1
|
Chen Y, Wu L, Niu Y, Li H, Sun T, Ye M, Wang L, Wang B, Shi F, Yao Y, Sun W. Waste-to-resource: Utilization of carbon dots derived from proliferating Sargassum aquifolium (Turner) C. Agardh for fluorescent detection of levofloxacin. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2025; 339:126283. [PMID: 40311252 DOI: 10.1016/j.saa.2025.126283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 04/04/2025] [Accepted: 04/22/2025] [Indexed: 05/03/2025]
Abstract
The recurrent proliferation and blooms of Sargassum have emerged as a major environmental challenge in tropical and temperate coastal regions, with profound implications for ecological balance, economic stability, and societal well-being. Levofloxacin (LVF), a fluoroquinolone antibiotic, is physiologically toxic and its residues and accumulation in the aquatic environment have become a serious threat to ecosystems and human health. This work presents a novel approach for the fluorescent detection of LVF, a common antibiotic in environmental waters, using carbon dots derived from the abundant invasive seaweed Sargassum aquifolium (Turner) C. Agardh. The Sargassum carbon dots (SACD) were prepared by a simple one-step hydrothermal method and can reduce the internal filtering effect (IFE) of LVF, thereby facilitating fluorescence detection of high-concentrations of LVF. SEM and TEM were used to characterize the structure, and SACD exhibited a spherical amorphous carbon with a partial lattice structure. The composition of SACD was further analyzed by using FTIR and XPS after synthesis. This method offers a wide detection range (1 μM-100 μM, R2 = 0.998) and low detection limit (0.3 μM) for LVF analysis, exhibiting high recovery rates (103.4 %-113.9 %) in spiked real environmental water samples, demonstrating its potential application performance. The SACD can selectively detect LVF in the presence of the common interfering substances. Therefore, SACD has promising applications in environmental pollutant detection. This study proposes a sustainable approach to utilizing and mitigating the impact of invasive seaweeds, highlighting the potential for converting biological waste into valuable resources for environmental monitoring and protection.
Collapse
Affiliation(s)
- Yuxue Chen
- Key Laboratory of Functional Materials and Photoelectrochemistry of Haikou, Key Laboratory of Laser Technology and Optoelectronic Functional Materials of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, Hainan 571158, China
| | - Luyong Wu
- Key Laboratory of Functional Materials and Photoelectrochemistry of Haikou, Key Laboratory of Laser Technology and Optoelectronic Functional Materials of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, Hainan 571158, China
| | - Yanyan Niu
- Key Laboratory of Functional Materials and Photoelectrochemistry of Haikou, Key Laboratory of Laser Technology and Optoelectronic Functional Materials of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, Hainan 571158, China
| | - Huihui Li
- School of Chemistry and Chemical Engineering, Hainan University, Haikou, Hainan 570228, China.
| | - Tingting Sun
- Key Laboratory of Functional Materials and Photoelectrochemistry of Haikou, Key Laboratory of Laser Technology and Optoelectronic Functional Materials of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, Hainan 571158, China
| | - Miantai Ye
- Key Laboratory of Functional Materials and Photoelectrochemistry of Haikou, Key Laboratory of Laser Technology and Optoelectronic Functional Materials of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, Hainan 571158, China
| | - Lisi Wang
- Key Laboratory of Functional Materials and Photoelectrochemistry of Haikou, Key Laboratory of Laser Technology and Optoelectronic Functional Materials of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, Hainan 571158, China
| | - Baoli Wang
- Key Laboratory of Functional Materials and Photoelectrochemistry of Haikou, Key Laboratory of Laser Technology and Optoelectronic Functional Materials of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, Hainan 571158, China; Haikou Key Laboratory of Marine Contaminants Monitoring Innovation and Application, Haikou Marine Geological Survey Center, China Geological Survey, Haikou, Hainan 571127, China
| | - Fan Shi
- Key Laboratory of Functional Materials and Photoelectrochemistry of Haikou, Key Laboratory of Laser Technology and Optoelectronic Functional Materials of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, Hainan 571158, China
| | - Yucen Yao
- Key Laboratory of Functional Materials and Photoelectrochemistry of Haikou, Key Laboratory of Laser Technology and Optoelectronic Functional Materials of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, Hainan 571158, China
| | - Wei Sun
- Key Laboratory of Functional Materials and Photoelectrochemistry of Haikou, Key Laboratory of Laser Technology and Optoelectronic Functional Materials of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, Hainan 571158, China.
| |
Collapse
|
2
|
Mukherjee S, Chakravarty S, Haldar J. Revitalizing Antibiotics with Macromolecular Engineering: Tackling Gram-Negative Superbugs and Mixed Species Bacterial Biofilm Infections In Vivo. Biomacromolecules 2025; 26:2211-2226. [PMID: 40040432 DOI: 10.1021/acs.biomac.4c01520] [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: 03/06/2025]
Abstract
The escalating prevalence of multidrug-resistant Gram-negative pathogens, coupled with dwindling antibiotic development, has created a critical void in the clinical pipeline. This alarming issue is exacerbated by the formation of biofilms by these superbugs and their frequent coexistence in mixed-species biofilms, conferring extreme antibiotic tolerance. Herein, we present an amphiphilic cationic macromolecule, ACM-AHex, as an innovative antibiotic adjuvant to rejuvenate and repurpose resistant antibiotics, for instance, rifampicin, fusidic acid, erythromycin, and chloramphenicol. ACM-AHex mildly perturbs the bacterial membrane, enhancing antibiotic permeability, hampers efflux machinery, and produces reactive oxygen species, resulting in a remarkable 64-1024-fold potentiation in antibacterial activity. The macromolecule reduces bacterial virulence and macromolecule-drug cocktail significantly eradicate both mono- and multispecies bacterial biofilms, achieving >99.9% bacterial reduction in the murine biofilm infection model. Demonstrating potent biocompatibility across multiple administration routes, ACM-AHex offers a promising strategy to restore obsolete antibiotics and combat recalcitrant Gram-negative biofilm-associated infections, advocating for further clinical evaluation as a next-generation macromolecular antibiotic adjuvant.
Collapse
Affiliation(s)
- Sudip Mukherjee
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru, Karnataka 560064, India
| | - Sayan Chakravarty
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru, Karnataka 560064, India
| | - Jayanta Haldar
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru, Karnataka 560064, India
- School of Advanced Materials, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru, Karnataka 560064, India
| |
Collapse
|
3
|
Kothinti RR, Basavegowda L, Bhoomandla S, Gosu NR, Muppavarapu S, Allaka TR, Raish M. Design, Synthesis, and Biological Evaluation of New Benzo[b]Oxepine-Based 1,2,3-Triazole Derivatives: Molecular Docking, DFT Analysis, In Silico Pharmacokinetics, and Identification of Antimicrobial Pharmacophore Sites. Chem Biodivers 2025:e202500520. [PMID: 40222953 DOI: 10.1002/cbdv.202500520] [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: 02/12/2025] [Revised: 03/26/2025] [Accepted: 04/13/2025] [Indexed: 04/15/2025]
Abstract
A novel series of benzo[b]oxepine-1,2,3-triazole derivatives (8a-8k) was synthesized through a well-defined CuAAC pathway utilizing 5 as a key intermediate. Compounds 8a-8k were synthesized in good-to-excellent yield through a multi-component reaction of 1 with phosphoryl trichloride under the Vilsmeier-Haack reaction, followed by reduction, chlorination, azido-methylation, and triazole, respectively. The obtained products were physicochemically characterized by melting points, FT-IR, 1H nuclear magnetic resonance (NMR), 13C-NMR, and high-resolution mass spectrometry (electron ionization) analysis. Among them, compounds 8d, 8h, and 8j (ZI = 37 ± 0.29, 36 ± 0.31, and 35 ± 0.15 mm, respectively) showed better activity against Escherichia coli than the reference drug MXF (ZI = 33 ± 0.15 mm). In vitro antimicrobial activity results showed that compounds 8d and 8h exhibited potent activity against Pseudomonas aeruginosa (with MIC values of 1.8 and 2.2 µg mL-1, respectively), which was significantly stronger than MXF. Notably, the substituted triazoles 8b and 8h exhibited superior antifungal activity against Candida albicans, with MICs of 5.0 and 3.8 µg mL-1, compared to other compounds in the series. The density functional theory (DFT) study was performed to investigate various electronic properties such as geometry optimization, global reactivity parameters, and frontier molecular orbitals (FMOs). Furthermore, the docking studies were executed on the synthesized derivatives in order to explain the binding interface of compounds with the active sites of the antibacterial protein. In addition, the potent compounds were also subjected to ADME-Tox analysis to evaluate their pharmacokinetic properties, suggesting that all the compounds exhibited comparable results. These potent compounds observed in the current work may lead to promising candidates for future drug development.
Collapse
Affiliation(s)
- Rushendra Reddy Kothinti
- Department of Chemistry, School of Applied Sciences, REVA University, Bengaluru, Karnataka, India
| | - Lakshmi Basavegowda
- Department of Chemistry, School of Applied Sciences, REVA University, Bengaluru, Karnataka, India
| | - Srinu Bhoomandla
- Department of Chemistry, Geethanjali College of Engineering and Technology, Medchal, Telangana, India
- Department of Chemistry, School of Science, GITAM (Deemed to be University) Hyderabad, Hyderabad, Telangana, India
| | - Nageswara Reddy Gosu
- Department of Chemistry, Vel Tech Rangarajan Dr. Sagunthala R & D Institute of Science and Technology, Avadi, Chennai, Tamil Nadu, India
| | - Sudha Muppavarapu
- Department of Chemistry, Nalla Narasimha Reddy Education Society's Group of Institutions, Hyderabad, Telangana, India
| | - Tejeswara Rao Allaka
- Centre for Chemical Sciences and Technology, University College of Engineering Science and Technology Hyderabad, Jawaharlal Nehru Technological University Hyderabad, Hyderabad, Telangana, India
| | - Mohammad Raish
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
4
|
Qu Q, Zhao M, Peng H, Zhu Z, Chen L, Wu H, Liu X, Dong Y, An K, Zheng Y, Zhang Z, Liu Y, Wang H, Dong N, Dong C, Li Y. Glabridin restore the sensitivity of colistin against mcr-1-positive Escherichia coli by polypharmacology mechanism. Microbiol Res 2025; 293:128070. [PMID: 39842376 DOI: 10.1016/j.micres.2025.128070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 01/13/2025] [Accepted: 01/13/2025] [Indexed: 01/24/2025]
Abstract
The clinical effectiveness of colistin against multidrug-resistant Gram-negative pathogen infections has been threatened by the emergence of the plasmid-mediated colistin-resistant gene mcr-1. This development underscores the urgent need for innovative therapeutic strategies that target resistance mechanisms. In this study, we demonstrated that glabridin can restore the sensitivity of colistin to mcr-1-positive Escherichia coli (E. coli) and exhibits a reduced propensity for resistance development. Our investigation into the underlying mechanisms revealed that glabridin may re-sensitize E. coli to colistin by targeting MCR-1 to inhibit its activity, regulating the expression of mcr-1, and restoring the Zeta potential at the cell membrane surface. Furthermore, the combination of glabridin and colistin increased bacterial membrane permeability, decreased membrane fluidity, disrupted transmembrane proton motive force (PMF), reduced the ratios of NAD+/NADH and FAD/FADH2, facilitated the tricarboxylic acid (TCA) cycle, and led to the accumulation of reactive oxygen species (ROS) in E. coli cells, ultimately resulting in bacterial death. In animal models, glabridin significantly enhanced the efficacy of colistin in treating E. coli infections. Our findings suggest that glabridin is a promising polypharmacological antibiotic adjuvant for addressing infections associated with colistin-resistant E. coli.
Collapse
Affiliation(s)
- Qianwei Qu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China; The Laboratory of Molecular Nutrition and Immunity, College of Animal Science and Technol, Northeast Agricultural University, Harbin, China
| | - Mengmeng Zhao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Haixin Peng
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Zhenxin Zhu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Long Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Haojie Wu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Xiaona Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yue Dong
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Kang An
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yadan Zheng
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Zhiyun Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yanyan Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Haoran Wang
- Southern Medical University, Guangzhou, China
| | - Na Dong
- The Laboratory of Molecular Nutrition and Immunity, College of Animal Science and Technol, Northeast Agricultural University, Harbin, China
| | - Chunliu Dong
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.
| | - Yanhua Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.
| |
Collapse
|
5
|
Dhanda G, Singh H, Gupta A, Abdul Mohid S, Biswas K, Mukherjee R, Mukherjee S, Bhunia A, Nair NN, Haldar J. Dual-Functional Antibiotic Adjuvant Displays Potency against Complicated Gram-Negative Bacterial Infections and Exhibits Immunomodulatory Properties. ACS CENTRAL SCIENCE 2025; 11:279-293. [PMID: 40028349 PMCID: PMC11868958 DOI: 10.1021/acscentsci.4c02060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 12/31/2024] [Accepted: 01/03/2025] [Indexed: 03/05/2025]
Abstract
The treatment of Gram-negative bacterial infections is challenged by antibiotic resistance and complicated forms of infection like persistence, multispecies biofilms, intracellular infection, as well as infection-associated hyperinflammation and sepsis. To overcome these challenges, a dual-functional antibiotic adjuvant has been developed as a novel strategy to target complicated forms of bacterial infection and exhibit immunomodulatory properties. The lead adjuvant, D-LBDiphe showed multimodal mechanisms of action like weak outer membrane permeabilization, weak membrane depolarization, and inhibition of efflux machinery, guided primarily by hydrogen bonding and electrostatic interactions, along with weak van der Waals forces. D-LBDiphe potentiated antibiotics up to ∼4100-fold, targeted phenotypic forms of antibiotic tolerance, and revitalized antibiotics against topical and systemic infections of P. aeruginosa in mice. The aromatic moiety in D-LBDiphe was instrumental for interaction with lipopolysaccharide (LPS) micelles, and this interaction was the driving factor in reducing pro-inflammatory cytokines by 61.8-79% in mice challenged with LPS. Such multifarious properties of a weak-membrane perturbing, nonactive and nontoxic adjuvant have been discussed for the first time, supported by detailed mechanistic understanding and elucidation of structure-guided properties. This work expands the scope of antibiotic adjuvants and validates them as a promising approach for treatment of complicated bacterial infections and inflammation.
Collapse
Affiliation(s)
- Geetika Dhanda
- Antimicrobial
Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru 560064, Karnataka, India
| | - Himani Singh
- Antimicrobial
Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru 560064, Karnataka, India
| | - Abhinav Gupta
- Department
of Chemistry, Indian Institute of Technology
Kanpur, Kanpur 208016, India
| | - Sk Abdul Mohid
- Department
of Chemical Sciences, Bose Institute, Kolkata 700091, India
| | - Karishma Biswas
- Department
of Chemical Sciences, Bose Institute, Kolkata 700091, India
| | - Riya Mukherjee
- Antimicrobial
Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru 560064, Karnataka, India
| | - Smriti Mukherjee
- Antimicrobial
Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru 560064, Karnataka, India
| | - Anirban Bhunia
- Department
of Chemical Sciences, Bose Institute, Kolkata 700091, India
| | - Nisanth N. Nair
- Department
of Chemistry, Indian Institute of Technology
Kanpur, Kanpur 208016, India
| | - Jayanta Haldar
- Antimicrobial
Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru 560064, Karnataka, India
- School
of Advanced Materials, Jawaharlal Nehru
Centre for Advanced Scientific Research, Jakkur, Bengaluru 560064, Karnataka, India
| |
Collapse
|
6
|
Xu L, Wang Y, Hu S, Pei Y, Qian C, Xue W, Zhang G, Wu S, Si X, Deng X, Xia J, Wang J. Discovery of Metabolic Reprogramming 2-Quinolones as Effective Antimicrobials for MRSA-Infected Wound Therapy. J Med Chem 2025; 68:3004-3019. [PMID: 39869779 DOI: 10.1021/acs.jmedchem.4c02185] [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: 01/29/2025]
Abstract
To date, the abuse of antibiotics and a gradual decline in novel antibiotic discovery enlarge the threat of drug-resistant bacterial infections, especially methicillin-resistant Staphylococcus aureus (MRSA). Herein, inspired by the unique structures and antibacterial activities of 2-quinolones, a class of novel 2-quinolones with substituted pyridines was synthesized. Notably, compound 11, the derivative with a methylpyridine fragment, showed potent antibacterial and antibiofilm activities, especially for MRSA strains (MIC = 0.02-0.04 μg/mL). A mechanistic study of compound 11 revealed that the increase of intracellular ROS and acceleration of the TCA cycle, which reprogrammed the bacterial metabolism, eventually triggered membrane damage and bacterial death. Most importantly, compound 11, with antibacterial and anti-inflammatory properties, accelerated the reconstruction and healing of MRSA-infected cutaneous wounds by decreasing bacterial loads, attenuating inflammation, and promoting angiogenesis. Overall, these findings provide a novel multifunctional chemotype with broad-spectrum antibacterial activity and highlight a promising strategy for MRSA-infected wound healing.
Collapse
Affiliation(s)
- Lei Xu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Yaling Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- School of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Sangyu Hu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Yuzhu Pei
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Chenliang Qian
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- School of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Wenjie Xue
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Gao Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Song Wu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Xinxin Si
- School of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Xuming Deng
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Jie Xia
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Jianfeng Wang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| |
Collapse
|
7
|
Rahman A, Ono K, Toyomoto T, Hanaoka K, Sawa T. Identification of Fungal Metabolite Gliotoxin as a Potent Inhibitor Against Bacterial O-Acetylserine Sulfhydrylase CysK and CysM. Int J Mol Sci 2025; 26:1106. [PMID: 39940875 PMCID: PMC11818871 DOI: 10.3390/ijms26031106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/21/2025] [Accepted: 01/25/2025] [Indexed: 02/16/2025] Open
Abstract
Cysteine is an essential amino acid for sustaining life, including protein synthesis, and serves as a precursor for antioxidant glutathione. Pathogenic bacteria synthesize cysteine via a two-step enzymatic process using serine as the starting material. The first step is catalyzed by serine acetyltransferase, also known as CysE, and the second by O-acetylserine sulfhydrylase (OASS), referred to as CysK or CysM. This cysteine biosynthetic pathway in bacteria differs significantly from that in mammals, making it an attractive target for the development of novel antibacterial agents. In this study, we aimed to identify OASS inhibitors. To achieve this, a high-throughput screening system was developed to analyze compounds capable of inhibiting CysK/CysM activity. Screening 168,640 compounds from a chemical library revealed that gliotoxin, a fungal metabolite, strongly inhibits both CysK and CysM. Furthermore, gliotoxin significantly suppressed the growth of Salmonella enterica serovar Typhimurium, a Gram-negative bacterium, under cystine-deficient conditions. Gliotoxin possesses a unique disulfide structure classified as epipolythiodioxopiperazine. To date, no studies have reported OASS inhibition by compounds with this structural motif, highlighting its potential for future structural optimization. The screening system developed in this study is expected to accelerate the discovery of functional CysK/CysM inhibitors, providing a foundation for novel antibacterial strategies.
Collapse
Grants
- 21H05262 Ministry of Education, Science, Sports, and Technology (MEXT), Japan
- 21H05267 Ministry of Education, Science, Sports, and Technology (MEXT), Japan
- 22K05443 Ministry of Education, Science, Sports, and Technology (MEXT), Japan
- 23K17979 Ministry of Education, Science, Sports, and Technology (MEXT), Japan
- 23K25210 Ministry of Education, Science, Sports, and Technology (MEXT), Japan
- 17fm0208029h0001 Japan Agency for Medical Research and Development (AMED), Japan
- Not applicable Association for Research on Lactic Acid Bacteria
- Not applicable Daiichi-Sankyo Foundation of Life Science
- the Scholarship Program The Chemo-Sero-Therapeutic Research Institute
- the Program for Leading Graduate Schools, HIGO (Health Life Science: Interdisciplinary and Global Oriented) Program Kumamoto University
Collapse
Affiliation(s)
- Azizur Rahman
- Department of Microbiology, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556, Japan; (A.R.); (K.O.); (T.T.)
| | - Katsuhiko Ono
- Department of Microbiology, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556, Japan; (A.R.); (K.O.); (T.T.)
| | - Touya Toyomoto
- Department of Microbiology, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556, Japan; (A.R.); (K.O.); (T.T.)
| | - Kenjiro Hanaoka
- Graduate School of Pharmaceutical Sciences, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo 105-8512, Japan;
| | - Tomohiro Sawa
- Department of Microbiology, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556, Japan; (A.R.); (K.O.); (T.T.)
| |
Collapse
|
8
|
Scoffone VC, Trespidi G, Barbieri G, Arshad A, Israyilova A, Buroni S. The Evolution of Antimicrobial Resistance in Acinetobacter baumannii and New Strategies to Fight It. Antibiotics (Basel) 2025; 14:85. [PMID: 39858372 PMCID: PMC11760889 DOI: 10.3390/antibiotics14010085] [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: 12/20/2024] [Revised: 01/10/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Acinetobacter baumannii is considered one of the prioritized ESKAPE microorganisms for the research and development of novel treatments by the World Health Organization, especially because of its remarkable persistence and drug resistance. In this review, we describe how this can be acquired by the enzymatic degradation of antibiotics, target site modification, altered membrane permeability, multidrug efflux pumps, and their ability to form biofilms. Also, the evolution of drug resistance in A. baumannii, which is mainly driven by mobile genetic elements, is reported, with particular reference to plasmid-associated resistance, resistance islands, and insertion sequences. Finally, an overview of existing, new, and alternative therapies is provided.
Collapse
Affiliation(s)
- Viola Camilla Scoffone
- Department of Biology and Biotechnology, University of Pavia, 27100 Pavia, Italy; (V.C.S.); (G.T.); (G.B.); (A.A.)
| | - Gabriele Trespidi
- Department of Biology and Biotechnology, University of Pavia, 27100 Pavia, Italy; (V.C.S.); (G.T.); (G.B.); (A.A.)
| | - Giulia Barbieri
- Department of Biology and Biotechnology, University of Pavia, 27100 Pavia, Italy; (V.C.S.); (G.T.); (G.B.); (A.A.)
| | - Arooba Arshad
- Department of Biology and Biotechnology, University of Pavia, 27100 Pavia, Italy; (V.C.S.); (G.T.); (G.B.); (A.A.)
| | - Aygun Israyilova
- Laboratory of Microbiology, Center of Excellence, Baku State University, AZ1148 Baku, Azerbaijan;
- Department of Biomedical Materials by ICESCO, Baku State University, AZ1148 Baku, Azerbaijan
| | - Silvia Buroni
- Department of Biology and Biotechnology, University of Pavia, 27100 Pavia, Italy; (V.C.S.); (G.T.); (G.B.); (A.A.)
| |
Collapse
|
9
|
Pederick JL, Vandborg BC, George A, Bovermann H, Boyd JM, Freundlich JS, Bruning JB. Identification of Cysteine Metabolism Regulator (CymR)-Derived Pentapeptides as Nanomolar Inhibitors of Staphylococcus aureus O-Acetyl-l-serine Sulfhydrylase (CysK). ACS Infect Dis 2025; 11:238-248. [PMID: 39705018 DOI: 10.1021/acsinfecdis.4c00832] [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] [Indexed: 12/21/2024]
Abstract
The pathway of bacterial cysteine biosynthesis is gaining traction for the development of antibiotic adjuvants. Bacterial cysteine biosynthesis is generally facilitated by two enzymes possessing O-acetyl-l-serine sulfhydrylases (OASS), CysK and CysM. In Staphylococcus aureus, there exists a single OASS homologue, SaCysK. Knockout of SaCysK was found to increase sensitivity to oxidative stress, making it a relevant target for inhibitor development. SaCysK also forms two functional complexes via interaction with the preceding enzyme in the pathway serine acetyltransferase (CysE) or the transcriptional regulator of cysteine metabolism (CymR). These interactions occur through insertion of a C-terminal peptide of CysE or CymR into the active site of SaCysK, inhibiting OASS activity, and therefore represent an excellent starting point for developing SaCysK inhibitors. Here, we detail the characterization of CysE and CymR-derived C-terminal peptides as inhibitors of SaCysK. Using a combination of X-ray crystallography, surface plasmon resonance, and enzyme inhibition assays, it was determined that the CymR-derived decapeptide forms extensive interactions with SaCysK and acts as a potent inhibitor (KD = 25 nM; IC50 = 180 nM), making it a promising lead for the development of SaCysK inhibitors. To understand the determinants of this high-affinity interaction, the structure-activity relationships of 16 rationally designed peptides were also investigated. This identified that the C-terminal pentapeptide of CymR facilitates the high-affinity interaction with SaCysK and that subtle structural modification of the pentapeptide is possible without impacting potency. Ultimately, this work identified CymR pentapeptides as a promising scaffold for the development of antibiotic adjuvants targeting SaCysK.
Collapse
Affiliation(s)
- Jordan L Pederick
- Institute for Photonics and Advanced Sensing (IPAS), School of Biological Sciences, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Bethiney C Vandborg
- Institute for Photonics and Advanced Sensing (IPAS), School of Biological Sciences, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Amir George
- Department of Pharmacology, Physiology, and Neuroscience, Rutgers University-New Jersey Medical School, Newark, New Jersey 07101, United States
| | - Hannah Bovermann
- Department of Biochemistry and Microbiology, Rutgers, The State University of New Jersey, New Brunswick, New Jersey 08901, United States
| | - Jeffrey M Boyd
- Department of Biochemistry and Microbiology, Rutgers, The State University of New Jersey, New Brunswick, New Jersey 08901, United States
| | - Joel S Freundlich
- Department of Pharmacology, Physiology, and Neuroscience, Rutgers University-New Jersey Medical School, Newark, New Jersey 07101, United States
| | - John B Bruning
- Institute for Photonics and Advanced Sensing (IPAS), School of Biological Sciences, The University of Adelaide, Adelaide, South Australia 5005, Australia
| |
Collapse
|
10
|
Gil-Gil T, Laborda P, Martínez JL, Hernando-Amado S. Use of adjuvants to improve antibiotic efficacy and reduce the burden of antimicrobial resistance. Expert Rev Anti Infect Ther 2025; 23:31-47. [PMID: 39670956 DOI: 10.1080/14787210.2024.2441891] [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: 06/26/2024] [Revised: 10/28/2024] [Accepted: 12/10/2024] [Indexed: 12/14/2024]
Abstract
INTRODUCTION The increase in antibiotic resistance, together with the absence of novel antibiotics, makes mandatory the introduction of novel strategies to optimize the use of existing antibiotics. Among these strategies, the use of molecules that increase their activity looks promising. AREAS COVERED Different categories of adjuvants have been reviewed. Anti-resistance adjuvants increase the activity of antibiotics by inhibiting antibiotic resistance determinants. Anti-virulence approaches focus on the infection process itself; reducing virulence in combination with an antibiotic can improve therapeutic efficacy. Combination of phages with antibiotics can also be useful, since they present different mechanisms of action and targets. Finally, combining antibiotics with adjuvants in the same molecule may serve to improve antibiotics' efficacy and to overcome potential problems of differential pharmacokinetics/pharmacodynamics. EXPERT OPINION The successful combination of inhibitors of β-lactamases with β-lactams has shown that adjuvants can improve the efficacy of current antibiotics. In this sense, novel anti-resistance adjuvants able to inhibit efflux pumps are still needed, as well as anti-virulence compounds that improve the efficacy of antibiotics by interfering with the infection process. Although adjuvants may present different pharmacodynamics/pharmacokinetics than antibiotics, conjugates containing both compounds can solve this problem. Finally, already approved drugs can be a promising source of antibiotic adjuvants.
Collapse
Affiliation(s)
- Teresa Gil-Gil
- Department of Biology, Emory University, Atlanta, GA, USA
| | - Pablo Laborda
- Department of Clinical Microbiology 9301, Rigshospitalet, Copenhagen, Denmark
| | | | | |
Collapse
|
11
|
Liu Z, Zhou Q, Xue J, Cui M, Xu L, Fang T, Wen Z, Li D, Wang J, Deng X, Zhou Y. Recent discoveries of propyl gallate restore the antibacterial effect of tigecycline against tet(X4)-positive Escherichia coli. Biochem Pharmacol 2025; 231:116638. [PMID: 39571915 DOI: 10.1016/j.bcp.2024.116638] [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: 06/19/2024] [Revised: 11/11/2024] [Accepted: 11/18/2024] [Indexed: 11/28/2024]
Abstract
Propyl gallate (PG), an approved food additive, can be added to different foods and drugs to provide health benefits with minimal danger. However, no clinical application of PG as an antibacterial agent for the treatment of antimicrobial resistance (AMR) has been documented. The aim of this study was to elucidate the effects and mechanisms by which PG inhibits the activity of Tet(X4). Enzyme activity inhibition assay, antimicrobial tests, scanning electron microscopy (SEM) assay, molecular docking and dynamics simulation assays, and animal infection models were used to confirm the synergistic efficacy and mechanism. Here, we found that PG efficiently inhibited Tet(X4) enzyme activity (IC50 = 34.83 μg/mL) while affecting the expression of tet(X4). PG has a synergistic effect with tigecycline (fractional inhibitory concentration index (FICI) < 0.5) against tet(X4)-positive Escherichia coli (E. coli) isolates of animal origin. The survival rates of G. mellonella larvae and the mouse systemic infection model increased by 60 % and 39 %, respectively. The combination of PG and tigecycline showed remarkable treatment benefits in terms of the bacterial load and inflammatory factors in mice. Our results indicate that PG is a valuable adjuvant with tetracyclines and can be considered to address the inevitable infection caused by tet(X4)-positive bacteria, which is a feasible way to extend the lifespan of existing antibiotics.
Collapse
Affiliation(s)
- Zhiying Liu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Qianyu Zhou
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Jinjing Xue
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western China, School of Life Sciences, Ningxia University, Yinchuan, China
| | - Minhe Cui
- Jilin Mushuo Breeding Co., Ltd, Changchun 130052, Jilin, China
| | - Lei Xu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Tianqi Fang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China; College of Food Science and Engineering, Jilin University, Changchun, China
| | - Zhongmei Wen
- Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun 130021, Jilin, China
| | - Dan Li
- Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun 130021, Jilin, China
| | - Jianfeng Wang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China; Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun 130021, Jilin, China
| | - Xuming Deng
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China; Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun 130021, Jilin, China.
| | - Yonglin Zhou
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China; Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western China, School of Life Sciences, Ningxia University, Yinchuan, China.
| |
Collapse
|
12
|
Safwan SM, Mehta D, Arora A, Khatol S, Singh M, Rana K, Gupta SK, Kumar Y, Verma V, Saini V, Bajaj A. Niacin-Cholic Acid-Peptide Conjugate Act as a Potential Antibiotic Adjuvant to Mitigate Polymicrobial Infections Caused by Gram-Negative Pathogens. ACS Infect Dis 2024; 10:4146-4155. [PMID: 39564818 DOI: 10.1021/acsinfecdis.4c00404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2024]
Abstract
Polymicrobial wound infections caused by Gram-negative bacteria and associated inflammation are challenging to manage, as many antibiotics do not work against these infections. Utilizing adjuvants to repurpose the existing antibiotics for mitigating microbial infections presents an alternative therapeutic strategy. We designed and developed a niacin-cholic acid-peptide conjugate (1) to rejuvenate the therapeutic efficacy of macrolide antibiotics against Gram-negative pathogens. We conjugated niacin with anti-inflammatory properties at the carboxyl terminal of the cholic acid and dipeptide (glycine-valine) at the three hydroxyl terminals of cholic acid to obtain the amphiphile 1. Our findings demonstrated that amphiphile 1 serves as a microbial membrane disruptor that facilitates the entry of erythromycin (ERY) in bacterial cells. The combination of amphiphile 1 and ERY is bactericidal and can effectively eliminate monomicrobial and polymicrobial Gram-negative bacterial biofilms. We further demonstrated the antibacterial effectiveness of combining 1 and ERY against monomicrobial and polymicrobial wound infections. Together, these findings indicate that amphiphile 1 revitalizes the remedial efficacy of ERY against Gram-negative bacteria.
Collapse
Affiliation(s)
- Sayed M Safwan
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, Third Milestone, Faridabad-Gurgaon Expressway, NCR Biotech Cluster, Faridabad 121001, Haryana, India
| | - Devashish Mehta
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, Third Milestone, Faridabad-Gurgaon Expressway, NCR Biotech Cluster, Faridabad 121001, Haryana, India
| | - Amit Arora
- Department of Chemistry, Guru Jambheshwar University of Science and Technology, Hisar 125001, Haryana, India
| | - Steffi Khatol
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, Third Milestone, Faridabad-Gurgaon Expressway, NCR Biotech Cluster, Faridabad 121001, Haryana, India
| | - Mohit Singh
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, Third Milestone, Faridabad-Gurgaon Expressway, NCR Biotech Cluster, Faridabad 121001, Haryana, India
| | - Kajal Rana
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, Third Milestone, Faridabad-Gurgaon Expressway, NCR Biotech Cluster, Faridabad 121001, Haryana, India
| | - Sonu K Gupta
- Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Third Milestone, Faridabad-Gurgaon Expressway, Faridabad 121001, Haryana, India
| | - Yashwant Kumar
- Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Third Milestone, Faridabad-Gurgaon Expressway, Faridabad 121001, Haryana, India
| | - Vikas Verma
- Department of Chemistry, Guru Jambheshwar University of Science and Technology, Hisar 125001, Haryana, India
| | - Varsha Saini
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, Third Milestone, Faridabad-Gurgaon Expressway, NCR Biotech Cluster, Faridabad 121001, Haryana, India
| | - Avinash Bajaj
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, Third Milestone, Faridabad-Gurgaon Expressway, NCR Biotech Cluster, Faridabad 121001, Haryana, India
| |
Collapse
|
13
|
Zhang X, Liu P, Zhang R, Zheng W, Qin D, Liu Y, Wang X, Sun T, Gao Y, Li LL. Action Programmed Nanoantibiotics with pH-Induced Collapse and Negative-Charged-Surface-Induced Deformation against Antibiotic-Resistant Bacterial Peritonitis. Adv Healthc Mater 2024; 13:e2401470. [PMID: 38924797 DOI: 10.1002/adhm.202401470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 06/19/2024] [Indexed: 06/28/2024]
Abstract
The incorporation of well-designed antibiotic nanocarriers, along with an antibiotic adjuvant effect, in combination with various antibiotics, offers an opportunity to combat drug-resistant strains. However, precise control over morphology and encapsulated payload release can significantly impact their antibacterial efficacy and synergistic effects when used alongside antibiotics. Here, this study focuses on developing lipopeptide-based nanoantibiotics, which demonstrate an antibiotic adjuvant effect by inducing pH-induced collapse and negative-charged-surface-induced deformation. This enhances the disruption of the bacterial outer membrane and facilitates drug penetration, effectively boosting the antimicrobial activity against drug-resistant strains. The modulation regulations of the lipopeptide nanocarriers with modular design are governed by the authors. The nanoantibiotics, made from lipopeptide and ciprofloxacin (Cip), have a drug loading efficiency of over 80%. The combination with Cip results in a significantly low fractional inhibitory concentration index of 0.375 and a remarkable reduction in the minimum inhibitory concentration of Cip against multidrug-resistant (MDR) Escherichia coli (clinical isolated strains) by up to 32-fold. The survival rate of MDR E. coli peritonitis treated with nanoantibiotics is significantly higher, reaching over 87%, compared to only 25% for Cip and no survival for the control group. Meanwhile, the nanoantibiotic shows no obvious toxicity to major organs.
Collapse
Affiliation(s)
- Xiao Zhang
- School of Pharmacy, Shandong Second Medical University, Weifang, Shandong, 261053, P. R. China
| | - Penghui Liu
- School of Bioscience and Technology, Shandong Second Medical University, Weifang, Shandong, 261053, P. R. China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing, 100190, P. R. China
| | - Ran Zhang
- School of Pharmacy, Shandong Second Medical University, Weifang, Shandong, 261053, P. R. China
| | - Wenhong Zheng
- School of Bioscience and Technology, Shandong Second Medical University, Weifang, Shandong, 261053, P. R. China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing, 100190, P. R. China
| | - Di Qin
- School of Bioscience and Technology, Shandong Second Medical University, Weifang, Shandong, 261053, P. R. China
| | - Yinghang Liu
- School of Pharmacy, Shandong Second Medical University, Weifang, Shandong, 261053, P. R. China
| | - Xin Wang
- School of Bioscience and Technology, Shandong Second Medical University, Weifang, Shandong, 261053, P. R. China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing, 100190, P. R. China
| | - Tongyi Sun
- School of Bioscience and Technology, Shandong Second Medical University, Weifang, Shandong, 261053, P. R. China
| | - Yuanyuan Gao
- School of Pharmacy, Shandong Second Medical University, Weifang, Shandong, 261053, P. R. China
| | - Li-Li Li
- School of Bioscience and Technology, Shandong Second Medical University, Weifang, Shandong, 261053, P. R. China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing, 100190, P. R. China
| |
Collapse
|
14
|
Zhong Y, Guo J, Zhang Z, Zheng Y, Yang M, Su Y. Exogenous NADH promotes the bactericidal effect of aminoglycoside antibiotics against Edwardsiella tarda. Virulence 2024; 15:2367647. [PMID: 38884466 PMCID: PMC11185186 DOI: 10.1080/21505594.2024.2367647] [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: 04/11/2024] [Accepted: 06/09/2024] [Indexed: 06/18/2024] Open
Abstract
The global surge in multidrug-resistant bacteria owing to antibiotic misuse and overuse poses considerable risks to human and animal health. With existing antibiotics losing their effectiveness and the protracted process of developing new antibiotics, urgent alternatives are imperative to curb disease spread. Notably, improving the bactericidal effect of antibiotics by using non-antibiotic substances has emerged as a viable strategy. Although reduced nicotinamide adenine dinucleotide (NADH) may play a crucial role in regulating bacterial resistance, studies examining how the change of metabolic profile and bacterial resistance following by exogenous administration are scarce. Therefore, this study aimed to elucidate the metabolic changes that occur in Edwardsiella tarda (E. tarda), which exhibits resistance to various antibiotics, following the exogenous addition of NADH using metabolomics. The effects of these alterations on the bactericidal activity of neomycin were investigated. NADH enhanced the effectiveness of aminoglycoside antibiotics against E. tarda ATCC15947, achieving bacterial eradication at low doses. Metabolomic analysis revealed that NADH reprogrammed the ATCC15947 metabolic profile by promoting purine metabolism and energy metabolism, yielding increased adenosine triphosphate (ATP) levels. Increased ATP levels played a crucial role in enhancing the bactericidal effects of neomycin. Moreover, exogenous NADH promoted the bactericidal efficacy of tetracyclines and chloramphenicols. NADH in combination with neomycin was effective against other clinically resistant bacteria, including Aeromonas hydrophila, Vibrio parahaemolyticus, methicillin-resistant Staphylococcus aureus, and Listeria monocytogenes. These results may facilitate the development of effective approaches for preventing and managing E. tarda-induced infections and multidrug resistance in aquaculture and clinical settings.
Collapse
Affiliation(s)
- Yilin Zhong
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou, People’s Republic of China
| | - Juan Guo
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou, People’s Republic of China
| | - Ziyi Zhang
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou, People’s Republic of China
| | - Yu Zheng
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou, People’s Republic of China
| | - Manjun Yang
- Xizang Key Laboratory of Veterinary Drug, Xizang Vocational Technical College, Lasa, Xizang, People’s Republic of China
| | - Yubin Su
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou, People’s Republic of China
| |
Collapse
|
15
|
Ma Z, Hou B, Liao A, Tan Y, Tan C, Jiang Y. Light-Activable Inhibitor Overcomes Antimicrobial Resistance and Regulates Antibacterial Activity. J Med Chem 2024; 67:20455-20466. [PMID: 39540544 DOI: 10.1021/acs.jmedchem.4c01923] [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: 11/16/2024]
Abstract
Overuse of antibiotics and the widespread environmental accumulation of antibiotics drive the evolution and spread of antimicrobial resistance, posing a significant global health threat by reducing the effectiveness of available treatments and increasing the risk of untreatable infections. We designed and synthesized PhoPS, a novel photocaged β-lactamase inhibitor, which incorporates the pharmacophore of sulbactam caged with a photoresponsive moiety of o-nitrobiphenyl derivative. Experimental results demonstrate its rapid photoactivation, good stability in solution, and light-activated β-lactamase inhibition in vitro. PhoPS displays synergy with a cephalosporin antibiotic cefoperazone against both susceptible and resistant strains of Escherichia coli and biofilm formation. Additionally, PhoPS treatment demonstrates the potential to suppress the development of resistance in E. coli. These findings suggest that PhoPS offers a promising approach for restoring the efficacy of existing antibiotics and mitigating the emergence of AMR.
Collapse
Affiliation(s)
- Zhuang Ma
- The State Key Laboratory of Chemical Oncogenomics, Institute of Biopharmaceutical and Health Engineering, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, P. R. China
| | - Boxuan Hou
- The State Key Laboratory of Chemical Oncogenomics, Institute of Biopharmaceutical and Health Engineering, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, P. R. China
| | - Anhui Liao
- The State Key Laboratory of Chemical Oncogenomics, Institute of Biopharmaceutical and Health Engineering, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, P. R. China
| | - Ying Tan
- The State Key Laboratory of Chemical Oncogenomics, Institute of Biopharmaceutical and Health Engineering, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, P. R. China
- Open FIESTA, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, P. R. China
| | - Chunyan Tan
- The State Key Laboratory of Chemical Oncogenomics, Institute of Biopharmaceutical and Health Engineering, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, P. R. China
- Open FIESTA, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, P. R. China
| | - Yuyang Jiang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, P. R. China
| |
Collapse
|
16
|
Nuske MR, Zhong J, Huang R, Sarojini V, Chen JLY, Squire CJ, Blaskovich MAT, Leung IKH. Adjuvant strategies to tackle mcr-mediated polymyxin resistance. RSC Med Chem 2024:d4md00654b. [PMID: 39539347 PMCID: PMC11556429 DOI: 10.1039/d4md00654b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024] Open
Abstract
The emergence of the mobile colistin resistance (mcr) gene is a demonstrable threat contributing to the worldwide antibiotic resistance crisis. The gene is encoded on plasmids and can easily spread between different bacterial strains. mcr encodes a phosphoethanolamine (pEtN) transferase, which catalyses the transfer of the pEtN moiety from phosphatidylethanolamine to lipid A, the head group of lipopolysaccharides (LPS). This neutralises the overall negative charge of the LPS and prevents the binding of polymyxins to bacterial membranes. We believe that the development of polymyxin adjuvants could be a promising approach to prolong the use of this important class of last-resort antibiotics. This review discusses recent progress in the identification, design and development of adjuvants to restore polymyxin sensitivity in these resistant bacteria, and focuses on both MCR inhibitors as well as alternative approaches that modulate polymyxin resistance.
Collapse
Affiliation(s)
- Madison R Nuske
- School of Chemistry, The University of Melbourne Parkville Victoria 3010 Australia
- Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne Parkville Victoria 3010 Australia
| | - Junlang Zhong
- School of Chemistry, The University of Melbourne Parkville Victoria 3010 Australia
- Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne Parkville Victoria 3010 Australia
| | - Renjie Huang
- School of Chemical Sciences, The University of Auckland Auckland 1010 New Zealand
| | | | - Jack L Y Chen
- Centre for Biomedical and Chemical Sciences, School of Science, Auckland University of Technology Auckland 1010 New Zealand
- Department of Biotechnology, Chemistry and Pharmaceutical Sciences, Università degli Studi di Siena 53100 Siena Italy
| | - Christopher J Squire
- School of Biological Sciences, The University of Auckland Auckland 1010 New Zealand
| | - Mark A T Blaskovich
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland St. Lucia Queensland 4072 Australia
| | - Ivanhoe K H Leung
- School of Chemistry, The University of Melbourne Parkville Victoria 3010 Australia
- Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne Parkville Victoria 3010 Australia
- School of Chemical Sciences, The University of Auckland Auckland 1010 New Zealand
| |
Collapse
|
17
|
Sun J, Wang X, Gao Y, Li S, Hu Z, Huang Y, Fan B, Wang X, Liu M, Qiao C, Zhang W, Wang Y, Ji X. H 2S scavenger as a broad-spectrum strategy to deplete bacteria-derived H 2S for antibacterial sensitization. Nat Commun 2024; 15:9422. [PMID: 39482291 PMCID: PMC11527999 DOI: 10.1038/s41467-024-53764-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 10/18/2024] [Indexed: 11/03/2024] Open
Abstract
Bacteria-derived H2S plays multifunctional protective roles against antibiotics insult, and the H2S biogenesis pathway is emerging as a viable target for the antibacterial adjuvant design. However, the development of a pan-inhibitor against H2S-synthesizing enzymes is challenging and underdeveloped. Herein, we propose an alternative strategy to downregulate the H2S levels in H2S-producing bacteria, which depletes the bacteria-derived H2S chemically by H2S scavengers without acting on the synthesizing enzymes. After the screening of chemically diversified scaffolds and a structural optimization campaign, a potent and specific H2S scavenger is successfully identified, which displays efficient H2S depletion in several H2S-producing bacteria, potentiates both bactericidal agents and photodynamic therapy, enhances the bacterial clearance of macrophages and polymorphonuclear neutrophils, disrupts the formation of bacterial biofilm and increases the sensitivity of bacterial persister cells to antibiotics. Most importantly, such an H2S scavenger exhibits sensitizing effects with gentamicin in Pseudomonas aeruginosa -infected pneumonia and skin wound female mouse models. In aggregate, our results not only provide an effective strategy to deplete bacteria-derived H2S and establish the H2S biogenesis pathway as a viable target for persisters and drug-resistant bacteria, but also deliver a promising antibacterial adjuvant for potential clinical translation.
Collapse
Affiliation(s)
- Jiekai Sun
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu, China
| | - Xu Wang
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu, China
| | - Ye Gao
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu, China
| | - Shuangyu Li
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu, China
| | - Ziwei Hu
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu, China
| | - Yan Huang
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu, China
| | - Baoqiang Fan
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, Shandong, China
| | - Xia Wang
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu, China
| | - Miao Liu
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu, China
| | - Chunhua Qiao
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu, China
| | - Wei Zhang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, Shandong, China.
| | - Yipeng Wang
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu, China.
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, China.
| | - Xingyue Ji
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
18
|
Zhao X, Zhang Z, Liu L, Wang D, Zhang X, Zhao L, Zhao Y, Jin X, Wang L, Liu X. Guanethidine Enhances the Antibacterial Activity of Rifampicin Against Multidrug-Resistant Bacteria. Microorganisms 2024; 12:2207. [PMID: 39597596 PMCID: PMC11596751 DOI: 10.3390/microorganisms12112207] [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: 09/05/2024] [Revised: 10/24/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024] Open
Abstract
The escalating global threat of antibiotic resistance necessitates innovative strategies, such as the combination of antibiotics with adjuvants. Monotherapy with rifampicin is more likely to induce resistance in pathogens compared to other antibiotics. Herein, we found that the antihypertensive drug guanethidine enhanced the activity of rifampicin against certain clinically resistant Gram-negative bacteria, resulting in a reduction of up to 128-fold in the minimum inhibitory concentration. In infected animal models, this combination has achieved treatment benefits, including increased survival and decreased bacterial burden. The antimicrobial mechanism of guanethidine in synergy with rifampicin involves the disruption of the outer membrane of Gram-negative bacteria, leading to dissipation of the proton motive force. This results in an increase in reactive oxygen species and a reduction in ATP synthesis, severely disturbing energy metabolism and ultimately increasing bacterial mortality. In summary, guanethidine has the potential to become a novel adjuvant for rifampicin, offering a new option for the treatment of clinical Gram-negative bacterial infections.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Lei Wang
- Institute of Animal Husbandry and Veterinary Medicine, Jilin Academy of Agricultural Science, Kemao Street No.186, Gongzhuling 136100, China; (X.Z.); (Z.Z.); (L.L.); (D.W.); (X.Z.); (L.Z.); (Y.Z.); (X.J.)
| | - Xiaoxiao Liu
- Institute of Animal Husbandry and Veterinary Medicine, Jilin Academy of Agricultural Science, Kemao Street No.186, Gongzhuling 136100, China; (X.Z.); (Z.Z.); (L.L.); (D.W.); (X.Z.); (L.Z.); (Y.Z.); (X.J.)
| |
Collapse
|
19
|
Liu Z, Liang X, Zhang Y, Deng W, Wang Y, Lu Z, Liu Q, Wei L. Drug Repurposing: Research Progress of Niclosamide and Its Derivatives on Antibacterial Activity. Infect Drug Resist 2024; 17:4539-4556. [PMID: 39464831 PMCID: PMC11505561 DOI: 10.2147/idr.s490998] [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: 08/10/2024] [Accepted: 10/09/2024] [Indexed: 10/29/2024] Open
Abstract
The development of antibiotic resistance complicates the treatment of infectious diseases and is a global public health threat. However, drug repurposing can address this resistance issue and reduce research and development costs. Niclosamide is a salicylanilide compound approved by the Food and Drug Administration (FDA), and it has been used clinically for treating parasitic infections for many years. Recent studies have shown that niclosamide can inhibit bacterial and fungus activity by affecting the quorum sensing system, biofilm formation, cell membrane potential, and other mechanisms. Here, we discuss recent advances in the antimicrobial applications of niclosamide and its derivatives to provide new perspectives in treating infectious diseases.
Collapse
Affiliation(s)
- Zhihong Liu
- School of Public Health, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu, People’s Republic of China
- Department of Clinical Laboratory, Gansu Provincial Hospital, Lanzhou, Gansu, People’s Republic of China
| | - Xiaofang Liang
- School of Public Health, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu, People’s Republic of China
| | - Yu Zhang
- School of Public Health, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu, People’s Republic of China
| | - Wenbo Deng
- School of Public Health, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu, People’s Republic of China
| | - Yulin Wang
- Ningxia Medical University, Yinchuan, Ningxia, People’s Republic of China
| | - Zhangping Lu
- Department of Clinical Laboratory, Gansu Provincial Hospital, Lanzhou, Gansu, People’s Republic of China
| | - Qianqian Liu
- Department of Clinical Laboratory, Gansu Provincial Hospital, Lanzhou, Gansu, People’s Republic of China
| | - Lianhua Wei
- Department of Clinical Laboratory, Gansu Provincial Hospital, Lanzhou, Gansu, People’s Republic of China
| |
Collapse
|
20
|
Zhao X, Zhang M, Zhang Z, Wang L, Wang Y, Liu L, Wang D, Zhang X, Zhao L, Zhao Y, Jin X, Liu X, Ma H. Guanethidine Restores Tetracycline Sensitivity in Multidrug-Resistant Escherichia coli Carrying tetA Gene. Antibiotics (Basel) 2024; 13:973. [PMID: 39452239 PMCID: PMC11504368 DOI: 10.3390/antibiotics13100973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/30/2024] [Accepted: 10/12/2024] [Indexed: 10/26/2024] Open
Abstract
The worrying issue of antibiotic resistance in pathogenic bacteria is aggravated by the scarcity of novel therapeutic agents. Antibiotic adjuvants offer a promising solution due to their cost-effectiveness and high efficacy in addressing this issue, such as the β-lactamase inhibitor sulbactam (a β-lactam adjuvant) and the dihydrofolate reductase inhibitor trimethoprim (a sulfonamide adjuvant). This study aimed to discover potential adjuvants for tetracyclines from a list of previously approved drugs to restore susceptibility to Escherichia coli carrying the tetA gene. We have screened guanethidine, a compound from the Chinese pharmacopoeia, which effectively potentiates the activity of tetracyclines by reversing resistance in tetA-positive Escherichia coli, enhancing its antibacterial potency, and retarding the development of resistance. Guanethidine functions via the inhibition of the TetA efflux pump, thereby increasing the intracellular concentration of tetracyclines. Our findings suggest that guanethidine holds promise as an antibiotic adjuvant.
Collapse
Affiliation(s)
- Xiaoou Zhao
- College of Animal Science and Technology, Jilin Agricultural University, Xincheng Street No. 2888, Changchun 130118, China; (X.Z.); (M.Z.); (Y.W.)
- Institute of Animal Husbandry and Veterinary Medicine, Jilin Academy of Agricultural Science, Kemao Street No. 186, Gongzhuling 136100, China; (Z.Z.); (L.W.); (L.L.); (D.W.); (X.Z.); (L.Z.); (Y.Z.); (X.J.)
| | - Mengna Zhang
- College of Animal Science and Technology, Jilin Agricultural University, Xincheng Street No. 2888, Changchun 130118, China; (X.Z.); (M.Z.); (Y.W.)
- College of Veterinary Medicine, Northwest A&F University, Xinong Street No. 22, Yangling 712100, China
| | - Zhendu Zhang
- Institute of Animal Husbandry and Veterinary Medicine, Jilin Academy of Agricultural Science, Kemao Street No. 186, Gongzhuling 136100, China; (Z.Z.); (L.W.); (L.L.); (D.W.); (X.Z.); (L.Z.); (Y.Z.); (X.J.)
| | - Lei Wang
- Institute of Animal Husbandry and Veterinary Medicine, Jilin Academy of Agricultural Science, Kemao Street No. 186, Gongzhuling 136100, China; (Z.Z.); (L.W.); (L.L.); (D.W.); (X.Z.); (L.Z.); (Y.Z.); (X.J.)
| | - Yu Wang
- College of Animal Science and Technology, Jilin Agricultural University, Xincheng Street No. 2888, Changchun 130118, China; (X.Z.); (M.Z.); (Y.W.)
- College of Life Sciences, Jilin Normal University, Haifeng Street No. 1301, Siping 136000, China
| | - Lizai Liu
- Institute of Animal Husbandry and Veterinary Medicine, Jilin Academy of Agricultural Science, Kemao Street No. 186, Gongzhuling 136100, China; (Z.Z.); (L.W.); (L.L.); (D.W.); (X.Z.); (L.Z.); (Y.Z.); (X.J.)
| | - Duojia Wang
- Institute of Animal Husbandry and Veterinary Medicine, Jilin Academy of Agricultural Science, Kemao Street No. 186, Gongzhuling 136100, China; (Z.Z.); (L.W.); (L.L.); (D.W.); (X.Z.); (L.Z.); (Y.Z.); (X.J.)
| | - Xin Zhang
- Institute of Animal Husbandry and Veterinary Medicine, Jilin Academy of Agricultural Science, Kemao Street No. 186, Gongzhuling 136100, China; (Z.Z.); (L.W.); (L.L.); (D.W.); (X.Z.); (L.Z.); (Y.Z.); (X.J.)
| | - Luobing Zhao
- Institute of Animal Husbandry and Veterinary Medicine, Jilin Academy of Agricultural Science, Kemao Street No. 186, Gongzhuling 136100, China; (Z.Z.); (L.W.); (L.L.); (D.W.); (X.Z.); (L.Z.); (Y.Z.); (X.J.)
| | - Yunhui Zhao
- Institute of Animal Husbandry and Veterinary Medicine, Jilin Academy of Agricultural Science, Kemao Street No. 186, Gongzhuling 136100, China; (Z.Z.); (L.W.); (L.L.); (D.W.); (X.Z.); (L.Z.); (Y.Z.); (X.J.)
| | - Xiangshu Jin
- Institute of Animal Husbandry and Veterinary Medicine, Jilin Academy of Agricultural Science, Kemao Street No. 186, Gongzhuling 136100, China; (Z.Z.); (L.W.); (L.L.); (D.W.); (X.Z.); (L.Z.); (Y.Z.); (X.J.)
| | - Xiaoxiao Liu
- Institute of Animal Husbandry and Veterinary Medicine, Jilin Academy of Agricultural Science, Kemao Street No. 186, Gongzhuling 136100, China; (Z.Z.); (L.W.); (L.L.); (D.W.); (X.Z.); (L.Z.); (Y.Z.); (X.J.)
| | - Hongxia Ma
- College of Animal Science and Technology, Jilin Agricultural University, Xincheng Street No. 2888, Changchun 130118, China; (X.Z.); (M.Z.); (Y.W.)
- The Engineering Research Center of Bioreactor and Pharmaceutical Development, Ministry of Education, Jilin Agricultural University, Xincheng Street No. 2888, Changchun 130118, China
| |
Collapse
|
21
|
Vladkova TG, Smani Y, Martinov BL, Gospodinova DN. Recent Progress in Terrestrial Biota Derived Antibacterial Agents for Medical Applications. Molecules 2024; 29:4889. [PMID: 39459256 PMCID: PMC11510244 DOI: 10.3390/molecules29204889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 10/07/2024] [Accepted: 10/13/2024] [Indexed: 10/28/2024] Open
Abstract
Conventional antibiotic and multidrug treatments are becoming less and less effective and the discovery of new effective and safe antibacterial agents is becoming a global priority. Returning to a natural antibacterial product is a relatively new current trend. Terrestrial biota is a rich source of biologically active substances whose antibacterial potential has not been fully utilized. The aim of this review is to present the current state-of-the-art terrestrial biota-derived antibacterial agents inspired by natural treatments. It summarizes the most important sources and newly identified or modified antibacterial agents and treatments from the last five years. It focuses on the significance of plant- animal- and bacteria-derived biologically active agents as powerful alternatives to antibiotics, as well as the advantages of utilizing natural antibacterial molecules alone or in combination with antibiotics. The main conclusion is that terrestrial biota-derived antibacterial products and substances open a variety of new ways for modern improved therapeutic strategies. New terrestrial sources of known antibacterial agents and new antibacterial agents from terrestrial biota were discovered during the last 5 years, which are under investigation together with some long-ago known but now experiencing their renaissance for the development of new medical treatments. The use of natural antibacterial peptides as well as combinational therapy by commercial antibiotics and natural products is outlined as the most promising method for treating bacterial infections. In vivo testing and clinical trials are necessary to reach clinical application.
Collapse
Affiliation(s)
- Todorka G. Vladkova
- Department of Polymer Engineering, University of Chemical Technology and Metallurgy, 8 “Kl. Ohridski” Blvd, 1756 Sofia, Bulgaria
| | - Younes Smani
- Andalusian Center of Developmental Biology, CSIC, Junta de Andalusia, University of Pablo de Olavide, 41013 Seville, Spain;
- Department of Molecular Biology and Biochemical Engineering, Andalusian Center of Developmental Biology, CSIC, Junta de Andalusia, University of Pablo de Olavide, 41013 Seville, Spain
| | - Boris L. Martinov
- Department of Biotechnology, University of Chemical Technology and Metallurgy, 8 “Kl. Ohridski” Blvd, 1756 Sofia, Bulgaria;
| | - Dilyana N. Gospodinova
- Faculty of Electrical Engineering, Technical University of Sofia, 8 “Kl. Ohridski” Blvd, 1756 Sofia, Bulgaria;
| |
Collapse
|
22
|
Li D, Chen L, Qiu X. Rapid synthesis of ferulic acid-derived lignin coated silver nanoparticles with low cytotoxicity and high antibacterial activity. Int J Biol Macromol 2024; 277:134471. [PMID: 39102905 DOI: 10.1016/j.ijbiomac.2024.134471] [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: 01/13/2024] [Revised: 07/09/2024] [Accepted: 08/02/2024] [Indexed: 08/07/2024]
Abstract
Antibiotic resistance and the rise of untreatable bacterial infections pose severe threats to human health. Silver nanoparticles (AgNPs) have emerged as a promising antibacterial solution due to their broad-spectrum effectiveness. However, their relatively high cytotoxicity has limited their widespread application. In this study, ferulic acid (FA) was used as a reducing agent, while silver oxide served as a silver precursor to rapidly prepare FA-derived lignin (FAL) coated AgNPs (AgNPs@FAL) with a size ranging from 34.8 to 77.1 nm. Density functional theory (DFT) calculations indicated that the coating of FAL endowed AgNPs@FAL with high stability, preventing the oxidation of AgNPs prior to antibacterial applications. Cell experiments further indicated that AgNPs@FAL exhibited lower cell toxicity (∼80 % viability of normal kidney cells cultured at 25 μg/mL AgNPs@FAL) compared to fully exposed commercially available citrate-modified AgNPs (AgNPs@CA). Antibacterial experiments revealed that the minimum inhibitory concentrations (MIC) of AgNPs@FAL against E. coli and S. aureus were 12.5 μg/mL and 25 μg/mL, respectively, surpassing the antibacterial effect of AgNPs@CA, as well as ampicillin and penicillin. Additionally, AgNPs@FAL was capable of disrupting E. coli and S. aureus biofilm formation. This novel AgNPs@FAL formulation presents a promising antibacterial solution, addressing limitations observed in conventional drugs.
Collapse
Affiliation(s)
- Dan Li
- Guangdong Provincial Key Laboratory of Plant Resources Biorefinery, School of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou 510006, China
| | - Liheng Chen
- Guangdong Provincial Key Laboratory of Plant Resources Biorefinery, School of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou 510006, China; Guangdong Provincial Laboratory of Chemistry and Fine Chemical Engineering Jieyang Center, Jieyang 515200, China.
| | - Xueqing Qiu
- Guangdong Provincial Key Laboratory of Plant Resources Biorefinery, School of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou 510006, China; Guangdong Provincial Laboratory of Chemistry and Fine Chemical Engineering Jieyang Center, Jieyang 515200, China.
| |
Collapse
|
23
|
Li W, Tao Z, Zhou M, Jiang H, Wang L, Ji B, Zhao Y. Antibiotic adjuvants against multidrug-resistant Gram-negative bacteria: important component of future antimicrobial therapy. Microbiol Res 2024; 287:127842. [PMID: 39032266 DOI: 10.1016/j.micres.2024.127842] [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: 04/24/2024] [Revised: 07/13/2024] [Accepted: 07/14/2024] [Indexed: 07/23/2024]
Abstract
The swift emergence and propagation of multidrug-resistant (MDR) bacterial pathogens constitute a tremendous global health crisis. Among these pathogens, the challenge of antibiotic resistance in Gram-negative bacteria is particularly pressing due to their distinctive structure, such as highly impermeable outer membrane, overexpressed efflux pumps, and mutations. Several strategies have been documented to combat MDR Gram-negative bacteria, including the structural modification of existing antibiotics, the development of antimicrobial adjuvants, and research on novel targets that MDR bacteria are sensitive to. Drugs functioning as adjuvants to mitigate resistance to existing antibiotics may play a pivotal role in future antibacterial therapy strategies. In this review, we provide a brief overview of potential antibacterial adjuvants against Gram-negative bacteria and their mechanisms of action, and discuss the application prospects and potential for bacterial resistance to these adjuvants, along with strategies to reduce this risk.
Collapse
Affiliation(s)
- Wenwen Li
- School of Life Science and Bio-Pharmaceutics, Shenyang Pharmaceutical University, Shenyang, Liaoning Province 110016, PR China
| | - Zhen Tao
- School of Life Science and Bio-Pharmaceutics, Shenyang Pharmaceutical University, Shenyang, Liaoning Province 110016, PR China
| | - Motan Zhou
- School of Life Science and Bio-Pharmaceutics, Shenyang Pharmaceutical University, Shenyang, Liaoning Province 110016, PR China
| | - Huilin Jiang
- School of Life Science and Bio-Pharmaceutics, Shenyang Pharmaceutical University, Shenyang, Liaoning Province 110016, PR China
| | - Liudi Wang
- School of Life Science and Bio-Pharmaceutics, Shenyang Pharmaceutical University, Shenyang, Liaoning Province 110016, PR China
| | - Bingjie Ji
- School of Life Science and Bio-Pharmaceutics, Shenyang Pharmaceutical University, Shenyang, Liaoning Province 110016, PR China
| | - Yongshan Zhao
- School of Life Science and Bio-Pharmaceutics, Shenyang Pharmaceutical University, Shenyang, Liaoning Province 110016, PR China.
| |
Collapse
|
24
|
Pederick JL, Vandborg BC, George A, Bovermann H, Boyd JM, Freundlich JS, Bruning JB. Identification of cysteine metabolism regulator (CymR)-derived pentapeptides as nanomolar inhibitors of Staphylococcus aureus O-acetyl-ʟ-serine sulfhydrylase (CysK). BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.19.614015. [PMID: 39345565 PMCID: PMC11429995 DOI: 10.1101/2024.09.19.614015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
The conditionally essential pathway of bacterial cysteine biosynthesis is gaining traction for the development of antibiotic adjuvants. Bacterial cysteine biosynthesis is generally facilitated by two enzymes possessing O-acetyl-ʟ-serine sulfhydrylase (OASS) activity, CysK and CysM. CysK enzymes can also form functional complexes with other proteins that regulate cysteine metabolism. In Staphylococcus aureus there exists a single OASS homologue, herein termed Sa CysK. Knockout of Sa CysK was found to increase sensitivity to oxidative stress, making it a relevant target for inhibitor development. Sa CysK forms two functional complexes via interaction with the preceding enzyme in the pathway serine acetyltransferase (CysE) or the transcriptional regulator of cysteine metabolism (CymR). These interactions occur through the insertion of a C-terminal peptide of CysE or CymR into the active site of Sa CysK, inhibiting OASS activity, and therefore represent an excellent starting point for developing Sa CysK inhibitors. Here we detail the characterization of CysE and CymR-derived C-terminal peptides as inhibitors of Sa CysK. First, interactions between CysE or CymR-derived C-terminal decapeptides and Sa CysK were assessed by X-ray crystallography. While both peptides occupied the active site of Sa CysK, the alternate sidechains of the CymR decapeptide formed more extensive interactions. Surface plasmon resonance binding assays and Sa CysK inhibition assays revealed that the CymR decapeptide bound to Sa CysK with nanomolar affinity (K D = 25 nM) and inhibited Sa CysK activity (IC 50 = 180 nM), making it a promising lead for the development of Sa CysK inhibitors. To understand the determinants of this high affinity interaction the structure-activity relationships of 16 rationally designed peptides were also investigated. This identified that the C-terminal pentapeptide of CymR alone facilitates the high affinity interaction with Sa CysK, and that subtle structural modification of the pentapeptide is possible without impacting potency. Ultimately, this work has identified CymR pentapeptides as a promising scaffold for the development of antibiotic adjuvants targeting Sa CysK. Author summary There is increasing interest in the investigation of non-essential pathways including bacterial cysteine metabolism for developing antibiotic adjuvants. Within this pathway the O-acetyl-ʟ-serine sulfhydrylase (OASS) enzymes CysK and CysM have been a focus. As such, the OASS enzyme of Staphylococcus aureus , Sa CysK, gained our interest. Previous efforts to inhibit CysK enzymes have mimicked the interaction between CysK and the C-terminus of serine acetyltransferase (CysE) which occurs inside the CysK active site and inhibits OASS activity. CysE peptides have only moderate potency, typically binding with micromolar affinity. In S. aureus another complex forms between Sa CysK and a transcriptional regulator CymR, but the ability of CymR peptides to inhibit CysK enzymes has not been investigated. We noticed there is variation between the C-terminus of CysE and CymR, suggesting that CymR peptides make distinct interactions with Sa CysK and may be superior inhibitors. Here we characterized CysE and CymR peptides as Sa CysK inhibitors. We found CymR peptides make more extensive molecular interactions with Sa CysK and bind with higher affinity, being the most potent peptide inhibitors of a CysK enzyme to date. A CymR pentapeptide is the minimal length required for this potency and provides a promising scaffold for developing antibiotic adjuvants targeting Sa CysK.
Collapse
|
25
|
Fararjeh A, Jaradat DMM, Al-Karablieh N, Al-Fawares O, Obeidat AIM, Bashabsheh RHF, Al-Khreshieh RO. Evaluation of synergism effect of human glucose-dependent insulinotropic polypeptide (GIP) on Klebsiella pneumoniae carbapenemases (KPC) producer isolated from clinical samples. Microb Pathog 2024; 194:106823. [PMID: 39059698 DOI: 10.1016/j.micpath.2024.106823] [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: 02/26/2024] [Revised: 06/23/2024] [Accepted: 07/23/2024] [Indexed: 07/28/2024]
Abstract
Antibiotic resistance is increasing among Gram-negative bacteria, prompting the development of new antibiotics as well as alternative treatment approaches. Klebsiella pneumoniae Carbapenemases (KPC) has become a major concern in the treatment of infections, since KPC-producing bacteria are resistant to a number of β -lactam and non β-lactam antibiotics in addition to hydrolyzing carbapenemases. The aim of this study is to examine the synergistic effect of human Glucose-dependent Insulinotropic Polypeptide (GIP) on KPC producer. The K. pneumoniae isolates were identified by using biochemical tests and PCR genotyping. The disc diffusion method was used to assess the antimicrobial susceptibility of each isolate, and the modified Hodge test (MHT) was used to find carbapenemases. Agar well diffusion and minimum inhibitory concentration (MIC) assays were used to validate the synergistic effect of GIP against Klebsiella species. MIC values of chosen antimicrobial compounds demonstrated a considerable synergism impact when combined with human GIP, particularly against KPC strains. The antibacterial activity of the antimicrobial compounds was boosted by 4-16 times due to human GIP, reducing the MIC values. The fractional inhibitory concentration (FIC) ranged from 0.032 to 0.25 for examined antibiotics. Thus, GIP can be considered an antibacterial adjuvant with the potential to supplement the current antibiotic spectrum.
Collapse
Affiliation(s)
- AbdulFattah Fararjeh
- Department of Medical Laboratory Sciences, Faculty of Science, Al-Balqa Applied University, Al-Salt, Jordan.
| | - Da'san M M Jaradat
- Department of Chemistry, Faculty of Science, Al-Balqa Applied University, Al-Salt, Jordan.
| | - Nehaya Al-Karablieh
- Department of Plant Protection, School of Agriculture, The University of Jordan, Amman, Jordan; Hamdi Mango Center for Scientific Research, The University of Jordan, Amman, Jordan
| | - O'la Al-Fawares
- Department of Medical Laboratory Sciences, Faculty of Science, Al-Balqa Applied University, Al-Salt, Jordan
| | - Abeer I M Obeidat
- Department of Chemistry, Faculty of Science, Al-Balqa Applied University, Al-Salt, Jordan
| | - Raghad H F Bashabsheh
- Department of Medical Laboratory Sciences, Faculty of Science, Al-Balqa Applied University, Al-Salt, Jordan; Histopathology department, Jordanian Royal Medical services, Amman, Jordan
| | - Rozan O Al-Khreshieh
- Department of Medical Laboratory Sciences, Faculty of Science, Al-Balqa Applied University, Al-Salt, Jordan
| |
Collapse
|
26
|
Liu M, Cheng JH, Zhao H, Yu C, Wu J. Targeting the outer membrane of gram-negative foodborne pathogens for food safety: compositions, functions, and disruption strategies. Crit Rev Food Sci Nutr 2024:1-14. [PMID: 39213149 DOI: 10.1080/10408398.2024.2397462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Foodborne pathogens are a major threat to both food safety and public health. The current trend toward fresh and less processed foods and the misuse of antibiotics in food production have made controlling these pathogens even more challenging. The outer membrane has been employed as a practical target to combat foodborne Gram-negative pathogens due to its accessibility and importance. In this review, the compositions of the outer membrane are extensively described firstly, to offer a thorough overview of this target. Current strategies for disrupting the outer membrane are also discussed, with emphasized on their mechanism of action. The disruption of the outer membrane structure, whether caused by severe damage of the lipid bilayer or by interference with the biosynthesis pathway, has been demonstrated to represent an effective antimicrobial strategy. Interference with the outer membrane-mediated functions of barrier, efflux and adhesion also contributes to the fight against Gram-negative pathogens. Their potential for control of foodborne pathogens in the production chain are also proposed. However, it is possible that multiple components in the food matrix may act as a protective barrier against microorganisms, and it is often the case that contamination is not caused by a single microorganism. Further investigation is needed to determine the effectiveness and safety of these methods in more complex systems, and it may be advisable to consider a multi-technology combined approach. Additionally, further studies on outer membranes are necessary to discover more promising mechanisms of action.
Collapse
Affiliation(s)
- Mengyuan Liu
- School of Food Science and Engineering, South China University of Technology, Guangzhou, China
- Academy of Contemporary Food Engineering, South China University of Technology, Guangzhou Higher Education Mega Centre, Guangzhou, China
| | - Jun-Hu Cheng
- School of Food Science and Engineering, South China University of Technology, Guangzhou, China
- Academy of Contemporary Food Engineering, South China University of Technology, Guangzhou Higher Education Mega Centre, Guangzhou, China
| | - Haigang Zhao
- School of Food Science and Engineering, South China University of Technology, Guangzhou, China
- ChemPartner PharmaTech Co., Ltd., Jiangmen, China
| | - Chongchong Yu
- Beijing Key Laboratory of Big Data Technology for Food Safety, Beijing Technology and Business University, Beijing, China
| | - Jingzhu Wu
- Beijing Key Laboratory of Big Data Technology for Food Safety, Beijing Technology and Business University, Beijing, China
| |
Collapse
|
27
|
Li H, Zhu X, Zhang X, Dong C. Caspofungin enhances the potency of rifampin against Gram-negative bacteria. Front Microbiol 2024; 15:1447485. [PMID: 39211315 PMCID: PMC11358092 DOI: 10.3389/fmicb.2024.1447485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 08/01/2024] [Indexed: 09/04/2024] Open
Abstract
Introduction Developing antibiotic adjuvants is an effective strategy to combat antimicrobial resistance (AMR). The envelope of Gram-negative bacteria (GNB) is a barrier to prevent the entry of antibiotics, making it an attractive target for novel antibiotic and adjuvant development. Methods and Results In this study, we identified Caspofungin acetate (CAS) as an antibiotic adjuvant against GNB in the repurposing screen of 3,158 FDA-approved drugs. Checkerboard assays suggested that CAS could enhance the antimicrobial activity of rifampin or colistin against various GNB strains in vitro, Moreover, Galleria mellonella larvae infection model also indicated that CAS significantly potentiated the efficacy of rifampin against multidrug-resistant Escherichia coli 72 strain in vivo. Most importantly, resistance development assay showed that CAS was less susceptible to accelerating the resistance development of drug-sensitive strain E. coli MG1655. Functional studies and RNA-seq analysis confirmed that the mechanisms by which CAS enhanced the antimicrobial activities of antibiotics were involved in permeabilizing the bacterial cell envelope, disrupting proton motive force and inhibiting bacterial biofilm formation. Additionally, it has been found that PgaC is the CAS target and enzymatic assay has confirmed the inhibition activity. Discussion Our results illustrate the feasibility of CAS as an antibiotic adjuvant against GNB, which is an alternative strategy of anti-infection.
Collapse
Affiliation(s)
- Haotian Li
- School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education and School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
| | - Xiaojing Zhu
- School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education and School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
| | - Xing Zhang
- School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education and School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
| | - Changjiang Dong
- School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education and School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
| |
Collapse
|
28
|
Berry L, Ramirez D, Domalaon R, Schweizer F. Sulfonamide Bioisosteres of Niclosamide Enhance Antibacterial Activity of Colistin and Bacitracin. ChemMedChem 2024; 19:e202400175. [PMID: 38679656 DOI: 10.1002/cmdc.202400175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/22/2024] [Accepted: 04/23/2024] [Indexed: 05/01/2024]
Abstract
Multicomponent therapy combining antibiotics with enhancer molecules known as adjuvants is an emerging strategy to combat antimicrobial resistance. Niclosamide is a clinically relevant anthelmintic drug with potential to be repurposed for its inherent antibacterial activity against Gram-positive bacteria and its ability to potentiate the antibacterial activity of colistin against susceptible and resistant Gram-negative bacteria. Herein, sulfonamide analogs of niclosamide were prepared and found to enhance colistin activity against Gram-negative bacteria. The ability of niclosamide and the new sulfonamide analogs to synergize with bacitracin against vancomycin-resistant Enterococcus faecium was also discovered.
Collapse
Affiliation(s)
- Liam Berry
- Department of Chemistry, University of Manitoba, Winnipeg, MB, Canada
| | - Danyel Ramirez
- Department of Chemistry, University of Manitoba, Winnipeg, MB, Canada
| | - Ronald Domalaon
- Department of Chemistry, University of Manitoba, Winnipeg, MB, Canada
| | - Frank Schweizer
- Department of Chemistry, University of Manitoba, Winnipeg, MB, Canada
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
29
|
Dhiman S, Ramirez D, Arora R, Arthur G, Schweizer F. Enhancing outer membrane permeability of tetracycline antibiotics in P. aeruginosa using TOB-CIP conjugates. RSC Med Chem 2024:d4md00329b. [PMID: 39131887 PMCID: PMC11305099 DOI: 10.1039/d4md00329b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 06/20/2024] [Indexed: 08/13/2024] Open
Abstract
Pseudomonas aeruginosa is an opportunistic critical 'priority 1' Gram-negative bacterium that poses a severe threat to public healthcare due to rising antibiotic resistance. Particularly, low membrane permeability and overexpression of efflux pumps in P. aeruginosa lead to intrinsic resistance that compromises the antibacterial activity of antibiotics. The broad-spectrum antibiotics class, tetracyclines, are rarely used to treat P. aeruginosa infections. In the present study, we describe a series of tobramycin-ciprofloxacin (TOB-CIP) conjugates in which the carboxylic acid of ciprofloxacin is linked to the aminoglycoside tobramycin using various tethers thereby generating a cationic amphiphile. The emerging amphiphilic conjugates potentiate tetracycline antibiotics including minocycline, doxycycline, tigecycline, and eravacycline against multidrug-resistant P. aeruginosa isolates. The structure-activity relationship investigation indicates that the flexible hydrophobic C12 carbon-chain linker in TOB-CIP conjugate 1a is an optimal potentiator of tetracyclines against tetracycline-resistant and -susceptible strains of P. aeruginosa. Furthermore, conjugate 1a consistently synergized with the 3rd generation tetracycline, eravacycline, in P. aeruginosa PAO1 in the presence of up to 25% fetal bovine serum (FBS).
Collapse
Affiliation(s)
- Shiv Dhiman
- Department of Chemistry, Faculty of Science, University of Manitoba Winnipeg Manitoba R3T 2N2 Canada
| | - Danyel Ramirez
- Department of Chemistry, Faculty of Science, University of Manitoba Winnipeg Manitoba R3T 2N2 Canada
| | - Rajat Arora
- Department of Chemistry, Faculty of Science, University of Manitoba Winnipeg Manitoba R3T 2N2 Canada
| | - Gilbert Arthur
- Department of Biochemistry and Medical Genetics, University of Manitoba Winnipeg Manitoba R3E 0J9 Canada
| | - Frank Schweizer
- Department of Chemistry, Faculty of Science, University of Manitoba Winnipeg Manitoba R3T 2N2 Canada
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba Winnipeg Manitoba R3E 0J9 Canada
| |
Collapse
|
30
|
Lin S, Song Y, Sun Y, Lin W, Yu G, Liao X, Yang Q. Morpholine-modified Ru-based agents with multiple antibacterial mechanisms as metalloantibiotic candidates against Staphylococcus aureus infection. RSC Adv 2024; 14:20130-20144. [PMID: 38915333 PMCID: PMC11194541 DOI: 10.1039/d4ra02667e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 06/06/2024] [Indexed: 06/26/2024] Open
Abstract
Multidrug-resistant bacteria resulting from the abuse and overuse of antibiotics have become a huge crisis in global public health security. Therefore, it is urgently needed to develop new antibacterial drugs with unique mechanisms of action. As a versatile moiety, morpholine has been widely employed to enhance the potency of numerous bioactive molecules. In this study, a series of ruthenium-based antibacterial agents modified with the morpholine moiety were designed and characterized, aiming to obtain a promising metalloantibiotic with a multitarget mechanism. Antibacterial activity screening demonstrated that the most active complex Ru(ii)-3 exhibited the strongest potency against Staphylococcus aureus (S. aureus) with an MIC value of only 0.78 μg mL-1, which is better than most clinically used antibiotics. Notably, Ru(ii)-3 not only possessed excellent bactericidal efficacy, but could also overcome bacterial resistance. Importantly, Ru(ii)-3 very efficiently removed biofilms produced by bacteria, inhibited the secretion of bacterial exotoxins, and enhanced the activity of many existing antibiotics. The results of mechanism studies confirmed that Ru(ii)-3 could destroy the bacterial membrane and induce ROS production in bacteria. Furthermore, animal infection models confirmed that Ru(ii)-3 showed significant anti-infective activity in vivo. Overall, this work demonstrated that a morpholine-modified ruthenium-based agent is a promising antibiotic candidate in tackling the crisis of drug-resistant bacteria.
Collapse
Affiliation(s)
- Shijie Lin
- Department of Pharmacy, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University) Haikou 570311 China
| | - Yun Song
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University Nanchang 330013 China
| | - Yajuan Sun
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University Nanchang 330013 China
| | - Wenjing Lin
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University Nanchang 330013 China
| | - Guangying Yu
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University Nanchang 330013 China
| | - Xiangwen Liao
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University Nanchang 330013 China
| | - Qiang Yang
- Department of Clinical Pharmacy, Hainan Cancer Hospital Haikou 570100 China
| |
Collapse
|
31
|
Tsutamoto S, Iwasaki Y, Shinohara A, Imamiya R, Samukawa K, Kawada-Matsuo M, Komatsuzawa H, Yamada Y, Mandokoro K, Iwao H, Horiguchi Y, Osada-Oka M. Triterpenoid saponin from Panax ginseng increases the sensitivity of methicillin-resistant Staphylococcus aureus to β-lactam and aminoglycoside antibiotics. Microbiol Spectr 2024; 12:e0322723. [PMID: 38647286 PMCID: PMC11237474 DOI: 10.1128/spectrum.03227-23] [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: 09/17/2023] [Accepted: 03/13/2024] [Indexed: 04/25/2024] Open
Abstract
The triterpenoid saponins, ginsenosides, are the major bioactive compound of red ginseng and can exert various physiological activities. In the present study, we examined whether red ginseng extract (RGE) exerts antibacterial activity against methicillin-resistant Staphylococcus aureus (MRSA). RGE had no bactericidal activity, at least in the range of dissolvable concentration. However, RGE reduced 0.03-0.25-fold the minimum inhibitory concentration (MIC) values of β-lactam antibiotics (oxacillin, ampicillin, carbenicillin, and cefazolin) and aminoglycoside antibiotics (kanamycin and gentamicin) against the two laboratory strains of MRSA. Moreover, the fractional inhibitory concentration index indicated the synergistic activity of RGE with each of the antibiotics. RGE also increased the kanamycin sensitivity of 15 MRSA strains isolated from human volunteers and increased the ampicillin sensitivity of five MRSA strains isolated from dairy cows diagnosed with bovine mastitis. In contrast, RGE did not alter the MIC values of fosfomycin, tetracycline, and erythromycin, suggesting that RGE acts selectively. In contrast, Triton X-100, which was reported to reduce the MIC value of β-lactam antibiotics to MRSA by increasing membrane permeability, reduced the MIC values of fosfomycin and tetracycline. These results indicate that RGE increases the bactericidal effect of antibiotics via a mechanism different from that used by Triton X-100. We found that ginsenoside Rg3 (Rg3), a component of RGE, was an essential compound that exhibits synergy activity with antibiotics. Furthermore, the non-natural compound K, which contains a common protopanaxadiol aglycon moiety with Rg3, also showed synergistic activity with antibiotics. Thus, Rg3 and compound K are potentially new antibiotic adjuvants against MRSA.IMPORTANCEMethicillin-resistant Staphylococcus aureus (MRSA) is a multidrug-resistant organism that is prevalent worldwide. Therefore, the research and development of new agents against MRSA are required. We first found that ginsenoside Rg3 (Rg3) in red ginseng, made from the roots of Panax ginseng C. A. Meyer, increased the sensitivity of β-lactam antibiotics and aminoglycoside antibiotics to MRSA. Furthermore, we identified that compound K, an unnatural ginsenoside analog, also increased the sensitivity of antibiotics to MRSA, similar to Rg3. By contrast, neither Rg3 nor compound K increased the sensitivity of fosfomycin, tetracycline, and erythromycin to MRSA, suggesting that these act selectively. In the present study, the natural compound Rg3 and its structural isomer, compound K, are potentially new antibiotic adjuvants against MRSA. Currently, multiple antibiotics are used to treat MRSA, but the use of these adjuvants is expected to enable the treatment of MRSA with a single antibiotic and low concentrations of antibiotics.
Collapse
Affiliation(s)
- Sakura Tsutamoto
- Food Hygiene and Environmental Health, Division of Applied Life Science, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto, Japan
| | - Yuina Iwasaki
- Food Hygiene and Environmental Health, Division of Applied Life Science, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto, Japan
| | - Akari Shinohara
- Food Hygiene and Environmental Health, Division of Applied Life Science, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto, Japan
| | - Risa Imamiya
- Food Hygiene and Environmental Health, Faculty of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto, Japan
| | - Keiichi Samukawa
- Department of Pharmacology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Miki Kawada-Matsuo
- Department of Bacteriology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Hitoshi Komatsuzawa
- Department of Bacteriology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yui Yamada
- Kyoto Prefectural Chutan Livestock Health Hygiene, Fukuchiyama, Japan
| | - Kouki Mandokoro
- Kyoto Prefectural Chutan Livestock Health Hygiene, Fukuchiyama, Japan
| | - Hiroshi Iwao
- Food Hygiene and Environmental Health, Division of Applied Life Science, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto, Japan
| | - Yasuhiko Horiguchi
- Department of Molecular Bacteriology, Research Institute of Microbial Diseases, Osaka University, Suita, Japan
| | - Mayuko Osada-Oka
- Food Hygiene and Environmental Health, Division of Applied Life Science, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto, Japan
| |
Collapse
|
32
|
Islam N, Reid D. Inhaled antibiotics: A promising drug delivery strategies for efficient treatment of lower respiratory tract infections (LRTIs) associated with antibiotic resistant biofilm-dwelling and intracellular bacterial pathogens. Respir Med 2024; 227:107661. [PMID: 38729529 DOI: 10.1016/j.rmed.2024.107661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 05/06/2024] [Accepted: 05/08/2024] [Indexed: 05/12/2024]
Abstract
Antibiotic-resistant bacteria associated with LRTIs are frequently associated with inefficient treatment outcomes. Antibiotic-resistant Streptococcus pneumoniae, Haemophilus influenzae, Pseudomonas aeruginosa, and Staphylococcus aureus, infections are strongly associated with pulmonary exacerbations and require frequent hospital admissions, usually following failed management in the community. These bacteria are difficult to treat as they demonstrate multiple adaptational mechanisms including biofilm formation to resist antibiotic threats. Currently, many patients with the genetic disease cystic fibrosis (CF), non-CF bronchiectasis (NCFB) and chronic obstructive pulmonary disease (COPD) experience exacerbations of their lung disease and require high doses of systemically administered antibiotics to achieve meaningful clinical effects, but even with high systemic doses penetration of antibiotic into the site of infection within the lung is suboptimal. Pulmonary drug delivery technology that reliably deliver antibacterials directly into the infected cells of the lungs and penetrate bacterial biofilms to provide therapeutic doses with a greatly reduced risk of systemic adverse effects. Inhaled liposomal-packaged antibiotic with biofilm-dissolving drugs offer the opportunity for targeted, and highly effective antibacterial therapeutics in the lungs. Although the challenges with development of some inhaled antibiotics and their clinicals trials have been studied; however, only few inhaled products are available on market. This review addresses the current treatment challenges of antibiotic-resistant bacteria in the lung with some clinical outcomes and provides future directions with innovative ideas on new inhaled formulations and delivery technology that promise enhanced killing of antibiotic-resistant biofilm-dwelling bacteria.
Collapse
Affiliation(s)
- Nazrul Islam
- Pharmacy Discipline, School of Clinical Sciences, Queensland University of Technology (QUT), Brisbane, QLD, Australia; Centre for Immunology and Infection Control (CIIC), Queensland University of Technology, Brisbane, Queensland, Australia; Centre for Materials Science, Queensland University of Technology, Brisbane, Queensland, Australia.
| | - David Reid
- Lung Inflammation and Infection, QIMR Berghofer Medical Research Institute, Australia
| |
Collapse
|
33
|
Lu Y, Kang W, Yu Y, Lu H, Wang Y, Xu Z, Zeng J, Qin M, Xu X. A synergistically antimicrobial and antioxidant hyaluronic acid hydrogel for infected wounds. Int J Biol Macromol 2024; 269:131795. [PMID: 38670175 DOI: 10.1016/j.ijbiomac.2024.131795] [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: 08/22/2023] [Revised: 02/05/2024] [Accepted: 04/13/2024] [Indexed: 04/28/2024]
Abstract
Bacterial infections during wound healing impede the healing process and trigger local or systemic inflammatory reactions. Consequently, there is an urgent need to develop a new material with antimicrobial and antioxidant properties to promote infected wound healing. A synergistically antimicrobial and antioxidant hyaluronic acid hydrogel (HMn) is prepared by employing MnO2 nanosheets into 4ARM-PEG5000-SH crosslinked methacrylated hyaluronic acid (HAMA) network. The coordination between sulfhydryl groups of 4ARM-PEG5000-SH and MnO2 nanosheets ensures entrapment of the nanosheets within the hydrogel, while the interaction between 4ARM-PEG5000-SH and HAMA results in facile gelation through thiol-ene click reaction. MnO2 nanosheets exhibit strong photothermal properties and reactive oxygen species (ROS) scavenging abilities, while hyaluronic acid promotes wound healing. When subjected to near-infrared (NIR) irradiation, the HMn achieves a bactericidal rate of 95.24 % for Staphylococcus aureus and nearly 100 % for Escherichia coli. In animal experiments, treatment with the HMn under NIR irradiation results in the best wound healing outcomes. Both in vitro and vivo biocompatible assays demonstrate that the HMn has rarely cell cytotoxicity and tissue damage. The HMn is easy to prepare and has good biocompatibility as well as efficient antibacterial and antioxidant properties, providing a novel method for the treatment of infected wounds.
Collapse
Affiliation(s)
- Yongping Lu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, PR China; Guangyuan Central Hospital, Guangyuan 628000, PR China
| | - Weiqi Kang
- Guangyuan Central Hospital, Guangyuan 628000, PR China
| | - Yue Yu
- Guangyuan Central Hospital, Guangyuan 628000, PR China
| | - Haiying Lu
- Guangyuan Central Hospital, Guangyuan 628000, PR China
| | - Yuemin Wang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, PR China
| | - Zhe Xu
- Guangyuan Central Hospital, Guangyuan 628000, PR China
| | - Jia Zeng
- Guangyuan Central Hospital, Guangyuan 628000, PR China
| | - Meng Qin
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, PR China.
| | - Xinyuan Xu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, PR China.
| |
Collapse
|
34
|
Geng X, Zhang ZD, Li YX, Hao RC, Yang YJ, Liu XW, Li JY. Fingolimod synergizes and reverses K. pneumoniae resistance to colistin. Front Microbiol 2024; 15:1396663. [PMID: 38873155 PMCID: PMC11169662 DOI: 10.3389/fmicb.2024.1396663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 05/13/2024] [Indexed: 06/15/2024] Open
Abstract
Klebsiella pneumoniae (K. pneumoniae) infection and the rapid spread of multi-drug resistant (MDR) bacteria pose a serious threat to global healthcare. Polymyxin E (colistin), a group of cationic antimicrobial polypeptides, is currently one of the last resort treatment options against carbapenem-resistant Gram-negative pathogens. The effectiveness of colistin has been compromised due to its intensive use. This study found that fingolimod (FLD), a natural product derivative, exhibited a significant synergistic bactericidal effect on K. pneumoniae when combined with colistin, both in vitro and in vivo. The checkerboard method was employed to assess the in vitro synergistic effect of FLD with colistin. FLD enhanced the susceptibility of bacteria to colistin and lowered effectively minimum inhibitory concentrations (MIC) when compared to colistin MIC, and the fractional inhibitory concentrations (FIC) value was less than 0.3. The time-kill curve demonstrated that the combination treatment of FLD and colistin had significant bactericidal efficacy. The in vitro concurrent administration of colistin and FLD resulted in heightening membrane permeability, compromising cell integrity, diminishing membrane fluidity, and perturbing membrane homeostasis. They also induced alterations in membrane potential, levels of reactive oxygen species, and adenosine triphosphate synthesis, ultimately culminating in bacterial death. Moreover, the combination of FLD with colistin significantly influenced fatty acid metabolism. In the mouse infection model, the survival rate of mice injected with K. pneumoniae was significantly improved to 67% and pathological damage was significantly relieved with combination treatment of FLD and colistin when compared with colistin treatment. This study highlights the potential of FLD in combining with colistin for treating infections caused by MDR isolates of K. pneumoniae.
Collapse
Affiliation(s)
| | | | | | | | | | - Xi-Wang Liu
- Key Lab of New Animal Drug of Gansu Province, Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou, China
| | - Jian-Yong Li
- Key Lab of New Animal Drug of Gansu Province, Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou, China
| |
Collapse
|
35
|
Dutta S, Eyolfson S, Zhu Y, Gao Y, Wang X. Practical synthesis of isoindolines yields potent colistin potentiators for multidrug-resistant Acinetobacter baumannii. Org Biomol Chem 2024; 22:4057-4061. [PMID: 38716633 DOI: 10.1039/d4ob00463a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2024]
Abstract
An efficient and practical one-pot synthesis of isoindolines from readily available starting materials was achieved under mild conditions by implementing an isoindole umpolung strategy. A variety of isoindolines were prepared with good to excellent yields. Biological screens of these identified compounds demonstrated that they are potent potentiators of colistin for multi-drug resistant Acinetobacter baumannii.
Collapse
Affiliation(s)
- Somnath Dutta
- Department of Chemistry, University of Colorado Boulder, Boulder, 80309, USA.
| | - Samantha Eyolfson
- Department of Chemistry, University of Colorado Boulder, Boulder, 80309, USA.
| | - Yuhang Zhu
- Department of Chemistry, University of Colorado Boulder, Boulder, 80309, USA.
| | - Yuefeng Gao
- Department of Chemistry, University of Colorado Boulder, Boulder, 80309, USA.
| | - Xiang Wang
- Department of Chemistry, University of Colorado Boulder, Boulder, 80309, USA.
| |
Collapse
|
36
|
Wei MZ, Zhu YY, Zu WB, Wang H, Bai LY, Zhou ZS, Zhao YL, Wang ZJ, Luo XD. Structure optimizing of flavonoids against both MRSA and VRE. Eur J Med Chem 2024; 271:116401. [PMID: 38640870 DOI: 10.1016/j.ejmech.2024.116401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 04/02/2024] [Accepted: 04/06/2024] [Indexed: 04/21/2024]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) and vancomycin-resistant Enterococci (VRE) cause more than 100,000 deaths each year, which need efficient and non-resistant antibacterial agents. SAR analysis of 162 flavonoids from the plant in this paper suggested that lipophilic group at C-3 was crucial, and then 63 novel flavonoid derivatives were designed and total synthesized. Among them, the most promising K15 displayed potent bactericidal activity against clinically isolated MRSA and VRE (MICs = 0.25-1.00 μg/mL) with low toxicity and high membrane selectivity. Moreover, mechanism insights revealed that K15 avoided resistance by disrupting biofilm and targeting the membrane, while vancomycin caused 256 times resistance against MRSA, and ampicillin caused 16 times resistance against VRE by the same 20 generations inducing. K15 eliminated residual bacteria in mice skin MRSA-infected model (>99 %) and abdominal VRE-infected model (>92 %), which was superior to vancomycin and ampicillin.
Collapse
Affiliation(s)
- Mei-Zhen Wei
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, People's Republic of China
| | - Yan-Yan Zhu
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, People's Republic of China
| | - Wen-Biao Zu
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, People's Republic of China
| | - Huan Wang
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, People's Republic of China
| | - Li-Yu Bai
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, People's Republic of China
| | - Zhong-Shun Zhou
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, People's Republic of China
| | - Yun-Li Zhao
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, People's Republic of China
| | - Zhao-Jie Wang
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, People's Republic of China
| | - Xiao-Dong Luo
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, People's Republic of China; State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, People's Republic of China.
| |
Collapse
|
37
|
Moussa AY. Endophytes: a uniquely tailored source of potential antibiotic adjuvants. Arch Microbiol 2024; 206:207. [PMID: 38581477 PMCID: PMC10998792 DOI: 10.1007/s00203-024-03891-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: 12/25/2023] [Revised: 02/06/2024] [Accepted: 02/08/2024] [Indexed: 04/08/2024]
Abstract
Multidrug microbial resistance is risking an annual loss of more than 10 million people' lives by 2050. Solutions include the rational use of antibiotics and the use of drugs that reduce resistance or completely obliterate them. Here endophytes come to play due to their high-yield production and inherent nature to produce antimicrobial molecules. Around 40%, 45% and 17% of antibacterial agents were obtained from fungi, actinomycetes, and bacteria, respectively, whose secondary metabolites revealed effectiveness against resistant microbes such as MRSA, MRSE, and Shigella flexneri. Endophyte's role was not confined to bactericidal effect but extended to other mechanisms against MDR microbes, among which was the adjuvant role or the "magic bullets". Scarce focus was given to antibiotic adjuvants, and many laboratories today just screen for the antimicrobial activity without considering combinations with traditional antibiotics, which means real loss of promising resistance combating molecules. While some examples of synthetic adjuvants were introduced in the last decade, the number is still far from covering the disused antibiotics and restoring them back to clinical use. The data compiled in this article demonstrated the significance of quorum sensing as a foreseen mechanism for adjuvants from endophytes secondary metabolites, which call for urgent in-depth studies of their molecular mechanisms. This review, comprehensively and for the first time, sheds light on the significance of endophytes secondary metabolites in solving AMR problem as AB adjuvants.
Collapse
Affiliation(s)
- Ashaimaa Y Moussa
- Department of Pharmacognosy, Faculty of Pharmacy, Ain-Shams University, African Union Organization Street, Abbassia, Cairo, 11566, Egypt.
| |
Collapse
|
38
|
Lovšin Ž, Kotnik T, Klančnik A. Antibiotic's target site affects the potentiation of Lactiplantibacillus plantarum inhibition and inactivation by electroporation. Front Microbiol 2024; 15:1331714. [PMID: 38585700 PMCID: PMC10996065 DOI: 10.3389/fmicb.2024.1331714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 03/12/2024] [Indexed: 04/09/2024] Open
Abstract
Introduction Antibiotic resistance represents a growing global threat, and thus the motivation to develop novel and combined methods of bacterial inactivation is increasing. Electroporation is a technique in which electric pulses of sufficient strength are applied to permeabilize cells, including bacteria. Combining antibacterials with electroporation is a promising strategy to potentiate their bactericidal and bacteriostatic effectiveness. This approach has already proved useful for increasing bacterial inactivation, yet most studies so far have mainly focused on the maximal achievable effects, and less on the underlying mechanisms. We recently demonstrated that in the Gram-negative (G-) bacterium Escherichia coli, electroporation potentiates antibacterials targeting the peptidoglycan wall more than those with intracellular targets. However, in Gram-positive (G+) bacteria, the wall is directly accessible from the outside, and thus the dependence of potentiation on the antibacterial's target may be rather different. Here, we compare the inactivation and growth inhibition of the G+ bacterium Lactiplantibacillus plantarum for two antibiotics with different modes of action: ampicillin (inhibits cell-wall synthesis) and tetracycline (inhibits intracellular protein synthesis). Methods We used antibiotic concentrations ranging from 0 to 30 × MIC (minimum inhibitory concentration that we predetermined for each antibiotic), a single 1-ms electric pulse with an amplitude from 0 to 20 kV/cm, and post-pulse pre-dilution incubation of 24 h or 1 h. Results Electroporation increased the inhibition and inactivation efficiency of both antibiotics, but this was more pronounced for tetracycline, with statistical significance mostly limited to 24-h incubation. In general, both inhibition and inactivation grew stronger with increasing antibiotic concentration and electric field amplitude. Discussion Our results indicate that electroporation potentiates inactivation of G+ bacteria to a larger extent for antibiotics that inhibit intracellular processes and require transport into the cytoplasm, and to a smaller extent for antibiotics that inhibit cell-wall synthesis. This is the inverse of the relation observed in G- bacteria, and can be explained by the difference in the envelope structure: in G- bacteria the outer membrane must be breached for wall-inhibiting antibiotics to access their target, whereas in G+ bacteria the wall is inherently accessible from the outside and permeabilization does not affect this access.
Collapse
Affiliation(s)
- Žana Lovšin
- Faculty of Electrical Engineering, University of Ljubljana, Ljubljana, Slovenia
| | - Tadej Kotnik
- Faculty of Electrical Engineering, University of Ljubljana, Ljubljana, Slovenia
| | - Anja Klančnik
- Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
39
|
Chen Y, Jiang Y, Xue T, Cheng J. Strategies for the eradication of intracellular bacterial pathogens. Biomater Sci 2024; 12:1115-1130. [PMID: 38284808 DOI: 10.1039/d3bm01498c] [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: 01/30/2024]
Abstract
Intracellular pathogens affect a significant portion of world population and cause millions of deaths each year. They can invade host cells and survive inside them and are extremely resistant to immune systems and antibiotics. Current treatments have limitations, and therefore, new effective therapies are needed to combat this ongoing health challenge. Active research efforts have been made to develop many new strategies to eradicate these intracellular pathogens. In this review, we focus on the intracellular bacterial pathogens and first introduce several representative intracellular bacteria and the diseases they cause. We then discuss the challenges in eradicating these bacteria and summarize the current therapeutics for intracellular bacteria. Finally, recent advances in intracellular bacteria eradication are highlighted.
Collapse
Affiliation(s)
- Yingying Chen
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA.
| | - Yunjiang Jiang
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA.
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
- BayRay Innovation Center, Shenzhen Bay Laboratory, Shenzhen, 518071, China
| | - Tianrui Xue
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Jianjun Cheng
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA.
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
- Biomaterials and Drug Delivery Laboratory, School of Engineering, Westlake University, Hangzhou 310024, China
| |
Collapse
|
40
|
Gong Y, Peng Q, Qiao Y, Tian D, Zhang Y, Xiong X, He M, Xu X, Shi B. Hyperbranched Polylysine Exhibits a Collaborative Enhancement of the Antibiotic Capacity to Kill Gram-Negative Pathogens. Antibiotics (Basel) 2024; 13:217. [PMID: 38534651 DOI: 10.3390/antibiotics13030217] [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: 12/20/2023] [Revised: 01/26/2024] [Accepted: 02/21/2024] [Indexed: 03/28/2024] Open
Abstract
In recent years, traditional antibiotic efficacy outcomes have rapidly diminished due to the advent of drug resistance, and the dose limitation value has increased due to the severe side effect of globalized healthcare. Therefore, novel strategies are required to resensitize resistant pathogens to antibiotics existing in the field and prevent the emergence of drug resistance. In this study, cationic hyperbranched polylysine (HBPL-6) was synthesized using the one-pot polymerization method. HBPL-6 exhibited excellent non-cytotoxicity and bio-solubility properties. The present study also showed that HBPL-6 altered the outer membrane (OM) integrity of Escherichia coli O157:H7, Salmonella typhimurium, and Pseudomonas aeruginosa PAO1 by improving their permeability levels. When administered at a safe dosage, HBPL-6 enhanced the accumulation of rifampicin (RIF) and erythromycin (ERY) in bacteria to restore the efficacy of the antibiotics used. Moreover, the combination of HBPL-6 with colistin (COL) reduced the antibiotic dosage, which was helpful in preventing further drug-resistance outcomes. Therefore, this research provides a new strategy for reducing the dosage of drugs used to combat Gram-negative (G-) bacteria through their synergistic effects.
Collapse
Affiliation(s)
- Yuxin Gong
- Feed Research Institute, Chinese Academy of Agricultural Sciences, No. 12 South Zhongguancun Street, Beijing 100081, China
| | - Qing Peng
- Feed Research Institute, Chinese Academy of Agricultural Sciences, No. 12 South Zhongguancun Street, Beijing 100081, China
| | - Yu Qiao
- Feed Research Institute, Chinese Academy of Agricultural Sciences, No. 12 South Zhongguancun Street, Beijing 100081, China
| | - Dandan Tian
- Feed Research Institute, Chinese Academy of Agricultural Sciences, No. 12 South Zhongguancun Street, Beijing 100081, China
| | - Yuwei Zhang
- Institute of Agro-Products Preservation and Processing Technology, Tianjin Academy of Agricultural Sciences, Tianjin 300380, China
| | - Xiaoyan Xiong
- Feed Research Institute, Chinese Academy of Agricultural Sciences, No. 12 South Zhongguancun Street, Beijing 100081, China
| | - Mengxin He
- Feed Research Institute, Chinese Academy of Agricultural Sciences, No. 12 South Zhongguancun Street, Beijing 100081, China
| | - Xiaoqing Xu
- Feed Research Institute, Chinese Academy of Agricultural Sciences, No. 12 South Zhongguancun Street, Beijing 100081, China
| | - Bo Shi
- Feed Research Institute, Chinese Academy of Agricultural Sciences, No. 12 South Zhongguancun Street, Beijing 100081, China
| |
Collapse
|
41
|
Bonacorsi A, Trespidi G, Scoffone VC, Irudal S, Barbieri G, Riabova O, Monakhova N, Makarov V, Buroni S. Characterization of the dispirotripiperazine derivative PDSTP as antibiotic adjuvant and antivirulence compound against Pseudomonas aeruginosa. Front Microbiol 2024; 15:1357708. [PMID: 38435690 PMCID: PMC10904629 DOI: 10.3389/fmicb.2024.1357708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 02/06/2024] [Indexed: 03/05/2024] Open
Abstract
Pseudomonas aeruginosa is a major human pathogen, able to establish difficult-to-treat infections in immunocompromised and people with cystic fibrosis (CF). The high rate of antibiotic treatment failure is due to its notorious drug resistance, often mediated by the formation of persistent biofilms. Alternative strategies, capable of overcoming P. aeruginosa resistance, include antivirulence compounds which impair bacterial pathogenesis without exerting a strong selective pressure, and the use of antimicrobial adjuvants that can resensitize drug-resistant bacteria to specific antibiotics. In this work, the dispirotripiperazine derivative PDSTP, already studied as antiviral, was characterized for its activity against P. aeruginosa adhesion to epithelial cells, its antibiotic adjuvant ability and its biofilm inhibitory potential. PDSTP was effective in impairing the adhesion of P. aeruginosa to various immortalized cell lines. Moreover, the combination of clinically relevant antibiotics with the compound led to a remarkable enhancement of the antibiotic efficacy towards multidrug-resistant CF clinical strains. PDSTP-ceftazidime combination maintained its efficacy in vivo in a Galleria mellonella infection model. Finally, the compound showed a promising biofilm inhibitory activity at low concentrations when tested both in vitro and using an ex vivo pig lung model. Altogether, these results validate PDSTP as a promising compound, combining the ability to decrease P. aeruginosa virulence by impairing its adhesion and biofilm formation, with the capability to increase antibiotic efficacy against antibiotic resistant strains.
Collapse
Affiliation(s)
- Andrea Bonacorsi
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia, Italy
| | - Gabriele Trespidi
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia, Italy
| | - Viola C. Scoffone
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia, Italy
| | - Samuele Irudal
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia, Italy
| | - Giulia Barbieri
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia, Italy
| | - Olga Riabova
- Research Center of Biotechnology RAS, Moscow, Russia
| | | | - Vadim Makarov
- Research Center of Biotechnology RAS, Moscow, Russia
| | - Silvia Buroni
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia, Italy
| |
Collapse
|
42
|
Safwan SM, Kumar N, Mehta D, Singh M, Saini V, Pandey N, Khatol S, Batheja S, Singh J, Walia P, Bajaj A. Xanthone Derivatives Enhance the Therapeutic Potential of Neomycin against Polymicrobial Gram-Negative Bacterial Infections. ACS Infect Dis 2024; 10:527-540. [PMID: 38294409 DOI: 10.1021/acsinfecdis.3c00471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Gram-negative bacterial infections are difficult to manage as many antibiotics are ineffective owing to the presence of impermeable bacterial membranes. Polymicrobial infections pose a serious threat due to the inadequate efficacy of available antibiotics, thereby necessitating the administration of antibiotics at higher doses. Antibiotic adjuvants have emerged as a boon as they can augment the therapeutic potential of available antibiotics. However, the toxicity profile of antibiotic adjuvants is a major hurdle in clinical translation. Here, we report the design, synthesis, and biological activities of xanthone-derived molecules as potential antibiotic adjuvants. Our SAR studies witnessed that the p-dimethylamino pyridine-derivative of xanthone (X8) enhances the efficacy of neomycin (NEO) against Escherichia coli and Pseudomonas aeruginosa and causes a synergistic antimicrobial effect without any toxicity against mammalian cells. Biochemical studies suggest that the combination of X8 and NEO, apart from inhibiting protein synthesis, enhances the membrane permeability by binding to lipopolysaccharide. Notably, the combination of X8 and NEO can disrupt the monomicrobial and polymicrobial biofilms and show promising therapeutic potential against a murine wound infection model. Collectively, our results unveil the combination of X8 and NEO as a suitable adjuvant therapy for the inhibition of the Gram-negative bacterial infections.
Collapse
Affiliation(s)
- Sayed Mohamad Safwan
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, NCR Biotech Science Cluster, Third Milestone, Faridabad-Gurgaon Expressway, Faridabad 121001, Haryana, India
| | - Neeraj Kumar
- Lord Shiva College of Pharmacy, Near Civil Hospital, Sirsa 125055, Haryana, India
| | - Devashish Mehta
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, NCR Biotech Science Cluster, Third Milestone, Faridabad-Gurgaon Expressway, Faridabad 121001, Haryana, India
| | - Mohit Singh
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, NCR Biotech Science Cluster, Third Milestone, Faridabad-Gurgaon Expressway, Faridabad 121001, Haryana, India
| | - Varsha Saini
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, NCR Biotech Science Cluster, Third Milestone, Faridabad-Gurgaon Expressway, Faridabad 121001, Haryana, India
| | - Nishant Pandey
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, NCR Biotech Science Cluster, Third Milestone, Faridabad-Gurgaon Expressway, Faridabad 121001, Haryana, India
| | - Steffi Khatol
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, NCR Biotech Science Cluster, Third Milestone, Faridabad-Gurgaon Expressway, Faridabad 121001, Haryana, India
| | - Shalini Batheja
- Lord Shiva College of Pharmacy, Near Civil Hospital, Sirsa 125055, Haryana, India
| | - Jitender Singh
- Lord Shiva College of Pharmacy, Near Civil Hospital, Sirsa 125055, Haryana, India
| | - Preeti Walia
- Lord Shiva College of Pharmacy, Near Civil Hospital, Sirsa 125055, Haryana, India
| | - Avinash Bajaj
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, NCR Biotech Science Cluster, Third Milestone, Faridabad-Gurgaon Expressway, Faridabad 121001, Haryana, India
| |
Collapse
|
43
|
Yarahmadi A, Afkhami H. The role of microbiomes in gastrointestinal cancers: new insights. Front Oncol 2024; 13:1344328. [PMID: 38361500 PMCID: PMC10867565 DOI: 10.3389/fonc.2023.1344328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 12/20/2023] [Indexed: 02/17/2024] Open
Abstract
Gastrointestinal (GI) cancers constitute more than 33% of new cancer cases worldwide and pose a considerable burden on public health. There exists a growing body of evidence that has systematically recorded an upward trajectory in GI malignancies within the last 5 to 10 years, thus presenting a formidable menace to the health of the human population. The perturbations in GI microbiota may have a noteworthy influence on the advancement of GI cancers; however, the precise mechanisms behind this association are still not comprehensively understood. Some bacteria have been observed to support cancer development, while others seem to provide a safeguard against it. Recent studies have indicated that alterations in the composition and abundance of microbiomes could be associated with the progression of various GI cancers, such as colorectal, gastric, hepatic, and esophageal cancers. Within this comprehensive analysis, we examine the significance of microbiomes, particularly those located in the intestines, in GI cancers. Furthermore, we explore the impact of microbiomes on various treatment modalities for GI cancer, including chemotherapy, immunotherapy, and radiotherapy. Additionally, we delve into the intricate mechanisms through which intestinal microbes influence the efficacy of GI cancer treatments.
Collapse
Affiliation(s)
- Aref Yarahmadi
- Department of Biology, Khorramabad Branch, Islamic Azad University, Khorramabad, Iran
| | - Hamed Afkhami
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Department of Medical Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| |
Collapse
|
44
|
Ding Y, Yang XC, Yu YY, Song SN, Li B, Pang XY, Cai JJ, Zhang CH, Huang S, Xia YM, Gao WW. Construction of Mn-N-C nanoparticles with multienzyme-like properties and photothermal performance for the effective treatment of bacterial infections. Biomater Sci 2024; 12:425-439. [PMID: 38050470 DOI: 10.1039/d3bm01228j] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2023]
Abstract
In this work, we successfully constructed Mn-coordinated nitrogen-carbon nanoparticles (Mn-N-C NPs) exhibiting multienzyme-like activities. In a bacterial infectious microenvironment, the POD-like and OXD-like activities of Mn-N-C NPs could synergistically trigger the generation of ROS (˙OH and O2˙-), causing oxidative damage to the bacterial cell membrane for killing bacteria. Alternatively, in neutral or weak alkaline normal tissues, the excessive O2˙- could be converted into O2 and H2O2via the SOD-like ability of Mn-N-C NPs, and subsequently their CAT-like activity catalyzed excess H2O2 into H2O and O2 for protecting normal cells through the antioxidant defense. Mn-N-C NPs also possessed a good NIR-photothermal performance, which could enhance their POD-like and OXD-like activities. Furthermore, Mn-N-C NPs could facilitate the GSH oxidation process and disrupt the intrinsic balance in the bacterial protection microenvironment with the assistance of H2O2, which is beneficial for rapid bacterial death. Undoubtedly, the Mn-N-C NPs + H2O2 system showed the highest antibacterial activity when irradiated with an 808 nm laser, destroying the bacterial membrane and causing the efflux of proteins. Moreover, the Mn-N-C NPs + H2O2 system was immune to the development of bacterial resistance and could efficiently disrupt the formation of a bacterial biofilm with negligible cytotoxicity and low hemolysis ratio. Finally, Mn-N-C NPs exhibited an excellent antibacterial performance in vivo and could accelerate wound healing without cellular inflammation production. Therefore, due to their significant therapeutic effects, Mn-N-C NPs show great potential in fighting antibiotic-resistant bacteria.
Collapse
Affiliation(s)
- Yong Ding
- State Key Laboratory Base of Eco-chemical Engineering, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China.
| | - Xiao-Chan Yang
- State Key Laboratory Base of Eco-chemical Engineering, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China.
| | - Ya-Ya Yu
- State Key Laboratory Base of Eco-chemical Engineering, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China.
| | - Sheng-Nan Song
- State Key Laboratory Base of Eco-chemical Engineering, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China.
| | - Bo Li
- State Key Laboratory Base of Eco-chemical Engineering, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China.
| | - Xue-Yao Pang
- State Key Laboratory Base of Eco-chemical Engineering, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China.
| | - Jian-Jian Cai
- Township Central Clinic of Masanzi, Binzhou 251907, China
| | | | - Shan Huang
- State Key Laboratory Base of Eco-chemical Engineering, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China.
- The Third Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, China
| | - Ya-Mu Xia
- State Key Laboratory Base of Eco-chemical Engineering, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China.
| | - Wei-Wei Gao
- State Key Laboratory Base of Eco-chemical Engineering, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China.
| |
Collapse
|
45
|
Li T, He X, Tao W, Zhang R, He Q, Gong H, Liu Y, Luo D, Zhang M, Zou C, Zhang SL, He Y. Development of membrane-targeting TPP +-chloramphenicol conjugates to combat methicillin-resistant staphylococcus aureus (MRSA) infections. Eur J Med Chem 2024; 264:115973. [PMID: 38096652 DOI: 10.1016/j.ejmech.2023.115973] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 11/15/2023] [Accepted: 11/16/2023] [Indexed: 12/30/2023]
Abstract
Infections caused by drug-resistant bacteria have become a new challenge in infection treatment, gravely endangering public health. Chloramphenicol (CL) is a well-known antibiotic which has lost its efficacy due to bacterial resistance. To address this issue, herein we report the design, synthesis and biological evaluations of novel triphenylphosphonium chloramphenicol conjugates (TPP+-CL). Study results indicated that compounds 39 and 42 possessed remarkable antibacterial effects against clinically isolated methicillin-resistant Staphylococcus aureus (MRSA) with MIC values ranging from 1 to 2 μg/mL, while CL was inactive to the tested MRSA strains. In addition, these conjugates exhibited rapid bactericidal properties and low toxicity, and did not readily induced bacterial resistance, obviously outperforming the parent drug CL. In a mouse model infected with a clinically isolated MRSA strain, compound 39 at a dose of 20 mg/kg exhibited a comparable or even better in vivo anti-MRSA efficacy than the golden standard drug vancomycin, while no toxicity was observed.
Collapse
Affiliation(s)
- Tao Li
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, No. 55 Daxuecheng South Rd., Shapingba, Chongqing, 401331, PR China
| | - Xiaoli He
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, 266 Fangzheng Ave, Shuitu Technology Development Zone, Beibei, Chongqing, 400714, PR China
| | - Wenlan Tao
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, 266 Fangzheng Ave, Shuitu Technology Development Zone, Beibei, Chongqing, 400714, PR China
| | - Ruixue Zhang
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, No. 55 Daxuecheng South Rd., Shapingba, Chongqing, 401331, PR China
| | - Qiaolin He
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, No. 55 Daxuecheng South Rd., Shapingba, Chongqing, 401331, PR China
| | - Hongzhi Gong
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, No. 55 Daxuecheng South Rd., Shapingba, Chongqing, 401331, PR China
| | - Ye Liu
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, No. 55 Daxuecheng South Rd., Shapingba, Chongqing, 401331, PR China
| | - Dong Luo
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, No. 55 Daxuecheng South Rd., Shapingba, Chongqing, 401331, PR China
| | - Maojie Zhang
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, No. 55 Daxuecheng South Rd., Shapingba, Chongqing, 401331, PR China
| | - Cheng Zou
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, No. 55 Daxuecheng South Rd., Shapingba, Chongqing, 401331, PR China
| | - Shao-Lin Zhang
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, No. 55 Daxuecheng South Rd., Shapingba, Chongqing, 401331, PR China; State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, 518055, PR China.
| | - Yun He
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, No. 55 Daxuecheng South Rd., Shapingba, Chongqing, 401331, PR China; Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, 266 Fangzheng Ave, Shuitu Technology Development Zone, Beibei, Chongqing, 400714, PR China.
| |
Collapse
|
46
|
Huang R, Hu Q, Ko CN, Tang FK, Xuan S, Wong HM, Jin L, Li X, Leung KCF. Nano-based theranostic approaches for infection control: current status and perspectives. MATERIALS CHEMISTRY FRONTIERS 2024; 8:9-40. [DOI: 10.1039/d3qm01048a] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Nano-based theranostic platforms constructed from various nanomaterials possess unique advantages in tackling bacterial and fungal infections while detecting pathogenic cells, making them a potential modality for addressing global healthcare burdens.
Collapse
Affiliation(s)
- Regina Huang
- Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, P. R. China
| | - Qin Hu
- Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, P. R. China
| | - Chung-Nga Ko
- Department of Chemistry, State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong SAR, P. R. China
| | - Fung Kit Tang
- Department of Chemistry, State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong SAR, P. R. China
| | - Shouhu Xuan
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230027, P. R. China
| | - Hai Ming Wong
- Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, P. R. China
| | - Lijian Jin
- Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, P. R. China
| | - Xuan Li
- Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, P. R. China
| | - Ken Cham-Fai Leung
- Department of Chemistry, State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong SAR, P. R. China
| |
Collapse
|
47
|
Vieira Da Cruz A, Jiménez-Castellanos JC, Börnsen C, Van Maele L, Compagne N, Pradel E, Müller RT, Meurillon V, Soulard D, Piveteau C, Biela A, Dumont J, Leroux F, Deprez B, Willand N, Pos KM, Frangakis AS, Hartkoorn RC, Flipo M. Pyridylpiperazine efflux pump inhibitor boosts in vivo antibiotic efficacy against K. pneumoniae. EMBO Mol Med 2024; 16:93-111. [PMID: 38177534 PMCID: PMC10897476 DOI: 10.1038/s44321-023-00007-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 11/09/2023] [Accepted: 11/17/2023] [Indexed: 01/06/2024] Open
Abstract
Antimicrobial resistance is a global problem, rendering conventional treatments less effective and requiring innovative strategies to combat this growing threat. The tripartite AcrAB-TolC efflux pump is the dominant constitutive system by which Enterobacterales like Escherichia coli and Klebsiella pneumoniae extrude antibiotics. Here, we describe the medicinal chemistry development and drug-like properties of BDM91288, a pyridylpiperazine-based AcrB efflux pump inhibitor. In vitro evaluation of BDM91288 confirmed it to potentiate the activity of a panel of antibiotics against K. pneumoniae as well as revert clinically relevant antibiotic resistance mediated by acrAB-tolC overexpression. Using cryo-EM, BDM91288 binding to the transmembrane region of K. pneumoniae AcrB was confirmed, further validating the mechanism of action of this inhibitor. Finally, proof of concept studies demonstrated that oral administration of BDM91288 significantly potentiated the in vivo efficacy of levofloxacin treatment in a murine model of K. pneumoniae lung infection.
Collapse
Affiliation(s)
- Anais Vieira Da Cruz
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177 - Drugs and Molecules for Living Systems, F-59000, Lille, France
| | - Juan-Carlos Jiménez-Castellanos
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, F-59000, Lille, France
| | - Clara Börnsen
- Buchmann Institute for Molecular Life Sciences and Institute for Biophysics, Goethe University Frankfurt, Max-von-Laue-Str. 15, D-60438, Frankfurt am Main, Germany
| | - Laurye Van Maele
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, F-59000, Lille, France
| | - Nina Compagne
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177 - Drugs and Molecules for Living Systems, F-59000, Lille, France
| | - Elizabeth Pradel
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, F-59000, Lille, France
| | - Reinke T Müller
- Institute of Biochemistry, Goethe-University Frankfurt, Max-von-Laue-Str. 9, D-60438, Frankfurt am Main, Germany
| | - Virginie Meurillon
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177 - Drugs and Molecules for Living Systems, F-59000, Lille, France
| | - Daphnée Soulard
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, F-59000, Lille, France
| | - Catherine Piveteau
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177 - Drugs and Molecules for Living Systems, F-59000, Lille, France
| | - Alexandre Biela
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177 - Drugs and Molecules for Living Systems, F-59000, Lille, France
| | - Julie Dumont
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177 - Drugs and Molecules for Living Systems, F-59000, Lille, France
| | - Florence Leroux
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177 - Drugs and Molecules for Living Systems, F-59000, Lille, France
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur Lille, US 41-UAR 2014-PLBS, F-59000, Lille, France
| | - Benoit Deprez
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177 - Drugs and Molecules for Living Systems, F-59000, Lille, France
| | - Nicolas Willand
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177 - Drugs and Molecules for Living Systems, F-59000, Lille, France
| | - Klaas M Pos
- Institute of Biochemistry, Goethe-University Frankfurt, Max-von-Laue-Str. 9, D-60438, Frankfurt am Main, Germany.
| | - Achilleas S Frangakis
- Buchmann Institute for Molecular Life Sciences and Institute for Biophysics, Goethe University Frankfurt, Max-von-Laue-Str. 15, D-60438, Frankfurt am Main, Germany.
| | - Ruben C Hartkoorn
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, F-59000, Lille, France.
| | - Marion Flipo
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177 - Drugs and Molecules for Living Systems, F-59000, Lille, France.
| |
Collapse
|
48
|
Sepordeh S, Jafari AM, Bazzaz S, Abbasi A, Aslani R, Houshmandi S, Rad AH. Postbiotic as Novel Alternative Agent or Adjuvant for the Common Antibiotic Utilized in the Food Industry. Curr Pharm Biotechnol 2024; 25:1245-1263. [PMID: 37702234 DOI: 10.2174/1389201025666230912123849] [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: 03/11/2023] [Revised: 07/11/2023] [Accepted: 07/27/2023] [Indexed: 09/14/2023]
Abstract
BACKGROUND Antibiotic resistance is a serious public health problem as it causes previously manageable diseases to become deadly infections that can cause serious disability or even death. Scientists are creating novel approaches and procedures that are essential for the treatment of infections and limiting the improper use of antibiotics in an effort to counter this rising risk. OBJECTIVES With a focus on the numerous postbiotic metabolites formed from the beneficial gut microorganisms, their potential antimicrobial actions, and recent associated advancements in the food and medical areas, this review presents an overview of the emerging ways to prevent antibiotic resistance. RESULTS Presently, scientific literature confirms that plant-derived antimicrobials, RNA therapy, fecal microbiota transplantation, vaccines, nanoantibiotics, haemofiltration, predatory bacteria, immunotherapeutics, quorum-sensing inhibitors, phage therapies, and probiotics can be considered natural and efficient antibiotic alternative candidates. The investigations on appropriate probiotic strains have led to the characterization of specific metabolic byproducts of probiotics named postbiotics. Based on preclinical and clinical studies, postbiotics with their unique characteristics in terms of clinical (safe origin, without the potential spread of antibiotic resistance genes, unique and multiple antimicrobial action mechanisms), technological (stability and feasibility of largescale production), and economic (low production costs) aspects can be used as a novel alternative agent or adjuvant for the common antibiotics utilized in the production of animal-based foods. CONCLUSION Postbiotic constituents may be a new approach for utilization in the pharmaceutical and food sectors for developing therapeutic treatments. Further metabolomics investigations are required to describe novel postbiotics and clinical trials are also required to define the sufficient dose and optimum administration frequency of postbiotics.
Collapse
Affiliation(s)
- Sama Sepordeh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Sara Bazzaz
- Department of Food Science and Technology, National Nutrition and Food Technology Research Institute, Faculty of Nutrition Science and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amin Abbasi
- Department of Food Science and Technology, National Nutrition and Food Technology Research Institute, Faculty of Nutrition Science and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ramin Aslani
- Food Safety and Hygiene Division, Department of Environmental Health Engineering, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Sousan Houshmandi
- Department of Midwifery, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Aziz Homayouni Rad
- Department of Food Science and Technology, Faculty of Nutrition & Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
49
|
Zhu YY, Wang ZJ, Zhu M, Zhou ZS, Hu BY, Wei MZ, Zhao YL, Dai Z, Luo XD. A dual mechanism with H 2S inhibition and membrane damage of morusin from Morus alba Linn. against MDR-MRSA. Bioorg Med Chem 2024; 97:117544. [PMID: 38071943 DOI: 10.1016/j.bmc.2023.117544] [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: 09/23/2023] [Revised: 11/15/2023] [Accepted: 12/04/2023] [Indexed: 12/30/2023]
Abstract
It's urgent to discover new antibiotics along with the increasing emergence and dissemination of multidrug resistant (MDR) bacterial pathogens. In the present investigation, morusin exhibited rapid bactericidal activity against methicillin-resistant Staphylococcus aureus (MRSA) and vancomycin-resistant Enterococcus (VRE) by targeting the phospholipid of bacterial inner membrane, increasing membrane rigidity and disrupting bacterial homeostasis together with the membrane permeability, which caused fundamental metabolic disorders. Furthermore, morusin can also accumulate ROS, suppress H2S production, and aggravate oxidative damage in bacteria. Importantly, morusin also inhibited the spread of wounds and reduced the bacterial burden in the mouse model of skin infection caused by MRSA. It's a chance to meet the challenge of existing antibiotic resistance and avoid the development of bacterial resistance, given the multiple targets of morusin.
Collapse
Affiliation(s)
- Yan-Yan Zhu
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650500, People's Republic of China
| | - Zhao-Jie Wang
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650500, People's Republic of China
| | - Meng Zhu
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650500, People's Republic of China
| | - Zhong-Shun Zhou
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650500, People's Republic of China
| | - Bin-Yuan Hu
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650500, People's Republic of China
| | - Mei-Zhen Wei
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650500, People's Republic of China
| | - Yun-Li Zhao
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650500, People's Republic of China
| | - Zhi Dai
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650500, People's Republic of China
| | - Xiao-Dong Luo
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650500, People's Republic of China; State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, People's Republic of China.
| |
Collapse
|
50
|
Dey R, Mukherjee S, Mukherjee R, Haldar J. Small molecular adjuvants repurpose antibiotics towards Gram-negative bacterial infections and multispecies bacterial biofilms. Chem Sci 2023; 15:259-270. [PMID: 38143555 PMCID: PMC10739173 DOI: 10.1039/d3sc05124b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 11/12/2023] [Indexed: 12/26/2023] Open
Abstract
Gram-negative bacterial infections pose a significant challenge due to two major resistance elements, including the impermeability of the outer membrane and the overexpression of efflux pumps, which contribute to antibiotic resistance. Additionally, the coexistence of multispecies superbugs in mixed species biofilms further complicates treatment, as these infections are refractory to most antibiotics. To address this issue, combining obsolete antibiotics with non-antibiotic adjuvants that target bacterial membranes has shown promise in combating antibacterial resistance. However, the clinical translation of this cocktail therapy has been hindered by the toxicity associated with these membrane active adjuvants, mainly due to a limited understanding of their structure and mechanism of action. Towards this goal, herein, we have designed a small molecular adjuvant by tuning different structural parameters, such as the balance between hydrophilic and hydrophobic groups, spatial positioning of hydrophobicity and hydrogen bonding interactions, causing moderate membrane perturbation in bacterial cells without any toxicity to mammalian cells. Moderate membrane perturbation not only enhances the internalization of antibiotics, but also increases the intracellular concentration of drugs by hampering the efflux machinery. This revitalises the efficacy of various classes of antibiotics by 32-512 fold, without inducing toxicity. The leading combination not only exhibits potent bactericidal activity against A. baumannii biofilms but also effectively disrupts mature multispecies biofilms composed of A. baumannii and methicillin-resistant Staphylococcus aureus (MRSA), which is typically resistant to most antibiotics. Importantly, the combination therapy demonstrates good biocompatibility and excellent in vivo antibacterial efficacy (>99% reduction) in a skin infection model of A. baumannii. Interestingly, A. baumannii shows reduced susceptibility to develop resistance against the leading combination, underscoring its potential for treating multi-drug resistant infections.
Collapse
Affiliation(s)
- Rajib Dey
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research Jakkur Bengaluru 560064 Karnataka India
| | - Sudip Mukherjee
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research Jakkur Bengaluru 560064 Karnataka India
| | - Riya Mukherjee
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research Jakkur Bengaluru 560064 Karnataka India
| | - Jayanta Haldar
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research Jakkur Bengaluru 560064 Karnataka India
- School of Advanced Materials, Jawaharlal Nehru Centre for Advanced Scientific Research Jakkur Bengaluru 560064 Karnataka India
| |
Collapse
|