influenced by the presence of modifications at position 37 of the A-site tRNA. Collectively, our study defines the paenilamicins as a new class of context-specific translocation inhibitors.
Collapse
Affiliation(s)
- Timm O. Koller
- Institute for Biochemistry and Molecular Biology, University of Hamburg, Martin-Luther-King-Platz 6, 20146 Hamburg, Germany
| | - Max J. Berger
- Institute for Biochemistry and Molecular Biology, University of Hamburg, Martin-Luther-King-Platz 6, 20146 Hamburg, Germany
| | - Martino Morici
- Institute for Biochemistry and Molecular Biology, University of Hamburg, Martin-Luther-King-Platz 6, 20146 Hamburg, Germany
| | - Helge Paternoga
- Institute for Biochemistry and Molecular Biology, University of Hamburg, Martin-Luther-King-Platz 6, 20146 Hamburg, Germany
| | - Timur Bulatov
- Institut für Chemie, Technische Universität Berlin, 10623 Berlin, Germany
| | - Adriana Di Stasi
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
| | - Tam Dang
- Institut für Chemie, Technische Universität Berlin, 10623 Berlin, Germany
| | - Andi Mainz
- Institut für Chemie, Technische Universität Berlin, 10623 Berlin, Germany
| | - Karoline Raulf
- Institute for Biochemistry and Molecular Biology, University of Hamburg, Martin-Luther-King-Platz 6, 20146 Hamburg, Germany
| | - Caillan Crowe-McAuliffe
- Institute for Biochemistry and Molecular Biology, University of Hamburg, Martin-Luther-King-Platz 6, 20146 Hamburg, Germany
| | - Marco Scocchi
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
| | - Mario Mardirossian
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
| | - Bertrand Beckert
- Dubochet Center for Imaging (DCI) at EPFL, EPFL SB IPHYS DCI, Lausanne, Switzerland
| | - Nora Vázquez-Laslop
- Center for Biomolecular Sciences, University of Illinois at Chicago, Chicago, IL 60607
| | - Alexander Mankin
- Center for Biomolecular Sciences, University of Illinois at Chicago, Chicago, IL 60607
| | | | - Daniel N. Wilson
- Institute for Biochemistry and Molecular Biology, University of Hamburg, Martin-Luther-King-Platz 6, 20146 Hamburg, Germany
| |
Collapse
|
47
|
Li J, Hu S, Feng P, Xia Y, Pei Z, Tian J, Jiang K, Liu L, Cai X, Wu P. Brucine Sulfate, a Novel Bacteriostatic Agent in 3D Printed Bone Scaffold Systems. Polymers (Basel) 2024; 16:1428. [PMID: 38794621 PMCID: PMC11124991 DOI: 10.3390/polym16101428] [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: 03/31/2024] [Revised: 04/28/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
Bacterial infection is a common complication in bone defect surgery, in which infection by clinically resistant bacteria has been a challenge for the medical community. Given this emerging problem, the discovery of novel natural-type inhibitors of drug-resistant bacteria has become imperative. Brucine, present in the traditional Chinese herb Strychnine semen, is reported to exert analgesic and anti-inflammatory effects. Brucine's clinical application was limited because of its water solubility. We extracted high-purity BS by employing reflux extraction and crystallization, greatly improved its solubility, and evaluated its antimicrobial activity against E. coli and S. aureus. Importantly, we found that BS inhibited the drug-resistant strains significantly better than standard strains and achieved sterilization by disrupting the bacterial cell wall. Considering the safety concerns associated with the narrow therapeutic window of BS, a 3D BS-PLLA/PGA bone scaffold system was constructed with SLS technology and tested for its performance, bacteriostatic behaviors, and biocompatibility. The results have shown that the drug-loaded bone scaffolds had not only long-term, slow-controlled release with good cytocompatibility but also demonstrated significant antimicrobial activity in antimicrobial testing. The above results indicated that BS may be a potential drug candidate for the treatment of antibiotic-resistant bacterial infections and that scaffolds with enhanced antibacterial activity and mechanical properties may have potential applications in bone tissue engineering.
Collapse
Affiliation(s)
- Jinying Li
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410208, China; (J.L.); (Y.X.); (Z.P.); (J.T.); (K.J.)
| | - Shi Hu
- State Key Laboratory of Precision Manufacturing for Extreme Service Performance, College of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China; (S.H.); (P.F.)
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Pei Feng
- State Key Laboratory of Precision Manufacturing for Extreme Service Performance, College of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China; (S.H.); (P.F.)
| | - Yang Xia
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410208, China; (J.L.); (Y.X.); (Z.P.); (J.T.); (K.J.)
- Changde First Chinese Medicine Hospital, Changde 415000, China
| | - Zihan Pei
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410208, China; (J.L.); (Y.X.); (Z.P.); (J.T.); (K.J.)
| | - Jiaxuan Tian
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410208, China; (J.L.); (Y.X.); (Z.P.); (J.T.); (K.J.)
| | - Kun Jiang
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410208, China; (J.L.); (Y.X.); (Z.P.); (J.T.); (K.J.)
| | - Liang Liu
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410208, China; (J.L.); (Y.X.); (Z.P.); (J.T.); (K.J.)
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Xiong Cai
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410208, China; (J.L.); (Y.X.); (Z.P.); (J.T.); (K.J.)
| | - Ping Wu
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410208, China; (J.L.); (Y.X.); (Z.P.); (J.T.); (K.J.)
| |
Collapse
|
48
|
Hofer W, Deschner F, Jézéquel G, Pessanha de Carvalho L, Abdel-Wadood N, Pätzold L, Bernecker S, Morgenstern B, Kany AM, Große M, Stadler M, Bischoff M, Hirsch AKH, Held J, Herrmann J, Müller R. Functionalization of Chlorotonils: Dehalogenil as Promising Lead Compound for In Vivo Application. Angew Chem Int Ed Engl 2024; 63:e202319765. [PMID: 38502093 DOI: 10.1002/anie.202319765] [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: 12/21/2023] [Revised: 02/15/2024] [Accepted: 02/22/2024] [Indexed: 03/20/2024]
Abstract
The natural product chlorotonil displays high potency against multidrug-resistant Gram-positive bacteria and Plasmodium falciparum. Yet, its scaffold is characterized by low solubility and oral bioavailability, but progress was recently made to enhance these properties. Applying late-stage functionalization, we aimed to further optimize the molecule. Previously unknown reactions including a sulfur-mediated dehalogenation were revealed. Dehalogenil, the product of this reaction, was identified as the most promising compound so far, as this new derivative displayed improved solubility and in vivo efficacy while retaining excellent antimicrobial activity. We confirmed superb activity against multidrug-resistant clinical isolates of Staphylococcus aureus and Enterococcus spp. and mature transmission stages of Plasmodium falciparum. We also demonstrated favorable in vivo toxicity, pharmacokinetics and efficacy in infection models with S. aureus. Taken together, these results identify dehalogenil as an advanced lead molecule.
Collapse
Affiliation(s)
- Walter Hofer
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI) and Department of Pharmacy at Saarland University Campus Building E8.1, 66123, Saarbrücken, Germany
- German Centre for Infection Research (DZIF), Braunschweig, 38124, Germany
| | - Felix Deschner
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI) and Department of Pharmacy at Saarland University Campus Building E8.1, 66123, Saarbrücken, Germany
- German Centre for Infection Research (DZIF), Braunschweig, 38124, Germany
| | - Gwenaëlle Jézéquel
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI) and Department of Pharmacy at Saarland University Campus Building E8.1, 66123, Saarbrücken, Germany
- German Centre for Infection Research (DZIF), Braunschweig, 38124, Germany
| | - Laìs Pessanha de Carvalho
- German Centre for Infection Research (DZIF), Braunschweig, 38124, Germany
- Institute of Tropical Medicine, Eberhard Karls University Tübingen, Wilhelmstraße 27, 72074, Tübingen, Germany
| | - Noran Abdel-Wadood
- Institute for Medical Microbiology and Hygiene, Saarland University, 66421, Homburg, Germany
- Institute of Anatomy and Cell Biology /, Saarland University, 66421, Homburg, Germany
| | - Linda Pätzold
- Institute for Medical Microbiology and Hygiene, Saarland University, 66421, Homburg, Germany
| | - Steffen Bernecker
- Microbial Drugs, Helmholtz Centre for Infection Research (HZI), Inhoffenstraße 7, 38124, Braunschweig, Germany
| | - Bernd Morgenstern
- Inorganic Solid State Chemistry, Saarland University Campus, 66123, Saarbrücken, Germany
| | - Andreas M Kany
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI) and Department of Pharmacy at Saarland University Campus Building E8.1, 66123, Saarbrücken, Germany
- German Centre for Infection Research (DZIF), Braunschweig, 38124, Germany
| | - Miriam Große
- German Centre for Infection Research (DZIF), Braunschweig, 38124, Germany
- Microbial Drugs, Helmholtz Centre for Infection Research (HZI), Inhoffenstraße 7, 38124, Braunschweig, Germany
| | - Marc Stadler
- German Centre for Infection Research (DZIF), Braunschweig, 38124, Germany
- Microbial Drugs, Helmholtz Centre for Infection Research (HZI), Inhoffenstraße 7, 38124, Braunschweig, Germany
| | - Markus Bischoff
- Institute for Medical Microbiology and Hygiene, Saarland University, 66421, Homburg, Germany
| | - Anna K H Hirsch
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI) and Department of Pharmacy at Saarland University Campus Building E8.1, 66123, Saarbrücken, Germany
- German Centre for Infection Research (DZIF), Braunschweig, 38124, Germany
- Helmholtz International Lab for Anti-Infectives, Saarbrücken, 66123, Germany
| | - Jana Held
- German Centre for Infection Research (DZIF), Braunschweig, 38124, Germany
- Institute of Tropical Medicine, Eberhard Karls University Tübingen, Wilhelmstraße 27, 72074, Tübingen, Germany
- Centre de Recherches Médicales de Lambaréné, Lambaréné, BP 242, BP 242, Gabon
| | - Jennifer Herrmann
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI) and Department of Pharmacy at Saarland University Campus Building E8.1, 66123, Saarbrücken, Germany
- German Centre for Infection Research (DZIF), Braunschweig, 38124, Germany
| | - Rolf Müller
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI) and Department of Pharmacy at Saarland University Campus Building E8.1, 66123, Saarbrücken, Germany
- German Centre for Infection Research (DZIF), Braunschweig, 38124, Germany
- Helmholtz International Lab for Anti-Infectives, Saarbrücken, 66123, Germany
| |
Collapse
|
49
|
Kimishima A, Honsho M, Terai J, Wasuwanich P, Honma S, Matsui H, Hanaki H, Asami Y. Efflux pump inhibitor, phenylalanine-arginine beta-naphthylamide analog potentiates the activity of 5-O-mycaminosyltylonolide for multi-drug resistant Pseudomonas aeruginosa. J Antibiot (Tokyo) 2024; 77:331-333. [PMID: 38467778 DOI: 10.1038/s41429-024-00713-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 02/01/2024] [Accepted: 02/12/2024] [Indexed: 03/13/2024]
Abstract
The emergence and spread of antimicrobial resistance are global threats. Pseudomonas aeruginosa (P. aeruginosa) is responsible for a substantial proportion of this global health issue because of its intrinsic resistance to many antibiotics due to the impermeability of its outer membrane and its multidrug efflux pump systems. Therefore, therapeutic drugs are limited, and the development of new drugs is extremely challenging. As an alternative approach, we focused on a combinational treatment strategy and found that 5-O-mycaminosyltylonolide (OMT) showed potent antibacterial activity against P. aeruginosa in the presence of an efflux pump inhibitor, phenylalanine-arginine beta-naphthylamide (PAβN). In this report, we prepared a PAβN derivative and compared the potentiation activity of OMT by PAβNs against multidrug-resistant P. aeruginosa clinical isolates.
Collapse
Affiliation(s)
- Aoi Kimishima
- Graduate School of Infection Control Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
- Ōmura Satoshi Memorial Institute, Kitasato University, Tokyo, 108-8641, Japan
| | - Masako Honsho
- Graduate School of Infection Control Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
- Ōmura Satoshi Memorial Institute, Kitasato University, Tokyo, 108-8641, Japan
| | - Junsei Terai
- Graduate School of Infection Control Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
| | - Paul Wasuwanich
- University of Florida College of Medicine, Gainesville, FL, USA
| | - Sota Honma
- Graduate School of Infection Control Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
- Ōmura Satoshi Memorial Institute, Kitasato University, Tokyo, 108-8641, Japan
| | - Hidehito Matsui
- Graduate School of Infection Control Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
- Ōmura Satoshi Memorial Institute, Kitasato University, Tokyo, 108-8641, Japan
| | - Hideaki Hanaki
- Graduate School of Infection Control Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
- Ōmura Satoshi Memorial Institute, Kitasato University, Tokyo, 108-8641, Japan
| | - Yukihiro Asami
- Graduate School of Infection Control Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan.
- Ōmura Satoshi Memorial Institute, Kitasato University, Tokyo, 108-8641, Japan.
| |
Collapse
|
50
|
Thompson TP, Gilmore BF. Exploring halophilic environments as a source of new antibiotics. Crit Rev Microbiol 2024; 50:341-370. [PMID: 37079280 DOI: 10.1080/1040841x.2023.2197491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 03/25/2023] [Indexed: 04/21/2023]
Abstract
Microbial natural products from microbes in extreme environments, including haloarchaea, and halophilic bacteria, possess a huge capacity to produce novel antibiotics. Additionally, enhanced isolation techniques and improved tools for genomic mining have expanded the efficiencies in the antibiotic discovery process. This review article provides a detailed overview of known antimicrobial compounds produced by halophiles from all three domains of life. We summarize that while halophilic bacteria, in particular actinomycetes, contribute the vast majority of these compounds the importance of understudied halophiles from other domains of life requires additional consideration. Finally, we conclude by discussing upcoming technologies- enhanced isolation and metagenomic screening, as tools that will be required to overcome the barriers to antimicrobial drug discovery. This review highlights the potential of these microbes from extreme environments, and their importance to the wider scientific community, with the hope of provoking discussion and collaborations within halophile biodiscovery. Importantly, we emphasize the importance of bioprospecting from communities of lesser-studied halophilic and halotolerant microorganisms as sources of novel therapeutically relevant chemical diversity to combat the high rediscovery rates. The complexity of halophiles will necessitate a multitude of scientific disciplines to unravel their potential and therefore this review reflects these research communities.
Collapse
Affiliation(s)
- Thomas P Thompson
- Biofilm Research Group, School of Pharmacy, Queen's University Belfast, Belfast, UK
| | - Brendan F Gilmore
- Biofilm Research Group, School of Pharmacy, Queen's University Belfast, Belfast, UK
| |
Collapse
|