1
|
Al Amin M, Hossain MS, Habib MK, Perveen FF, Sabuj SI, Imam H, Islam S, Mahmud S. Pharmacophore-guided computational modeling of quinolone-ATPase conjugate inhibitors targeting DNA GyrB subunit of Staphylococcus aureus. Comput Biol Med 2025; 186:109727. [PMID: 39967191 DOI: 10.1016/j.compbiomed.2025.109727] [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: 11/03/2024] [Revised: 01/17/2025] [Accepted: 01/18/2025] [Indexed: 02/20/2025]
Abstract
The rapid rise of antibiotic-resistant bacterial strains is a significant global health issue, necessitating the development of new and effective antimicrobial agents. This study focuses on designing synthetic de novo models of fluoroquinolone (FQ) descriptors by fusing the quinolone ring-a derivative of FQs-with potential ATPase inhibitors, which is identified through pharmacophore modeling targeting the DNA gyrase B (gyrB) protein of S. aureus. Initially, the pharmacophore model was generated based on the DNA gyrB protein (PDB IDs: 3TTZ, 3U2D, and 3U2K), specifically targeting their co-crystalized ATPase inhibitors to develop a shared feature pharmacophore (SFP) with key features including hydrophobic regions, hydrogen bond acceptors (HBA), hydrogen bond donors (HBD), aromatic moieties (Ar), and halogen bond donors (XBD). The map was further evaluated using the goodness-of-hit (GH) score of 0.2641, indicating the map's strength in capturing potential compounds. The SFP was used for virtual screening against 160,000 compounds from ZINCpharmer and ChEMBL, resulting in 74 hits (48 from ZINCpharmer and 26 from ChEMBL) with similar ATPase features and exhibiting the best-fit scores ranging from 73.50 to 76.80 and RMSD values from 0.1 to 0.5. These identified ligands were fused with the quinolone ring of FQs using genetic algorithms and fragment-based design to create 50 new synthetic models of FQs. Most of these models contain Pyrrole rings, average similarity of more than 55 %, and a Synthetic Accessibility Score (SAScore) ≤ 3.5 for practical syntheses in the lab. These conformers were evaluated through pharmacokinetics and molecular docking, revealing three top compounds-Molecule 13 (-9.1 kcal/mol), Molecule 20 (-9.1 kcal/mol), and Molecule 49 (-9.4 kcal/mol)-which showed greater binding affinity with the DNA gyrase protein (PDB ID: 4PLB) compare to control, Ciprofloxacin (-7.8 kcal/mol). Additionally, 200 ns Molecular Dynamics Simulations (MDS) were conducted using the Schrödinger suite for these three compounds and control, where Molecule 13 showed potential structural stability. To ensure practical feasibility, computational sequence-to-sequence retrosynthesis, and chemical scaffold comparison analysis were employed to design synthetic routes for the most promising compounds, confirming their likelihood of successful synthesis in the lab. The findings of this study contribute to the ongoing efforts to treat antibiotic resistance by providing a framework for designing and evaluating new antimicrobial agents with improved efficacy.
Collapse
Affiliation(s)
- Md Al Amin
- Department of Biotechnology and Genetic Engineering, Mawlana Bhashani Science and Technology University, Santosh, Tangail-1902, Bangladesh
| | - Md Sakhawat Hossain
- Department of Biotechnology and Genetic Engineering, Mawlana Bhashani Science and Technology University, Santosh, Tangail-1902, Bangladesh
| | - Md Kawsar Habib
- Department of Biotechnology and Genetic Engineering, Mawlana Bhashani Science and Technology University, Santosh, Tangail-1902, Bangladesh
| | - Faria Farzana Perveen
- Department of Pharmacy, Mawlana Bhashani Science and Technology University, Santosh, Tangail-1902, Bangladesh
| | - Sahinur Islam Sabuj
- Bachelor of Medicine and Bachelor of Surgery (M.B.B.S.), Universal Medical College, Mohakhali, Dhaka-1215, Bangladesh
| | - Hasan Imam
- Department of Biochemistry and Molecular Biology, Siddheswari College, Moghbazar, Dhaka-1217, Bangladesh
| | - Sirajul Islam
- Department of Biotechnology and Genetic Engineering, Mawlana Bhashani Science and Technology University, Santosh, Tangail-1902, Bangladesh
| | - Shahin Mahmud
- Department of Biotechnology and Genetic Engineering, Mawlana Bhashani Science and Technology University, Santosh, Tangail-1902, Bangladesh.
| |
Collapse
|
2
|
Hasnat S, Rahman S, Alam MB, Suin FM, Yeasmin F, Suha T, Supty NT, Sabila S, Chowdhury A, Shahinuzzaman ADA, Mahbub MM, Islam T, Hoque MN. High throughput screening identifies potential inhibitors targeting trimethoprim resistant DfrA1 protein in Klebsiella pneumoniae and Escherichia coli. Sci Rep 2025; 15:7141. [PMID: 40021806 PMCID: PMC11871338 DOI: 10.1038/s41598-025-91410-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 02/20/2025] [Indexed: 03/03/2025] Open
Abstract
The DfrA1 protein provides trimethoprim resistance in bacteria, especially Klebsiella pneumoniae and Escherichia coli, by modifying dihydrofolate reductase, which reduces the binding efficacy of the antibiotic. This study identified inhibitors of the trimethoprim-resistant DfrA1 protein through high-throughput computational screening and optimization of 3,601 newly synthesized chemical compounds from the ChemDiv database, aiming to discover potential drug candidates targeting DfrA1 in K. pneumoniae and E. coli. Through this approach, we identified six promising DCs, labeled DC1 to DC6, as potential inhibitors of DfrA1. Each DC showed a strong ability to bind effectively to the DfrA1 protein and formed favorable chemical interactions at the binding sites. These interactions were comparable to those of Iclaprim, a well-known antibiotic effective against DfrA1. To confirm our findings, we explored how the promising DCs work at the molecular level, focusing on their thermodynamic properties. Additionally, molecular dynamics simulations confirmed the ability of these six DCs to effectively inhibit the DfrA1 protein. Our results showed that DC4 (an organofluorinated compound) and DC6 (a benzimidazole compound) exhibited potential efficacy against the DfrA1 protein than the control drug, particularly regarding stability, solvent-accessible surface area, solvent exposure, polarity, and binding site interactions, which influence their residence time and efficacy. Overall, findings of this study suggest that DC4 and DC6 have the potential to act as inhibitors against the DfrA1, offering promising prospects for the treatment and management of infections caused by trimethoprim-resistant K. pneumoniae and E. coli in both humans and animals. However, further in vitro validations are necessary.
Collapse
Affiliation(s)
- Soharth Hasnat
- Department of Genetic Engineering and Biotechnology, East West University, Dhaka, 1212, Bangladesh
| | - Soaibur Rahman
- Department of Genetic Engineering and Biotechnology, East West University, Dhaka, 1212, Bangladesh
| | - Meherun Binta Alam
- Department of Genetic Engineering and Biotechnology, East West University, Dhaka, 1212, Bangladesh
| | - Farha Mohi Suin
- Department of Genetic Engineering and Biotechnology, East West University, Dhaka, 1212, Bangladesh
| | - Farzana Yeasmin
- Institute of Biotechnology and Genetic Engineering, Gazipur Agricultural University (GAU), Gazipur, 1706, Bangladesh
| | - Tanjila Suha
- Department of Genetic Engineering and Biotechnology, East West University, Dhaka, 1212, Bangladesh
| | - Nahuna Tanjin Supty
- Department of Genetic Engineering and Biotechnology, East West University, Dhaka, 1212, Bangladesh
| | - Sal Sabila
- Department of Genetic Engineering and Biotechnology, East West University, Dhaka, 1212, Bangladesh
| | - Animesh Chowdhury
- Department of Genetic Engineering and Biotechnology, East West University, Dhaka, 1212, Bangladesh
| | - A D A Shahinuzzaman
- Pharmaceutical Sciences Research Division, Bangladesh Council of Scientific and Industrial Research (BCSIR), Dhaka, 1205, Bangladesh
| | - M Murshida Mahbub
- Department of Genetic Engineering and Biotechnology, East West University, Dhaka, 1212, Bangladesh.
| | - Tofazzal Islam
- Institute of Biotechnology and Genetic Engineering, Gazipur Agricultural University (GAU), Gazipur, 1706, Bangladesh.
| | - M Nazmul Hoque
- Molecular Biology and Bioinformatics Laboratory, Department of Gynecology, Obstetrics and Reproductive Health, Gazipur Agricultural University (GAU), Gazipur, 1706, Bangladesh.
| |
Collapse
|
3
|
Pflégr V, Konečná K, Stolaříková J, Ősterreicher J, Janďourek O, Krátký M. Enhancing the antimycobacterial efficacy of pyridine-4-carbohydrazide: linkage to additional antimicrobial agents via oxocarboxylic acids. RSC Med Chem 2025; 16:767-778. [PMID: 39568598 PMCID: PMC11575622 DOI: 10.1039/d4md00663a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 10/13/2024] [Indexed: 11/22/2024] Open
Abstract
This study evaluates the antimycobacterial potential of novel "mutual" bioactive amides, combining pyridine-4-carbohydrazide (isoniazid, INH) with various antimicrobial agents (sulphonamides, 4-aminosalicylic acid, thiosemicarbazide, diphenyl (thio)ethers) via oxocarboxylic acids. The aim was to enhance activity against both drug-susceptible and multidrug-resistant (MDR) Mycobacterium tuberculosis and non-tuberculous strains, while overcoming drug resistance through dual-action mechanisms. Many derivatives exhibited potent antimycobacterial activity, with minimum inhibitory concentrations (MICs) as low as ≤0.25 μM, outperforming INH, especially diphenyl (thio)ethers and biphenyl analogues. Additionally, the compounds were effective against M. kansasii (MICs ≤1 μM) and inhibited MDR strains at higher concentrations (≥8 μM). The cytotoxicity assay indicated a favourable safety profile, with no significant haemolysis at 125 μM, and some compounds were even protective. Selectivity for mycobacteria was confirmed by low inhibition of Gram-positive bacteria and inactivity against Gram-negative bacteria or fungi, highlighting the potential for further development as antimycobacterial agents.
Collapse
Affiliation(s)
- Václav Pflégr
- Department of Organic and Bioorganic Chemistry, Faculty of Pharmacy in Hradec Králové, Charles University Akademika Heyrovského 1203 500 03 Hradec Králové Czech Republic +420 495067166 +420 495067302
| | - Klára Konečná
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Králové, Charles University Akademika Heyrovského 1203 500 03 Hradec Králové Czech Republic
| | - Jiřina Stolaříková
- Laboratory for Mycobacterial Diagnostics and Tuberculosis, Regional Institute of Public Health in Ostrava Partyzánské náměstí 7 Ostrava Czech Republic
| | - Jan Ősterreicher
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Králové, Charles University Akademika Heyrovského 1203 500 03 Hradec Králové Czech Republic
| | - Ondřej Janďourek
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Králové, Charles University Akademika Heyrovského 1203 500 03 Hradec Králové Czech Republic
| | - Martin Krátký
- Department of Organic and Bioorganic Chemistry, Faculty of Pharmacy in Hradec Králové, Charles University Akademika Heyrovského 1203 500 03 Hradec Králové Czech Republic +420 495067166 +420 495067302
| |
Collapse
|
4
|
Sharma V, Das R, Mehta DK, Sharma D, Aman S, Khan MU. Quinolone scaffolds as potential drug candidates against infectious microbes: a review. Mol Divers 2025; 29:711-737. [PMID: 38683488 DOI: 10.1007/s11030-024-10862-4] [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: 02/16/2024] [Accepted: 03/26/2024] [Indexed: 05/01/2024]
Abstract
Prevalence of microbial infections and new rising pathogens are signified as causative agent for variety of serious and lethal health crisis in past years. Despite medical advances, bacterial and fungal infections continue to be a rising problem in the health care system. As more bacteria develop resistance to antibiotics used in therapy, and as more invasive microbial species develop resistance to conventional antimicrobial drugs. Relevant published publications from the last two decades, up to 2024, were systematically retrieved from the MEDLINE/PubMed, SCOPUS, EMBASE, and WOS databases using keywords such as quinolones, anti-infective, antibacterial, antimicrobial resistance and patents on quinolone derivatives. With an approach of considerable interest towards novel heterocyclic derivatives as novel anti-infective agents, researchers have explored these as essential tools in vistas of drug design and development. Among heterocycles, quinolones have been regarded extremely essential for the development of novel derivatives, even able to tackle the associated resistance issues. The quinolone scaffold with its bicyclic structure and specific functional groups such as the carbonyl and acidic groups, is indeed considered a valuable functionalities for further lead generation and optimization in drug discovery. Besides, the substitution at N-1, C-3 and C-7 positions also subjected to be having a significant role in anti-infective potential. In this article, we intend to highlight recent quinolone derivatives based on the SAR approach and anti-infective potential such as antibacterial, antifungal, antimalarial, antitubercular, antitrypanosomal and antiviral activities. Moreover, some recent patents granted on quinolone-containing derivatives as anti-infective agents have also been highlighted in tabular form. Due consideration of this, future research in this scaffold is expected to be useful for aspiring scientists to get pharmacologically significant leads.
Collapse
Affiliation(s)
- Vishal Sharma
- Department of Pharmaceutical Chemistry, MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, 133207, India
| | - Rina Das
- Department of Pharmaceutical Chemistry, MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, 133207, India
| | - Dinesh Kumar Mehta
- Department of Pharmaceutical Chemistry, MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, 133207, India.
| | - Diksha Sharma
- Swami Devidyal College of Pharmacy, Barwala, 134118, India
| | - Shahbaz Aman
- Department of Microbiology, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, 133207, India
| | - M U Khan
- Department of pharmaceutical Chemistry & Pharmacognosy, Unaizah College of Pharmacy, Qassim University, Uniazah, Al Qassim, Saudi Arabia
| |
Collapse
|
5
|
Saifi Z, Ali A, Inam A, Azam A, Kamthan M, Abid M, Ali I. Synthesis and antibacterial evaluation of quinoline-sulfonamide hybrid compounds: a promising strategy against bacterial resistance. RSC Adv 2025; 15:1680-1689. [PMID: 39831044 PMCID: PMC11740869 DOI: 10.1039/d4ra05069j] [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: 07/13/2024] [Accepted: 11/28/2024] [Indexed: 01/22/2025] Open
Abstract
Antibiotic-resistant bacteria are a serious global health threat, making infections harder to treat and increasing medical costs and mortality rates. To combat resistant bacterial strains, a series of compounds (QS1-12) were synthesized with an excellent yield of 85-92%. Initial assessments of these analogues as potential antibacterial agents were conducted through a preliminary screening against a panel of diverse bacterial strains. The results identified compound QS-3 as the most effective antibacterial candidate, exhibiting exceptional inhibitory activity against P. aeruginosa with a minimum inhibitory concentration (MIC) of 64 μg mL-1. Furthermore, QS-3 demonstrated a favorable synergistic effect when combined with ciprofloxacin. Notably, the compound displayed minimal cytotoxicity, inducing less than 5% lysis of red blood cells (RBCs). Significantly, QS-3 exhibited enhanced inhibitory activity, particularly against the antibiotic-resistant strains AA202 and AA290. In silico predictions of physicochemical properties underscored the drug-like qualities of the designed compounds. Additionally, molecular docking poses, ligPlot images, and a binding affinity of -8.0 kcal mol-1 further reinforced their potential as promising antibacterial agents. Briefly, the reported compound QS3 may be a future broad-range antibacterial agent.
Collapse
Affiliation(s)
- Zohaib Saifi
- Department of Chemistry, Jamia Millia Islamia Jamia Nagar New Delhi-110025 India
| | - Asghar Ali
- Department of Biosciences, Jamia Millia Islamia New Delhi-110025 India
- Department of Biochemistry, School of Chemical and Life Sciences, Jamia Hamdard New Delhi-110062 India
| | - Afreen Inam
- Department of Chemistry, Jamia Millia Islamia Jamia Nagar New Delhi-110025 India
| | - Amir Azam
- Department of Chemistry, Jamia Millia Islamia Jamia Nagar New Delhi-110025 India
| | - Mohan Kamthan
- Department of Biochemistry, School of Chemical and Life Sciences, Jamia Hamdard New Delhi-110062 India
| | - Mohammad Abid
- Department of Biosciences, Jamia Millia Islamia New Delhi-110025 India
| | - Imran Ali
- Department of Chemistry, Jamia Millia Islamia Jamia Nagar New Delhi-110025 India
| |
Collapse
|
6
|
Sharma V, Saini M, Das R, Chauhan S, Sharma D, Mujwar S, Gupta S, Mehta DK. Recent Updates on Antibacterial Quinolones: Green Synthesis, Mode of Interaction and Structure-Activity Relationship. Chem Biodivers 2025:e202401936. [PMID: 39756027 DOI: 10.1002/cbdv.202401936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 12/21/2024] [Accepted: 12/23/2024] [Indexed: 01/07/2025]
Abstract
Quinolone antibiotics are a crucial class of synthetic antibacterial agents, widely utilized due to their broad spectrum of antibacterial activity. Due to the development of antimicrobial resistance, the potency of quinolone drugs decreased. Many conventional methods have been developed to elevate amination rate and to improve yield. These methods are generally characterized by prolonged reaction durations, high boiling solvents, harsh conditions, costly reagents and excessive heat generation, which have adversely affected the therapeutic efficacy of these compounds. Recently, green chemistry has focused on sustainable chemistry-dependent quinolone analogue synthesis methods that significantly reduce bacterial infections. These methods include one-pot synthesis, photoredox catalysis, phase transfer catalysis, ultrasonic irradiation, microwave-assisted, green solvent and catalyst-free synthesis, which often utilize energy-efficient, non-toxic and less time-consuming techniques, aligning with green chemistry principles to improve safety and environmental impact. Researchers continuously explore innovative approaches to applying these methods in synthetic reactions. This review includes a comprehensive analysis of synthetic literature from the past 15 years from Scopus, PubMed, Embase and WOS using keywords, such as green chemistry, quinolone and antibacterial, highlighting significant advancements and emerging trends. This work's importance lies in its extensive literature overview on green synthesis methods for quinolones and related heterocyclic compounds. Furthermore, to provide useful information for the generation of future antibacterial drugs, some structural-activity relationship studies and in silico studies have also been included to investigate the stable binding interactions between quinolone leads and various target proteins.
Collapse
Affiliation(s)
- Vishal Sharma
- Department of Pharmaceutical Chemistry, MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Ambala, India
| | - Monika Saini
- Department of Pharmaceutical Chemistry, MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Ambala, India
| | - Rina Das
- Department of Pharmaceutical Chemistry, MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Ambala, India
| | - Samrat Chauhan
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Diksha Sharma
- Department of Pharmaceutical Chemistry, Swami Devidyal College of Pharmacy, Barwala, India
| | - Somdutt Mujwar
- Department of Pharmaceutical Chemistry, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Sumeet Gupta
- Department of Pharmacology, MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Ambala, India
| | - Dinesh Kumar Mehta
- Department of Pharmaceutical Chemistry, MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Ambala, India
| |
Collapse
|
7
|
Felix MAJ, Ragumoorthy C, Chen TW, Chen SM, Kiruthiga G, Singh A, Ghazaryan K, Al-Mohaimeed AM, Elshikh MS. Fluid-specific detection of environmental pollutant moxifloxacin hydrochloride utilizing a rare-earth niobate decorated functionalized carbon nanofiber sensor platform. ENVIRONMENTAL RESEARCH 2025; 264:120349. [PMID: 39542161 DOI: 10.1016/j.envres.2024.120349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 11/03/2024] [Accepted: 11/09/2024] [Indexed: 11/17/2024]
Abstract
The development of precise and efficient detection methods is essential for the real-time monitoring of antibiotics, especially in environmental and biological matrices. This study aims to address this challenge by introducing a novel electrochemical sensor for the targeted detection of moxifloxacin hydrochloride (MFN), a fourth-generation fluoroquinolone. The sensor is based on a holmium niobate (HNO) and functionalized carbon nanofiber (f-CNF) nanocomposite, synthesized via a hydrothermal approach and subsequently characterized for its structural and electrochemical properties. When deposited onto a glassy carbon electrode (GCE), the HNO/f-CNF nanocomposite demonstrated exceptional electrochemical performance, as assessed using cyclic voltammetry (CV) and differential pulse voltammetry (DPV). The sensor exhibited remarkable sensitivity, with a detection limit of 0.034 μM, a quantification limit of 0.11 μM, and a sensitivity of 0.69 μA μM-1 cm-2. It also achieved a broad linear detection range from 0.001 μM to 1166.11 μM, making it highly effective for MFN detection across various complex matrices, including environmental waters, biological fluids, and artificial saliva, with recovery rates between 98.15% and 101.75%. The novelty of this work lies in the unique combination of HNO's catalytic properties and f-CNF's enhanced electron transport, establishing a highly selective and sensitive platform for MFN detection. This sensor not only advances the field of electrochemical sensing but also offers a promising tool for real-time environmental and pharmaceutical monitoring.
Collapse
Affiliation(s)
- Mariya Antony John Felix
- Department of Chemical Engineering and Biotechnology, National Taipei University of Technology, No. 1, Section 3, Chung-Hsiao East Road, Taipei, 106, Taiwan
| | - Chandini Ragumoorthy
- Department of Chemical Engineering and Biotechnology, National Taipei University of Technology, No. 1, Section 3, Chung-Hsiao East Road, Taipei, 106, Taiwan
| | - Tse-Wei Chen
- Department of Materials, Imperial College London, London, SW7 2AZ, United Kingdom.
| | - Shen-Ming Chen
- Department of Chemical Engineering and Biotechnology, National Taipei University of Technology, No. 1, Section 3, Chung-Hsiao East Road, Taipei, 106, Taiwan.
| | - G Kiruthiga
- Department of Physics, Avinashilingam Institute for Home Science and Higher Education for Women, Tamil Nadu, Coimbatore, 641 043, India
| | - Abhishek Singh
- Faculty of Biology, Yerevan State University, Yerevan, 0025, Armenia
| | - Karen Ghazaryan
- Faculty of Biology, Yerevan State University, Yerevan, 0025, Armenia
| | - Amal M Al-Mohaimeed
- Department of Chemistry, College of Science, King Saud University, P.O. Box 22452, Riyadh, 11495, Saudi Arabia
| | - Mohamed S Elshikh
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia
| |
Collapse
|
8
|
Qin Q, Wu W, Che L, Zhou X, Wu D, Li X, Yang Y, Lou J. Computer-Aided Construction and Evaluation of Poly-L-Lysine/Hyodeoxycholic Acid Nanoparticles for Hemorrhage and Infection Therapy. Pharmaceutics 2024; 17:7. [PMID: 39861658 PMCID: PMC11768166 DOI: 10.3390/pharmaceutics17010007] [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: 11/10/2024] [Revised: 12/14/2024] [Accepted: 12/20/2024] [Indexed: 01/27/2025] Open
Abstract
Background: Traumatic hemorrhage and infection are major causes of mortality in wounds caused by battlefield injuries, hospital procedures, and traffic accidents. Developing a multifunctional nano-drug capable of simultaneously controlling bleeding, preventing infection, and promoting wound healing is critical. This study aimed to design and evaluate a nanoparticle-based solution to address these challenges effectively. Methods: Using a one-pot assembly approach, we prepared a series of nanoparticles composed of poly-L-lysine and hyodeoxycholic acid (PLL-HDCA NPs). Theoretical simulations and experimental studies were combined to optimize their structure and functionality. In vitro platelet aggregation, antibacterial assays, cytotoxicity tests, and hemolysis evaluations were performed. In vivo efficacy was assessed in various hemorrhage models, a full-thickness skin defect model, and a skin irritation test. Results: PLL-HDCA NPs demonstrated effective induction of platelet aggregation and significantly reduced bleeding time and blood loss in mouse models, including tail vein, femoral vein, artery, and liver bleeding. Antibacterial assays revealed strong activity against E. coli and S. aureus. Wound healing studies showed that PLL-HDCA NPs promoted tissue repair in a full-thickness skin defect model. Cytotoxicity and hemolysis tests indicated minimal impact on human cells and significantly reduced hemolysis rates compared to PLL alone. Skin irritation tests confirmed the safety of PLL-HDCA NPs for external application. Conclusions: PLL-HDCA NPs represent a safe, efficient, and multifunctional nano-drug suitable for topical applications to control bleeding, combat infection, and facilitate wound healing, making them promising candidates for use in battlefield and hospital settings.
Collapse
Affiliation(s)
- Qin Qin
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China; (Q.Q.); (D.W.)
| | - Wenxing Wu
- Department of Pharmaceutics, College of Pharmacy, Army Medical University, Chongqing 400038, China; (W.W.); (X.L.)
| | - Ling Che
- Department of Pharmacy, Medical Supplies Center of PLA General Hospital, Beijing 100853, China;
| | - Xing Zhou
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming 650500, China;
| | - Diedie Wu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China; (Q.Q.); (D.W.)
| | - Xiaohui Li
- Department of Pharmaceutics, College of Pharmacy, Army Medical University, Chongqing 400038, China; (W.W.); (X.L.)
| | - Yumin Yang
- Department of Pharmacy, Medical Supplies Center of PLA General Hospital, Beijing 100853, China;
| | - Jie Lou
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China; (Q.Q.); (D.W.)
- Department of Pharmaceutics, College of Pharmacy, Army Medical University, Chongqing 400038, China; (W.W.); (X.L.)
| |
Collapse
|
9
|
Huzum B, Aprotosoaie AC, Alexa O, Sîrbu PD, Puha B, Veliceasa B, Huzum RM. Antimicrobials in Orthopedic Infections: Overview of Clinical Perspective and Microbial Resistance. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1988. [PMID: 39768868 PMCID: PMC11728363 DOI: 10.3390/medicina60121988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 11/18/2024] [Accepted: 11/21/2024] [Indexed: 01/04/2025]
Abstract
Orthopedic infections are challenging pathologies that impose a heavy burden on patients and the healthcare system. Antimicrobial therapy is a critical component of the successful management of orthopedic infections, but its effectiveness depends on patient-, surgery-, drug-, and hospital-related factors. The dramatic increase in the emergence of multidrug-resistant microbial strains necessitates new clinical approaches in order to prevent or limit this phenomenon and to ensure a favorable therapeutic outcome. The present paper reviews the currently available antimicrobial strategies in the management of orthopedic infections, highlighting their clinical use related to the occurrence of microbial resistance. Some approaches for reducing antibiotic resistance emergence in orthopedics are also presented. The use of antibiotics tailored to the microorganism's sensitivity profile, patient factors, and pharmacokinetic profile in terms of monotherapy or combinations, the understanding of microbial pathogenicity and resistance patterns, strict control measures in healthcare facilities, the development of new antimicrobial therapies (drugs, devices, technologies), and patient education for improving compliance and tolerance are some of the most important tools for overcoming microbial resistance.
Collapse
Affiliation(s)
- Bogdan Huzum
- Department of Orthopaedic and Traumatology, Faculty of Medicine “Grigore T. Popa”, University of Medicine and Pharmacy of Iasi, 700115 Iasi, Romania; (B.H.); (O.A.); (P.D.S.); (B.P.); (B.V.)
| | - Ana Clara Aprotosoaie
- Faculty of Pharmacy “Grigore T. Popa”, University of Medicine and Pharmacy of Iasi, 700115 Iasi, Romania
| | - Ovidiu Alexa
- Department of Orthopaedic and Traumatology, Faculty of Medicine “Grigore T. Popa”, University of Medicine and Pharmacy of Iasi, 700115 Iasi, Romania; (B.H.); (O.A.); (P.D.S.); (B.P.); (B.V.)
| | - Paul Dan Sîrbu
- Department of Orthopaedic and Traumatology, Faculty of Medicine “Grigore T. Popa”, University of Medicine and Pharmacy of Iasi, 700115 Iasi, Romania; (B.H.); (O.A.); (P.D.S.); (B.P.); (B.V.)
| | - Bogdan Puha
- Department of Orthopaedic and Traumatology, Faculty of Medicine “Grigore T. Popa”, University of Medicine and Pharmacy of Iasi, 700115 Iasi, Romania; (B.H.); (O.A.); (P.D.S.); (B.P.); (B.V.)
| | - Bogdan Veliceasa
- Department of Orthopaedic and Traumatology, Faculty of Medicine “Grigore T. Popa”, University of Medicine and Pharmacy of Iasi, 700115 Iasi, Romania; (B.H.); (O.A.); (P.D.S.); (B.P.); (B.V.)
| | - Riana Maria Huzum
- Department of Radiology, Faculty of Medicine “Grigore T. Popa”, University of Medicine and Pharmacy of Iasi, 700115 Iasi, Romania;
| |
Collapse
|
10
|
Aleksandrova EV, Ma CX, Klepacki D, Alizadeh F, Vázquez-Laslop N, Liang JH, Polikanov YS, Mankin AS. Macrolones target bacterial ribosomes and DNA gyrase and can evade resistance mechanisms. Nat Chem Biol 2024; 20:1680-1690. [PMID: 39039256 PMCID: PMC11686707 DOI: 10.1038/s41589-024-01685-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 06/19/2024] [Indexed: 07/24/2024]
Abstract
Growing resistance toward ribosome-targeting macrolide antibiotics has limited their clinical utility and urged the search for superior compounds. Macrolones are synthetic macrolide derivatives with a quinolone side chain, structurally similar to DNA topoisomerase-targeting fluoroquinolones. While macrolones show enhanced activity, their modes of action have remained unknown. Here, we present the first structures of ribosome-bound macrolones, showing that the macrolide part occupies the macrolide-binding site in the ribosomal exit tunnel, whereas the quinolone moiety establishes new interactions with the tunnel. Macrolones efficiently inhibit both the ribosome and DNA topoisomerase in vitro. However, in the cell, they target either the ribosome or DNA gyrase or concurrently both of them. In contrast to macrolide or fluoroquinolone antibiotics alone, dual-targeting macrolones are less prone to select resistant bacteria carrying target-site mutations or to activate inducible macrolide resistance genes. Furthermore, because some macrolones engage Erm-modified ribosomes, they retain activity even against strains with constitutive erm resistance genes.
Collapse
Affiliation(s)
- Elena V Aleksandrova
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Cong-Xuan Ma
- Key Laboratory of Medicinal Molecule Science and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, China
| | - Dorota Klepacki
- Center for Biomolecular Sciences, University of Illinois at Chicago, Chicago, IL, USA
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Faezeh Alizadeh
- Center for Biomolecular Sciences, University of Illinois at Chicago, Chicago, IL, USA
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Nora Vázquez-Laslop
- Center for Biomolecular Sciences, University of Illinois at Chicago, Chicago, IL, USA
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Jian-Hua Liang
- Key Laboratory of Medicinal Molecule Science and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, China.
| | - Yury S Polikanov
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL, USA.
- Center for Biomolecular Sciences, University of Illinois at Chicago, Chicago, IL, USA.
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, USA.
| | - Alexander S Mankin
- Center for Biomolecular Sciences, University of Illinois at Chicago, Chicago, IL, USA.
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
11
|
Rusu A, Oancea OL, Tanase C, Uncu L. Unlocking the Potential of Pyrrole: Recent Advances in New Pyrrole-Containing Compounds with Antibacterial Potential. Int J Mol Sci 2024; 25:12873. [PMID: 39684580 PMCID: PMC11640851 DOI: 10.3390/ijms252312873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Nitrogen heterocycles are valuable structural elements in the molecules of antibacterial drugs approved and used to treat bacterial infections. Pyrrole is a five-atom heterocycle found in many natural compounds with biological activity, including antibacterial activity. Numerous compounds are being develop based on the pyrrole heterocycle as new potential antibacterial drugs. Due to the phenomenon of antibacterial resistance, there is a continuous need to create new effective antibacterials. In the scientific literature, we have identified the most relevant studies that aim to develop new compounds, such as pyrrole derivatives, that are proven to have antibacterial activity. Nature is an endless reservoir of inspiration for designing new compounds based on the structure of pyrrole heterocycles such as calcimycin, lynamycins, marinopyrroles, nargenicines, phallusialides, and others. However, many other synthetic compounds based on the pyrrole heterocycle have been developed and can be optimized in the future. The identified compounds were classified according to the type of chemical structure. The chemical structure-activity relationships, mechanisms of action, and antibacterial effectiveness of the most valuable compounds were highlighted. This review highlights scientific progress in designing new pyrrole-containing compounds and provides examples of lead compounds that can be successfully optimized further.
Collapse
Affiliation(s)
- Aura Rusu
- Pharmaceutical and Therapeutic Chemistry Department, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania;
| | - Octavia-Laura Oancea
- Organic Chemistry Department, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania;
| | - Corneliu Tanase
- Pharmaceutical Botany Department, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania
| | - Livia Uncu
- Scientific Center for Drug Research, Pharmaceutical and Toxicological Chemistry Department, “Nicolae Testemitanu” State University of Medicine and Pharmacy, 165 Bd. Stefan Cel Mare si Sfant, MD-2004 Chisinau, Moldova;
| |
Collapse
|
12
|
Lungu IA, Oancea OL, Rusu A. In Silico Study of the Potential Inhibitory Effects on Escherichia coli DNA Gyrase of Some Hypothetical Fluoroquinolone-Tetracycline Hybrids. Pharmaceuticals (Basel) 2024; 17:1540. [PMID: 39598450 PMCID: PMC11597511 DOI: 10.3390/ph17111540] [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: 10/15/2024] [Revised: 11/12/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND/OBJECTIVES Despite the discovery of antibiotics, bacterial infections persist globally, exacerbated by rising antimicrobial resistance that results in millions of cases, increased healthcare costs, and more extended hospital stays. The urgent need for new antibacterial drugs continues as resistance evolves. Fluoroquinolones and tetracyclines are versatile antibiotics that are effective against various bacterial infections. A hybrid antibiotic combines two or more molecules to enhance antimicrobial effectiveness and combat resistance better than monotherapy. Fluoroquinolones are ideal candidates for hybridization due to their potent bactericidal effects, ease of synthesis, and ability to form combinations with other molecules. METHODS This study explored the mechanisms of action for 40 hypothetical fluoroquinolone-tetracycline hybrids, all of which could be obtained using a simple, eco-friendly synthesis method. Their interaction with Escherichia coli DNA Gyrase and similarity to albicidin were evaluated using the FORECASTER platform. RESULTS Hybrids such as Do-Ba, Mi-Fi, and Te-Ba closely resembled albicidin in physicochemical properties and FITTED Scores, while Te-De surpassed it with a better score. Similar to fluoroquinolones, these hybrids likely inhibit DNA synthesis by binding to enzyme-DNA complexes. CONCLUSIONS These hybrids could offer broad-spectrum activity and help mitigate bacterial resistance, though further in vitro and in vivo studies are needed to validate their potential.
Collapse
Affiliation(s)
- Ioana-Andreea Lungu
- Medicine and Pharmacy Doctoral School, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Targu Mures, Romania
| | - Octavia-Laura Oancea
- Organic Chemistry Department, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Targu Mures, Romania;
| | - Aura Rusu
- Pharmaceutical and Therapeutic Chemistry Department, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Targu Mures, Romania;
| |
Collapse
|
13
|
Ommi O, Dhopat PS, Sau S, Estharla MR, Nanduri S, Kalia NP, Yaddanapudi VM. Design, synthesis, and biological evaluation of pyrazole-ciprofloxacin hybrids as antibacterial and antibiofilm agents against Staphylococcus aureus. RSC Med Chem 2024; 16:d4md00623b. [PMID: 39493222 PMCID: PMC11528910 DOI: 10.1039/d4md00623b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 10/15/2024] [Indexed: 11/05/2024] Open
Abstract
In our continued efforts to tackle antibiotic resistance, a new series of pyrazole-ciprofloxacin hybrids were designed, synthesized, and evaluated for their antibacterial activity against Staphylococcus aureus (S. aureus), Pseudomonas aeruginosa (P. aeruginosa), and Mycobacterium tuberculosis (Mtb). Most of the compounds exhibited good to excellent activities against S. aureus, and six compounds (7a, 7b, 7d, 7g, 7k, and 7p) exhibited higher or comparable activity (MIC = 0.125-0.5 μg mL-1) to ciprofloxacin (0.125 μg mL-1). Further, these selected compounds were non-toxic (CC50 ≥ 1000 μg mL-1) when evaluated for cell viability test against the Hep-G2 cell line. Three compounds (7a, 7d, and 7g) demonstrated excellent activity against ciprofloxacin-resistant S. aureus with MIC values ranging from 0.125-0.5 μg mL-1 and good antibiofilm activity. Among them, 7g displayed remarkable antibiofilm activity with an MBIC50 value of 0.02 μg mL-1, which is 50 times lower than ciprofloxacin (MBIC50 = 1.06 μg mL-1). A time-kill kinetics study indicated that 7g showed both concentration and time-dependent bactericidal properties. In addition, 7g effectively inhibited DNA-gyrase supercoiling activity at 1 μg mL-1 (8× MIC). Two compounds 7b and 7d exhibited the highest activity against Mtb with a MIC of 0.5 μg mL-1, while 7c showed the highest activity against P. aeruginosa with a MIC value of 2 μg mL-1. Molecular docking studies revealed that 7g formed stable interactions at the DNA active site.
Collapse
Affiliation(s)
- Ojaswitha Ommi
- Work carried out at Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER) Balanagar Hyderabad 500037 Telangana India
| | - Priyanka Sudhir Dhopat
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER) Balanagar Hyderabad 500037 Telangana India
| | - Shashikanta Sau
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER) Balanagar Hyderabad 500037 Telangana India
| | - Madhu Rekha Estharla
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER) Balanagar Hyderabad 500037 Telangana India
| | - Srinivas Nanduri
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER) Balanagar Hyderabad 500037 Telangana India
| | - Nitin Pal Kalia
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER) Balanagar Hyderabad 500037 Telangana India
| | - Venkata Madhavi Yaddanapudi
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER) Balanagar Hyderabad 500037 Telangana India
| |
Collapse
|
14
|
Dang X, Li N, Yu Z, Ji X, Yang M, Wang X. Advances in the preparation and application of cellulose-based antimicrobial materials: A review. Carbohydr Polym 2024; 342:122385. [PMID: 39048226 DOI: 10.1016/j.carbpol.2024.122385] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 06/06/2024] [Accepted: 06/07/2024] [Indexed: 07/27/2024]
Abstract
The rise of polymer materials in modern life has drawn attention to renewable, easily biodegradable, environmentally-friendly bio-based polymers. Notably, significant research has been dedicated to creating green antimicrobial functional materials for the biomedical field using natural polymer materials. Cellulose is a rich natural biomass organic polymer material. Given its favorable attributes like film-forming capability, biodegradability, and biocompatibility, it is extensively employed to tackle a wide range of challenges confronting humanity today. However, its inherent drawbacks, such as insolubility in water and most organic solvents, hygroscopic nature, difficulty in melting, and limited antimicrobial properties, continue to pose challenges for realizing the high-value applications of cellulose. Achieving multifunctionality and more efficient application of cellulose still poses major challenges. In this regard, the current development status of cellulose materials was reviewed, covering the classification, preparation methods, and application status of cellulose-based antimicrobial materials. The application value of cellulose-based antimicrobial materials in biomedicine, textiles, food packaging, cosmetics and wastewater treatment was summarised. Finally, insights were provided into the developing prospects of cellulose-based antimicrobial materials were provided.
Collapse
Affiliation(s)
- Xugang Dang
- State Key Laboratory of Biobased Material and Green Papermaking, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China; Institute for Biomass and Function Materials & College of Bioresources Chemistry and Materials Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China.
| | - Nan Li
- Institute for Biomass and Function Materials & College of Bioresources Chemistry and Materials Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China
| | - Zhenfu Yu
- Institute for Biomass and Function Materials & College of Bioresources Chemistry and Materials Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China
| | - Xingxiang Ji
- State Key Laboratory of Biobased Material and Green Papermaking, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China.
| | - Mao Yang
- State Key Laboratory of Biobased Material and Green Papermaking, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China
| | - Xuechuan Wang
- Institute for Biomass and Function Materials & College of Bioresources Chemistry and Materials Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China
| |
Collapse
|
15
|
Nguyen TD, Itayama T, Iwami N, Shimizu K, Dao TS, Pham TL, Tran VQ, Maseda H. Toxicity of ciprofloxacin and ofloxacin to Moina macrocopa and investigation of p-value adjustments for (eco)toxicological studies. Drug Chem Toxicol 2024; 47:662-673. [PMID: 37491899 DOI: 10.1080/01480545.2023.2239524] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 06/19/2023] [Accepted: 07/14/2023] [Indexed: 07/27/2023]
Abstract
Ciprofloxacin (CFX) and ofloxacin (OFX) are commonly found as residual contaminants in aquatic environments, posing potential risks to various species. To ensure the safety of aquatic wildlife, it is essential to determine the toxicity of these antibiotics and establish appropriate concentration limits. Additionally, in (eco)toxicological studies, addressing the issue of multiple hypothesis testing through p-value adjustments is crucial for robust decision-making. In this study, we assessed the no observed adverse effect concentration (NOAEC) of CFX and OFX on Moina macrocopa across a concentration range of 0-400 µg L-1. Furthermore, we investigated multiple p-value adjustments to determine the NOAECs. Our analysis yielded consistent results across seven different p-value adjustments, indicating NOAECs of 100 µg CFX L-1 for age at first reproduction and 200 µg CFX L-1 for fertility. For OFX treatment, a NOAEC of 400 µg L-1 was observed for both biomarkers. However, further investigation is required to establish the NOAEC of OFX at higher concentrations with greater certainty. Our findings demonstrate that CFX exhibits higher toxicity compared to OFX, consistent with previous research. Moreover, this study highlights the differential performance of p-value adjustment methods in terms of maintaining statistical power while controlling the multiplicity problem, and their practical applicability. The study emphasizes the low NOAECs for these antibiotics in the zooplanktonic group, highlighting their significant risks to ecological and environmental safety. Additionally, our investigation of p-value adjustment approaches contributes to a deeper understanding of their performance characteristics, enabling (eco)toxicologists to select appropriate methods based on their specific needs and priorities.
Collapse
Affiliation(s)
- Tan-Duc Nguyen
- Graduate School of Engineering, Nagasaki University, Nagasaki City, Japan
| | - Tomoaki Itayama
- Graduate School of Engineering, Nagasaki University, Nagasaki City, Japan
| | - Norio Iwami
- School of Science and Engineering, Meisei University, Hino City, Japan
| | - Kazuya Shimizu
- Faculty of Life Sciences, Toyo University, Gunma City, Japan
| | - Thanh-Son Dao
- Faculty of Environment and Natural Resources, Ho Chi Minh City University of Technology (HCMUT), Ho Chi Minh City, Vietnam
- Vietnam National University Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Thanh Luu Pham
- Vietnam Academy of Science and Technology (VAST), Graduate University of Science and Technology, Hanoi City, Vietnam
- Institute of Tropical Biology, Vietnam Academy of Science and Technology (VAST), Ho Chi Minh City, Vietnam
| | - Vinh Quang Tran
- Asian Centre for Water Research (CARE), Ho Chi Minh City University of Technology (HCMUT), Ho Chi Minh City, Vietnam
| | - Hideaki Maseda
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology, Ikeda City, Japan
| |
Collapse
|
16
|
Tang F, Zou T, Wang Z, Zhang J. Fabrication of fluorinated triazine-based covalent organic frameworks for selective extraction of fluoroquinolone in milk. J Chromatogr A 2024; 1730:465078. [PMID: 38889582 DOI: 10.1016/j.chroma.2024.465078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 05/12/2024] [Accepted: 06/11/2024] [Indexed: 06/20/2024]
Abstract
A novel fluorinated triazine-based covalent organic frameworks (F-CTFs) was designed and synthesized by using melamine and 2,3,5,6-tetrafluoroterephthalaldehydeas as organic ligands for selective pipette tip solid-phase extraction (PT-SPE) of amphiphilic fluoroquinolones (FQs). The competitive adsorption experiment and mechanism study were carried out and verified that this F-CTFs possesses favorable adsorption affinity for FQs. The abundant fluorine affinity sites endowed the F-CTFs high selectivity to FQs extraction through F-F interactions. The adsorption capacity of F-CTFs can reach up to 109.1 mg g-1 for enrofloxacin. The detailed characterization of the F-CTFs adsorbent involved the application of various techniques to examine its morphology and structure. Under optimized conditions, a method combining F-CTF-based PT-SPE with high-performance liquid chromatography (PT-SPE-HPLC) was established, which exhibited a broad linear range, excellent precision, and an impressively low limit of detection, and could be used for the determination of six FQs in milk, with LODs as low as 0.0010 μg mL-1. The recovery rates during extraction varied between 92.1% and 111.4%, exhibiting RSDs below 6.8% at different spiked concentrations.
Collapse
Affiliation(s)
- Furong Tang
- School of Chemistry and Environmental Engineering, Hubei Key Laboratory of Novel Reactor and Green Chemical Technology, Key Laboratory for Green Chemical Process of Ministry of Education, Wuhan Institute of Technology, Wuhan 430205, China
| | - Ting Zou
- School of Chemistry and Environmental Engineering, Hubei Key Laboratory of Novel Reactor and Green Chemical Technology, Key Laboratory for Green Chemical Process of Ministry of Education, Wuhan Institute of Technology, Wuhan 430205, China
| | - Ziyi Wang
- School of Chemistry and Environmental Engineering, Hubei Key Laboratory of Novel Reactor and Green Chemical Technology, Key Laboratory for Green Chemical Process of Ministry of Education, Wuhan Institute of Technology, Wuhan 430205, China
| | - Juan Zhang
- School of Chemistry and Environmental Engineering, Hubei Key Laboratory of Novel Reactor and Green Chemical Technology, Key Laboratory for Green Chemical Process of Ministry of Education, Wuhan Institute of Technology, Wuhan 430205, China; School of Chemical Engineering and Pharmacy, Key Laboratory of Novel Biomass-Based Environmental and Energy Materials in Petroleum and Chemical Industry, Wuhan Institute of Technology, Wuhan 430205, China.
| |
Collapse
|
17
|
Thitirungreangchai T, Roytrakul S, Aunpad R. Deciphering the Intracellular Action of the Antimicrobial Peptide A11 via an In-Depth Analysis of Its Effect on the Global Proteome of Acinetobacter baumannii. ACS Infect Dis 2024; 10:2795-2813. [PMID: 39075773 PMCID: PMC11320580 DOI: 10.1021/acsinfecdis.4c00160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 07/17/2024] [Accepted: 07/17/2024] [Indexed: 07/31/2024]
Abstract
The potential antimicrobial activity and low propensity to induce the development of bacterial resistance have rendered antimicrobial peptides (AMPs) as novel and ideal candidate therapeutic agents for the treatment of infections caused by drug-resistant pathogenic bacteria. The targeting of bacterial membranes by AMPs has been typically considered their sole mode of action; however, increasing evidence supports the existence of multiple and complementary functions of AMPs that result in bacterial death. An in-depth characterization of their mechanism of action could facilitate further research and development of AMPs with higher potency. The current study employs biophysics and proteomics approaches to unveil the mechanisms underlying the antibacterial activity of A11, a potential candidate AMP, against Acinetobacter baumannii, a leading cause of hospital-acquired infections (HAIs) and consequently, a serious global threat. A11 peptide was found to induce membrane depolarization to a high extent, as revealed by flow cytometry and electron microscopy analyses. The prompt intracellular penetration of A11 peptide, observed using confocal microscopy, was found to occur concomitantly with a very low degree of membrane lysis, suggesting that its mode of action predominantly involves a nonlytic killing mechanism. Quantitative proteomics analysis employed for obtaining insights into the mechanisms underlying the antimicrobial activity of A11 peptide revealed that it disrupted energy metabolism, interfered with protein homeostasis, and inhibited fatty acid synthesis that is essential for cell membrane integrity; all these impacted the cellular functions of A. baumannii. A11 treatment also impacted signal transduction associated with the regulation of biofilm formation, hindered the stress response, and influenced DNA repair processes; these are all crucial survival mechanisms of A. baumannii. Additionally, robust antibacterial activity was exhibited by A11 peptide against multidrug-resistant (MDR) and extensively drug-resistant (XDR) clinical isolates of A. baumannii; moreover, A11 peptide exhibited synergy with levofloxacin and minocycline as well as low propensity for inducing resistance. Taken together, the findings emphasize the therapeutic potential of A11 peptide as an antibacterial agent against drug-resistant A. baumannii and underscore the need for further investigation.
Collapse
Affiliation(s)
- Thanit Thitirungreangchai
- Graduate
Program in Biomedical Sciences, Faculty of Allied Health Sciences, Thammasat University, Khlong Luang, Pathum Thani 12120, Thailand
| | - Sittiruk Roytrakul
- Functional
Proteomics Technology Laboratory, National Center for Genetic Engineering
and Biotechnology, National Science and
Technology Development Agency, Khlong Luang, Pathum Thani 12120, Thailand
| | - Ratchaneewan Aunpad
- Graduate
Program in Biomedical Sciences, Faculty of Allied Health Sciences, Thammasat University, Khlong Luang, Pathum Thani 12120, Thailand
| |
Collapse
|
18
|
Najafi D, Siri G, Sadri M, Yazdani O, Esbati R, Karimi P, Keshavarz A, Mehmandar-Oskuie A, Ilktac M. Combination MEG3 lncRNA and Ciprofloxacin dramatically decreases cell migration and viability as well as induces apoptosis in GC cells in vitro. Biotechnol Appl Biochem 2024; 71:809-816. [PMID: 38499448 DOI: 10.1002/bab.2578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 02/25/2024] [Indexed: 03/20/2024]
Abstract
Gastric cancer (GC) is a prominent cause of cancer-related mortality worldwide. Long noncoding RNA (lncRNA) maternal expression gene3 (MEG3) participates in numerous signaling pathways by targeting the miRNA-mRNA axis. Studies on human tumors have demonstrated that the antibiotic Ciprofloxacin induces cell cycle changes, programmed cell death, and growth suppression. In this study, we transfected MEG3 lncRNA and Ciprofloxacin into the MKN-45 GC cell line. qRT-PCR was employed to evaluate the effects on the specific microRNA and mRNA. The wound healing test, MTT assay, and flow cytometry were used to assess the impact of their administration on cell migration, viability, and apoptosis, respectively. Research showed that miR-147 expression fell even more after MEG3 lncRNA transfection, leading to an increase in B-cell lymphoma 2 (BCL-2) levels. Ciprofloxacin transfection did not significantly affect the axis, except for MEG3, which led to its slight upregulation. MEG3 lncRNA inhibited the migration of MKN-45 cells compared to the control group. When MEG3 lncRNA was coupled with Ciprofloxacin, there was a significant reduction in cell migration compared to untreated groups and controls. MTT assay and flow cytometry demonstrated that MEG3 lncRNA decreased cell viability and triggered apoptosis. Simultaneous administration of MEG3 lncRNA and Ciprofloxacin revealed a significant reduction in cell viability caused by increased apoptosis obtained from MTT or flow cytometry assays. Modulating the miR-147-BCL-2 axis decreases cell migration and survival while promoting cell death. In conclusion, combining MEG3 lncRNA with Ciprofloxacin may be an effective therapeutic approach for GC treatment by influencing the miR-14-BCl-2 axis, resulting in reduced cell viability, migration, and increased apoptosis.
Collapse
Affiliation(s)
- Dena Najafi
- Faculty of Pharmacy, Eastern Mediterranean University, Famagusta, North Cyprus, Turkey
| | - Goli Siri
- Department of Internal Medicine, Amir Alam Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Sadri
- Department of Internal Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Omid Yazdani
- Department of Medical Science, School of Medicine, Shahid Beheshti University, Tehran, Iran
| | - Romina Esbati
- Research Center for Social Determinants of Health, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parvin Karimi
- Fars Population-Based Cancer Registry, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Keshavarz
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amirreza Mehmandar-Oskuie
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehmet Ilktac
- Faculty of Pharmacy, Eastern Mediterranean University, Famagusta, North Cyprus, Turkey
| |
Collapse
|
19
|
Ross CL, Lawer A, Sircombe KJ, Pletzer D, Gamble AB, Hook S. Site-Specific Antimicrobial Activity of a Dual-Responsive Ciprofloxacin Prodrug. J Med Chem 2024; 67:9599-9612. [PMID: 38780408 DOI: 10.1021/acs.jmedchem.4c00724] [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: 05/25/2024]
Abstract
Bacterial infections create distinctive microenvironments with a unique mix of metabolites and enzymes compared with healthy tissues that can be used to trigger the activation of antibiotic prodrugs. Here, a single and dual prodrug masking the C3 carboxylate and C7 piperazine of the fluoroquinolone, ciprofloxacin, responsive to nitroreductase (NTR) and/or hydrogen sulfide (H2S), was developed. Masking both functional groups reduced the activity of the prodrug against Staphylococcus aureus and Escherichia coli, increasing its minimum inhibitory concentration (MIC) by ∼512-fold (S. aureus) and ∼8000-fold (E. coli strains), while masking a single group only increased the MIC by ∼128-fold. Bacteria subjected to prolonged prodrug exposure did not show any increase in resistance. Triggering assays demonstrated the conversion of prodrugs to ciprofloxacin, and in a murine infection model, responsive prodrugs showed antibacterial activity comparable to that of ciprofloxacin, suggesting in vivo activation of prodrugs. Thus, the potential for site-specific antibiotic treatment with reduced threat of resistance is demonstrated.
Collapse
Affiliation(s)
- Catherine L Ross
- School of Pharmacy, University of Otago, Dunedin 9054, New Zealand
- Department of Microbiology and Immunology, University of Otago, Dunedin 9054, New Zealand
| | - Aggie Lawer
- School of Pharmacy, University of Otago, Dunedin 9054, New Zealand
| | - Kathleen J Sircombe
- School of Pharmacy, University of Otago, Dunedin 9054, New Zealand
- Department of Microbiology and Immunology, University of Otago, Dunedin 9054, New Zealand
| | - Daniel Pletzer
- Department of Microbiology and Immunology, University of Otago, Dunedin 9054, New Zealand
| | - Allan B Gamble
- School of Pharmacy, University of Otago, Dunedin 9054, New Zealand
| | - Sarah Hook
- School of Pharmacy, University of Otago, Dunedin 9054, New Zealand
| |
Collapse
|
20
|
Teng T, Chen S, Huo F, Jia J, Zhao L, Jiang G, Wang F, Chu N, Huang H. Efflux pump effects on levofloxacin resistance in Mycobacterium abscessus. Antimicrob Agents Chemother 2024; 68:e0134823. [PMID: 38572960 PMCID: PMC11064541 DOI: 10.1128/aac.01348-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 02/27/2024] [Indexed: 04/05/2024] Open
Abstract
Mycobacterium abscessus (M. abscessus) inherently displays resistance to most antibiotics, with the underlying drug resistance mechanisms remaining largely unexplored. Efflux pump is believed to play an important role in mediating drug resistance. The current study examined the potential of efflux pump inhibitors to reverse levofloxacin (LFX) resistance in M. abscessus. The reference strain of M. abscessus (ATCC19977) and 60 clinical isolates, including 41 M. abscessus subsp. abscessus and 19 M. abscessus subsp. massilense, were investigated. The drug sensitivity of M. abscessus against LFX alone or in conjunction with efflux pump inhibitors, including verapamil (VP), reserpine (RSP), carbonyl cyanide 3-chlorophenylhydrazone (CCCP), or dicyclohexylcarbodiimide (DCC), were determined by AlarmarBlue microplate assay. Drug-resistant regions of the gyrA and gyrB genes from the drug-resistant strains were sequenced. The transcription level of the efflux pump genes was monitored using qRT-PCR. All the tested strains were resistant to LFX. The drug-resistant regions from the gyrA and gyrB genes showed no mutation associated with LFX resistance. CCCP, DCC, VP, and RSP increased the susceptibility of 93.3% (56/60), 91.7% (55/60), 85% (51/60), and 83.3% (50/60) isolates to LFX by 2 to 32-fold, respectively. Elevated transcription of seven efflux pump genes was observed in isolates with a high reduction in LFX MIC values in the presence of efflux pump inhibitors. Efflux pump inhibitors can improve the antibacterial activity of LFX against M. abscessus in vitro. The overexpression of efflux-related genes in LFX-resistant isolates suggests that efflux pumps are associated with the development of LFX resistance in M. abscessus.
Collapse
Affiliation(s)
- Tianlu Teng
- Department of Respiratory and Critical Care Medicine, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing, China
- National Clinical Laboratory on Tuberculosis, Beijing Key Laboratory for Drug Resistant Tuberculosis Research, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing, China
| | - Suting Chen
- National Clinical Laboratory on Tuberculosis, Beijing Key Laboratory for Drug Resistant Tuberculosis Research, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing, China
| | - Fengmin Huo
- National Clinical Laboratory on Tuberculosis, Beijing Key Laboratory for Drug Resistant Tuberculosis Research, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing, China
| | - Junnan Jia
- National Clinical Laboratory on Tuberculosis, Beijing Key Laboratory for Drug Resistant Tuberculosis Research, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing, China
| | - Liping Zhao
- National Clinical Laboratory on Tuberculosis, Beijing Key Laboratory for Drug Resistant Tuberculosis Research, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing, China
| | - Guanglu Jiang
- National Clinical Laboratory on Tuberculosis, Beijing Key Laboratory for Drug Resistant Tuberculosis Research, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing, China
| | - Fen Wang
- National Clinical Laboratory on Tuberculosis, Beijing Key Laboratory for Drug Resistant Tuberculosis Research, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing, China
| | - Naihui Chu
- Department of Tuberculosis, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing, China
| | - Hairong Huang
- National Clinical Laboratory on Tuberculosis, Beijing Key Laboratory for Drug Resistant Tuberculosis Research, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing, China
| |
Collapse
|
21
|
Wang A, Wen Y, Zhu X, Zhou J, Chen Y, Liu H, Liang C, Liu E, Zhang Y, Ai G, Gaiping Z. Quantum dot-based fluorescence-linked immunosorbent assay for the rapid detection of lomefloxacin in animal-derived foods. Food Addit Contam Part A Chem Anal Control Expo Risk Assess 2024; 41:513-524. [PMID: 38502862 DOI: 10.1080/19440049.2023.2267144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 10/01/2023] [Indexed: 03/21/2024]
Abstract
Lomefloxacin (LMF), a third-generation fluoroquinolone antibacterial agent, is often used to treat bacterial and mycoplasma infections. However, due to its prolonged half-life and slow metabolism, it is prone to residues in animal-derived foods, posing a potential food safety risk. Therefore, it is particularly urgent and important to establish a method for detecting lomefloxacin. In this study, direct and indirect competitive fluorescence-linked immunosorbent assay (dc-FLISA and ic-FLISA) based on quantum dots (QDs) was established for the detection of LMF. As for dc-FLISA, the half-maximal inhibitory concentration (IC50) and limit of detection (LOD) were 0.84 ng/mL, 0.04 ng/mL, respectively, the detection ranges from 0.08 to 9.11 ng/mL. The IC50 and LOD of ic-FLISA were 0.43 ng/mL and 0.03 ng/mL, respectively, meanwhile the detection ranges from 0.05 to 3.49 ng/mL. The recoveries of dc-FLISA and ic-FLISA in animal-derived foods (milk, fish, chicken, and honey), ranged from 95.8% to 105.2% and from 96.3% to 103.4%, respectively, with the coefficients of variation less than 8%. These results suggest that the dc-FLISA and ic-FLISA methods, which are based on QD labelling, are highly sensitive and cost-effective, and can be effectively used to detect LMF in animal-derived foods.
Collapse
Affiliation(s)
- Aiping Wang
- School of Life Sciences, Zhengzhou University, Zhengzhou, P.R. China
- Longhu Laboratory, Zhengzhou, P.R. China
- Henan Key Laboratory of Immunobiology, Zhengzhou, P.R. China
| | - Yihong Wen
- School of Life Sciences, Zhengzhou University, Zhengzhou, P.R. China
- Henan Key Laboratory of Immunobiology, Zhengzhou, P.R. China
| | - Xifang Zhu
- School of Life Sciences, Zhengzhou University, Zhengzhou, P.R. China
- Longhu Laboratory, Zhengzhou, P.R. China
- Henan Key Laboratory of Immunobiology, Zhengzhou, P.R. China
| | - Jingming Zhou
- School of Life Sciences, Zhengzhou University, Zhengzhou, P.R. China
- Longhu Laboratory, Zhengzhou, P.R. China
- Henan Key Laboratory of Immunobiology, Zhengzhou, P.R. China
| | - Yumei Chen
- School of Life Sciences, Zhengzhou University, Zhengzhou, P.R. China
- Longhu Laboratory, Zhengzhou, P.R. China
- Henan Key Laboratory of Immunobiology, Zhengzhou, P.R. China
| | - Hongliang Liu
- School of Life Sciences, Zhengzhou University, Zhengzhou, P.R. China
- Longhu Laboratory, Zhengzhou, P.R. China
- Henan Key Laboratory of Immunobiology, Zhengzhou, P.R. China
| | - Chao Liang
- School of Life Sciences, Zhengzhou University, Zhengzhou, P.R. China
- Longhu Laboratory, Zhengzhou, P.R. China
- Henan Key Laboratory of Immunobiology, Zhengzhou, P.R. China
| | - Enping Liu
- School of Life Sciences, Zhengzhou University, Zhengzhou, P.R. China
- Longhu Laboratory, Zhengzhou, P.R. China
- Henan Key Laboratory of Immunobiology, Zhengzhou, P.R. China
| | - Ying Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou, P.R. China
- Longhu Laboratory, Zhengzhou, P.R. China
- Henan Key Laboratory of Immunobiology, Zhengzhou, P.R. China
| | - Guoping Ai
- School of Life Sciences, Zhengzhou University, Zhengzhou, P.R. China
- Henan Key Laboratory of Immunobiology, Zhengzhou, P.R. China
| | - Zhang Gaiping
- School of Life Sciences, Zhengzhou University, Zhengzhou, P.R. China
- Longhu Laboratory, Zhengzhou, P.R. China
- Henan Key Laboratory of Immunobiology, Zhengzhou, P.R. China
- School of Advanced Agricultural Sciences, Peking University, Beijing, P.R. China
| |
Collapse
|
22
|
Volynkina IA, Bychkova EN, Karakchieva AO, Tikhomirov AS, Zatonsky GV, Solovieva SE, Martynov MM, Grammatikova NE, Tereshchenkov AG, Paleskava A, Konevega AL, Sergiev PV, Dontsova OA, Osterman IA, Shchekotikhin AE, Tevyashova AN. Hybrid Molecules of Azithromycin with Chloramphenicol and Metronidazole: Synthesis and Study of Antibacterial Properties. Pharmaceuticals (Basel) 2024; 17:187. [PMID: 38399402 PMCID: PMC10892836 DOI: 10.3390/ph17020187] [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: 12/19/2023] [Revised: 01/23/2024] [Accepted: 01/25/2024] [Indexed: 02/25/2024] Open
Abstract
The sustained rise of antimicrobial resistance (AMR) causes a strong need to develop new antibacterial agents. One of the methods for addressing the problem of antibiotic resistance is through the design of hybrid antibiotics. In this work, we proposed a synthetic route for the conjugation of an azithromycin derivative with chloramphenicol and metronidazole hemisuccinates and synthesized two series of new hybrid molecules 4a-g and 5a-g. While a conjugation did not result in tangible synergy for wild-type bacterial strains, new compounds were able to overcome AMR associated with the inducible expression of the ermC gene on a model E. coli strain resistant to macrolide antibiotics. The newly developed hybrids demonstrated a tendency to induce premature ribosome stalling, which might be crucial since they will not induce a macrolide-resistant phenotype in a number of pathogenic bacterial strains. In summary, the designed structures are considered as a promising direction for the further development of hybrid molecules that can effectively circumvent AMR mechanisms to macrolide antibiotics.
Collapse
Affiliation(s)
- Inna A. Volynkina
- Department of Chemistry, Lomonosov Moscow State University, Leninskie Gory 1, 119234 Moscow, Russia; (A.O.K.); (P.V.S.); (O.A.D.); (I.A.O.)
| | - Elena N. Bychkova
- Gause Institute of New Antibiotics, B. Pirogovskaya 11, 119021 Moscow, Russia; (E.N.B.); (A.S.T.); (G.V.Z.); (S.E.S.); (M.M.M.); (N.E.G.); (A.E.S.)
| | - Anastasiia O. Karakchieva
- Department of Chemistry, Lomonosov Moscow State University, Leninskie Gory 1, 119234 Moscow, Russia; (A.O.K.); (P.V.S.); (O.A.D.); (I.A.O.)
| | - Alexander S. Tikhomirov
- Gause Institute of New Antibiotics, B. Pirogovskaya 11, 119021 Moscow, Russia; (E.N.B.); (A.S.T.); (G.V.Z.); (S.E.S.); (M.M.M.); (N.E.G.); (A.E.S.)
| | - George V. Zatonsky
- Gause Institute of New Antibiotics, B. Pirogovskaya 11, 119021 Moscow, Russia; (E.N.B.); (A.S.T.); (G.V.Z.); (S.E.S.); (M.M.M.); (N.E.G.); (A.E.S.)
| | - Svetlana E. Solovieva
- Gause Institute of New Antibiotics, B. Pirogovskaya 11, 119021 Moscow, Russia; (E.N.B.); (A.S.T.); (G.V.Z.); (S.E.S.); (M.M.M.); (N.E.G.); (A.E.S.)
| | - Maksim M. Martynov
- Gause Institute of New Antibiotics, B. Pirogovskaya 11, 119021 Moscow, Russia; (E.N.B.); (A.S.T.); (G.V.Z.); (S.E.S.); (M.M.M.); (N.E.G.); (A.E.S.)
| | - Natalia E. Grammatikova
- Gause Institute of New Antibiotics, B. Pirogovskaya 11, 119021 Moscow, Russia; (E.N.B.); (A.S.T.); (G.V.Z.); (S.E.S.); (M.M.M.); (N.E.G.); (A.E.S.)
| | - Andrey G. Tereshchenkov
- Department of Chemistry, Lomonosov Moscow State University, Leninskie Gory 1, 119234 Moscow, Russia; (A.O.K.); (P.V.S.); (O.A.D.); (I.A.O.)
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Leninskie Gory 1, 119234 Moscow, Russia
| | - Alena Paleskava
- Department of Molecular and Radiation Biophysics, Petersburg Nuclear Physics Institute Named by B.P. Konstantiniv of NRC “Kurchatov Institute”, Mkr. Orlova Roshcha 1, 188300 Gatchina, Russia; (A.P.); (A.L.K.)
- Institute of Biomedical Systems and Biotechnologies, Peter the Great St. Petersburg Polytechnic University, Khlopina 11, 195251 Saint Petersburg, Russia
| | - Andrey L. Konevega
- Department of Molecular and Radiation Biophysics, Petersburg Nuclear Physics Institute Named by B.P. Konstantiniv of NRC “Kurchatov Institute”, Mkr. Orlova Roshcha 1, 188300 Gatchina, Russia; (A.P.); (A.L.K.)
- Institute of Biomedical Systems and Biotechnologies, Peter the Great St. Petersburg Polytechnic University, Khlopina 11, 195251 Saint Petersburg, Russia
- NBICS Center, NRC “Kurchatov Institute”, Kurchatov Square 1, 123182 Moscow, Russia
| | - Petr V. Sergiev
- Department of Chemistry, Lomonosov Moscow State University, Leninskie Gory 1, 119234 Moscow, Russia; (A.O.K.); (P.V.S.); (O.A.D.); (I.A.O.)
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Leninskie Gory 1, 119234 Moscow, Russia
- Institute of Functional Genomics, Lomonosov Moscow State University, Leninskie Gory 1, 119234 Moscow, Russia
| | - Olga A. Dontsova
- Department of Chemistry, Lomonosov Moscow State University, Leninskie Gory 1, 119234 Moscow, Russia; (A.O.K.); (P.V.S.); (O.A.D.); (I.A.O.)
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Leninskie Gory 1, 119234 Moscow, Russia
- Department of Functioning of Living Systems, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| | - Ilya A. Osterman
- Department of Chemistry, Lomonosov Moscow State University, Leninskie Gory 1, 119234 Moscow, Russia; (A.O.K.); (P.V.S.); (O.A.D.); (I.A.O.)
| | - Andrey E. Shchekotikhin
- Gause Institute of New Antibiotics, B. Pirogovskaya 11, 119021 Moscow, Russia; (E.N.B.); (A.S.T.); (G.V.Z.); (S.E.S.); (M.M.M.); (N.E.G.); (A.E.S.)
| | - Anna N. Tevyashova
- Gause Institute of New Antibiotics, B. Pirogovskaya 11, 119021 Moscow, Russia; (E.N.B.); (A.S.T.); (G.V.Z.); (S.E.S.); (M.M.M.); (N.E.G.); (A.E.S.)
- School of Science, Constructor University, Campus Ring 1, 28759 Bremen, Germany
| |
Collapse
|
23
|
Prasher P, Sharma M. Hybridization of antimicrobial oxazolidinones with commercial drugs: A fight against the "superbugs". Drug Dev Res 2023; 84:1337-1345. [PMID: 37583273 DOI: 10.1002/ddr.22107] [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: 06/03/2023] [Revised: 07/16/2023] [Accepted: 08/05/2023] [Indexed: 08/17/2023]
Abstract
Antimicrobial resistance caused by the emergence of antibiotic-resistant microbes, termed as "superbugs," poses a grave healthcare concern in the contemporary era. Though this phenomenon is natural, an incessant use of antibiotics due to their unregulated over-the-counter availability, and a lack of compliance with the legislation seem to be major contributing factors. This phenomenon has further complicated the treatment of common infectious diseases thereby leading to prolonged illness, disability, and even death. In addition, a sizeable impact on the healthcare cost is met due to a prolonged stay at the medical facilities to receive an intensive care. Overall, the gains of "Millennium Development Goals" and the accomplishment of Sustainable Development Goals are at risk due to the emerging antimicrobial resistance. Since an early identification and development of novel antibiotic classes that evade antimicrobial resistance appears improbable, the strategy of hybridization of the existing antibiotics with efficacious pharmacophores and drug molecules with a different mechanism of antimicrobial action can be a silver lining for the management of superbugs. In this regard, we aim to provide a perspective for the applicability of the hybridization of oxazolidinone class of antibiotics with other drugs for evading antimicrobial resistance.
Collapse
Affiliation(s)
- Parteek Prasher
- Department of Chemistry, University of Petroleum & Energy Studies, Energy Acres, Dehradun, India
| | - Mousmee Sharma
- Department of Chemistry, Uttaranchal University, Dehradun, India
| |
Collapse
|
24
|
Feng J, Zheng Y, Ma W, Ihsan A, Hao H, Cheng G, Wang X. Multitarget antibacterial drugs: An effective strategy to combat bacterial resistance. Pharmacol Ther 2023; 252:108550. [PMID: 39492518 DOI: 10.1016/j.pharmthera.2023.108550] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 10/19/2023] [Accepted: 10/23/2023] [Indexed: 11/05/2024]
Abstract
The rise of antibiotic resistance and the decrease in the discovery of new antibiotics have caused a global health crisis. Of particular concern is the fact that despite efforts to develop new antibiotics, drug discovery is unable to keep up with the rapid development of resistance. This ongoing crisis highlights the fact that single-target drugs may not always exhibit satisfactory therapeutic effects and are prone to target mutations and resistance due to the complexity of bacterial mechanisms. Retrospective studies have shown that most successful antibiotics have multiple targets. Compared with single-target drugs, successfully designed multitarget drugs can simultaneously regulate multiple targets to reduce resistance caused by single-target mutations or expression changes. In addition to a lower risk of drug-drug interactions, multitarget drugs show superior pharmacokinetics and higher patient compliance compared with combination therapies. Therefore, to reduce resistance, many efforts have been made to discover and design multitarget drugs with different chemical structures and functions. Although there have been numerous studies on how to develop drugs and slow down the development of drug resistance, the reduction of bacterial resistance by multitarget antibacterial drugs has not received widespread attention and is rarely mentioned in the peer-reviewed literature. This review summarises the development of antibiotic resistance and the mechanisms proposed for its emergence, examines the potential of multitarget drugs as an effective strategy to slow the development of resistance, and discusses the rationale for multitarget drug therapy. We also describe multitarget antibacterial compounds with the potential to reduce drug resistance and the available strategies to develop multitarget drugs.
Collapse
Affiliation(s)
- Jin Feng
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Youle Zheng
- MAO Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Wanqing Ma
- MAO Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Awais Ihsan
- Department of Biosciences, COMSATS University Islamabad, Sahiwal Campus, Islamabad 45550, Pakistan
| | - Haihong Hao
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China; MAO Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Guyue Cheng
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China; MAO Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Xu Wang
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China; MAO Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei 430070, China.
| |
Collapse
|
25
|
Aziz M, Ejaz SA, Alsfouk BA, Sultan A, Li C. Identification of potential inhibitors against E.coli via novel approaches based on deep learning and quantum mechanics-based atomistic investigations. Arch Biochem Biophys 2023; 747:109761. [PMID: 37734644 DOI: 10.1016/j.abb.2023.109761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/31/2023] [Accepted: 09/18/2023] [Indexed: 09/23/2023]
Abstract
Currently, drug resistance to commercially available antibiotics is imparting negative consequences to global health, and the development of novel antibiotics in a timely manner is a prime need of the hour. In the current study, an e-pharmacophore model was built using the 3D structure of DNA gyrase in complex with a standard inhibitor. The generated model was subjected to a pharmacophore based virtual screening against 45,257,086 molecules having 223,460,579 conformers available in MCULE database. Pharmacophore based screening retrieved eight molecules as top hit based on pharmacophoric features in comparison to standard inhibitors. Afterward, all eight compounds were subjected molecular docking based on deep learning algorithm. The molecular docking revealed that compound MCULE-6042843173 and MCULE-2362244223 had significant binding orientation inside active pocket of targeted protein with binding affinity of -9.52 and -9.24 kcal/mol respectively. In addition, density functional theory studies (DFT) were performed to evaluate quantum mechanics of top ranked compounds which were investigated through quantum mechanics (QM) computations which strongly assisted the findings of other in-silico investigations. Consequently, the MCULE-6042843173 and MCULE-2362244223 were subjected to MD simulation studies for evaluation of stability, hydrogen bond analysis, van der Waals interactions, and the contact profile of compounds with targeted amino acid residues. Findings of current study suggested MCULE-6042843173 and MCULE-2362244223 as potential and novel inhibitor of DNA Gyrase enzyme.
Collapse
Affiliation(s)
- Mubashir Aziz
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, 63100, Pakistan
| | - Syeda Abida Ejaz
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, 63100, Pakistan.
| | - Bshra A Alsfouk
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah Bint Abdulrahman University, P.O Box 84428, Riyadh, 11671, Saudi Arabia
| | - Ahlam Sultan
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah Bint Abdulrahman University, P.O Box 84428, Riyadh, 11671, Saudi Arabia
| | - Chen Li
- Department of Biology, Chemistry, Pharmacy, Free University of Berlin, Berlin, 14195, Germany.
| |
Collapse
|
26
|
Grigor’eva AE, Bardasheva AV, Ryabova ES, Tupitsyna AV, Zadvornykh DA, Koroleva LS, Silnikov VN, Tikunova NV, Ryabchikova EI. Changes in the Ultrastructure of Staphylococcus aureus Cells Make It Possible to Identify and Analyze the Injuring Effects of Ciprofloxacin, Polycationic Amphiphile and Their Hybrid. Microorganisms 2023; 11:2192. [PMID: 37764036 PMCID: PMC10537381 DOI: 10.3390/microorganisms11092192] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/18/2023] [Accepted: 08/28/2023] [Indexed: 09/29/2023] Open
Abstract
The purposeful development of synthetic antibacterial compounds requires an understanding of the relationship between effects of compounds and their chemical structure. This knowledge can be obtained by studying changes in bacteria ultrastructure under the action of antibacterial compounds of a certain chemical structure. Our study was aimed at examination of ultrastructural changes in S. aureus cells caused by polycationic amphiphile based on 1,4‒diazabicyclo[2.2.2]octane (DL412), ciprofloxacin and their hybrid (DL5Cip6); the samples were incubated for 15 and 45 min. DL412 first directly interacted with bacterial cell wall, damaging it, then penetrated into the cell and disrupted cytoplasm. Ciprofloxacin penetrated into cell without visually damaging the cell wall, but altered the cell membrane and cytoplasm, and inhibited the division of bacteria. The ultrastructural characteristics of S. aureus cells damaged by the hybrid clearly differed from those under ciprofloxacin or DL412 action. Signs associated with ciprofloxacin predominated in cell damage patterns from the hybrid. We studied the effect of ciprofloxacin, DL412 and their hybrid on S. aureus biofilm morphology using paraffin sections. Clear differences in compound effects on S. aureus biofilm (45 min incubation) were observed. The results obtained allow us to recommend this simple and cheap approach for the initial assessment of antibiofilm properties of synthesized compounds.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Elena I. Ryabchikova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of Russian Academy of Science, Lavrent’ev av., 8, 630090 Novosibirsk, Russia; (A.E.G.); (A.V.B.); (E.S.R.); (A.V.T.); (D.A.Z.); (L.S.K.); (V.N.S.); (N.V.T.)
| |
Collapse
|
27
|
Kim KS. Editorial: Global excellence in pharmacology of infectious diseases: Australia and Asia. Front Pharmacol 2023; 14:1243284. [PMID: 37521482 PMCID: PMC10374358 DOI: 10.3389/fphar.2023.1243284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 07/07/2023] [Indexed: 08/01/2023] Open
Affiliation(s)
- Kwang-sun Kim
- Department of Chemistry, Chemistry Institute for Functional Materials, Pusan National University, Busan, Republic of Korea
| |
Collapse
|
28
|
Gulyás D, Kamotsay K, Szabó D, Kocsis B. Investigation of Delafloxacin Resistance in Multidrug-Resistant Escherichia coli Strains and the Detection of E. coli ST43 International High-Risk Clone. Microorganisms 2023; 11:1602. [PMID: 37375104 DOI: 10.3390/microorganisms11061602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/13/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
Delafloxacin is a novel fluoroquinolone agent that is approved for clinical application. In this study, we analyzed the antibacterial efficacy of delafloxacin in a collection of 47 Escherichia coli strains. Antimicrobial susceptibility testing was performed by the broth microdilution method and minimum inhibitory concentration (MIC) values were determined for delafloxacin, ciprofloxacin, levofloxacin, moxifloxacin, ceftazidime, cefotaxime, and imipenem. Two multidrug-resistant E. coli strains, which exhibited delafloxacin and ciprofloxacin resistance as well as extended-spectrum beta-lactamase (ESBL) phenotype, were selected for whole-genome sequencing (WGS). In our study, delafloxacin and ciprofloxacin resistance rates were 47% (22/47) and 51% (24/47), respectively. In the strain collection, 46 E. coli were associated with ESBL production. The MIC50 value for delafloxacin was 0.125 mg/L, while all other fluoroquinolones had an MIC50 value of 0.25 mg/L in our collection. Delafloxacin susceptibility was detected in 20 ESBL positive and ciprofloxacin resistant E. coli strains; by contrast, E. coli strains that exhibited a ciprofloxacin MIC value above 1 mg/L were delafloxacin-resistant. WGS analysis on the two selected E. coli strains (920/1 and 951/2) demonstrated that delafloxacin resistance is mediated by multiple chromosomal mutations, namely, five mutations in E. coli 920/1 (gyrA S83L, D87N, parC S80I, E84V, and parE I529L) and four mutations in E. coli 951/2 (gyrA S83L, D87N, parC S80I, and E84V). Both strains carried an ESBL gene, blaCTX-M-1 in E. coli 920/1 and blaCTX-M-15 in E. coli 951/2. Based on multilocus sequence typing, both strains belong to the E. coli sequence type 43 (ST43). In this paper, we report a remarkable high rate (47%) of delafloxacin resistance among multidrug-resistant E. coli as well as the E. coli ST43 international high-risk clone in Hungary.
Collapse
Affiliation(s)
- Dániel Gulyás
- Institute of Medical Microbiology, Semmelweis University, 1089 Budapest, Hungary
| | - Katalin Kamotsay
- Central Microbiology Laboratory, National Institute of Hematology and Infectious Disease, Central Hospital of Southern-Pest, 1097 Budapest, Hungary
| | - Dóra Szabó
- Institute of Medical Microbiology, Semmelweis University, 1089 Budapest, Hungary
- Human Microbiota Study Group, Semmelweis University-Eötvös Lóránd Research Network, 1089 Budapest, Hungary
| | - Béla Kocsis
- Institute of Medical Microbiology, Semmelweis University, 1089 Budapest, Hungary
| |
Collapse
|
29
|
Koh AJJ, Thombare V, Hussein M, Rao GG, Li J, Velkov T. Bifunctional antibiotic hybrids: A review of clinical candidates. Front Pharmacol 2023; 14:1158152. [PMID: 37397488 PMCID: PMC10313405 DOI: 10.3389/fphar.2023.1158152] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 06/02/2023] [Indexed: 07/04/2023] Open
Abstract
Antibiotic resistance is a top threat to human health and a priority across the globe. This problematic issue is accompanied by the decline of new antibiotics in the pipeline over the past 30 years. In this context, an urgent need to develop new strategies to combat antimicrobial resistance is in great demand. Lately, among the possible approaches used to deal with antimicrobial resistance is the covalent ligation of two antibiotic pharmacophores that target the bacterial cells through a dissimilar mode of action into a single hybrid molecule, namely hybrid antibiotics. This strategy exhibits several advantages, including better antibacterial activity, overcoming the existing resistance towards individual antibiotics, and may ultimately delay the onset of bacterial resistance. This review sheds light on the latest development of the dual antibiotic hybrids pipeline, their potential mechanisms of action, and challenges in their use.
Collapse
Affiliation(s)
- Augustine Jing Jie Koh
- Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIP, Australia
| | - Varsha Thombare
- Monash Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Parkville, VIP, Australia
| | - Maytham Hussein
- Monash Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Parkville, VIP, Australia
| | - Gauri G. Rao
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, United States
| | - Jian Li
- Monash Biomedicine Discovery Institute, Department of Microbiology, Monash University, Parkville, VIP, Australia
| | - Tony Velkov
- Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIP, Australia
- Monash Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Parkville, VIP, Australia
| |
Collapse
|
30
|
Overview of Side-Effects of Antibacterial Fluoroquinolones: New Drugs versus Old Drugs, a Step Forward in the Safety Profile? Pharmaceutics 2023; 15:pharmaceutics15030804. [PMID: 36986665 PMCID: PMC10056716 DOI: 10.3390/pharmaceutics15030804] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 02/07/2023] [Accepted: 02/17/2023] [Indexed: 03/05/2023] Open
Abstract
Antibacterial fluoroquinolones (FQs) are frequently used in treating infections. However, the value of FQs is debatable due to their association with severe adverse effects (AEs). The Food and Drug Administration (FDA) issued safety warnings concerning their side-effects in 2008, followed by the European Medicine Agency (EMA) and regulatory authorities from other countries. Severe AEs associated with some FQs have been reported, leading to their withdrawal from the market. New systemic FQs have been recently approved. The FDA and EMA approved delafloxacin. Additionally, lascufloxacin, levonadifloxacin, nemonoxacin, sitafloxacin, and zabofloxacin were approved in their origin countries. The relevant AEs of FQs and their mechanisms of occurrence have been approached. New systemic FQs present potent antibacterial activity against many resistant bacteria (including resistance to FQs). Generally, in clinical studies, the new FQs were well-tolerated with mild or moderate AEs. All the new FQs approved in the origin countries require more clinical studies to meet FDA or EMA requirements. Post-marketing surveillance will confirm or infirm the known safety profile of these new antibacterial drugs. The main AEs of the FQs class were addressed, highlighting the existing data for the recently approved ones. In addition, the general management of AEs when they occur and the rational use and caution of modern FQs were outlined.
Collapse
|
31
|
Gao J, Hou H, Gao F. Current scenario of quinolone hybrids with potential antibacterial activity against ESKAPE pathogens. Eur J Med Chem 2023; 247:115026. [PMID: 36577217 DOI: 10.1016/j.ejmech.2022.115026] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/04/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022]
Abstract
The ESKAPE (Escherichia coli/E. coli, Staphylococcus aureus/S. aureus, Klebsiella pneumonia/K. pneumoniae, Acinetobacter Baumannii/A. baumannii, Pseudomonas aeroginosa/P. aeroginosa and Enterobacter spp.) pathogens, which could escape or evade common therapies through diverse antimicrobial resistance mechanisms and biofilm formation, are deemed as highly virulent bacteria responsible for life-threatening diseases, calling for novel chemotherapeutics. Quinolones including 2-quinolones and 4-quinolones have occupied a propitious place in drug design and development due to their excellent pharmacological profiles. Quinolones especially fluoroquinolones could inhibit the synthesis of nucleic acid of ESKAPE pathogens, leading to the rupture of bacterial chromosome. However, the resistance of ESKAPE pathogens to quinolones develops rapidly and spreads widely. Accordingly, it has become increasingly urgent to enhance the potency of quinolones against both drug-susceptible and drug-resistant ESKAPE pathogens. Quinolone hybrids can bind with different drug targets simultaneously and have been considered as useful prototypes to circumvent drug resistance. The purpose of this review is to summarize the current scenario (2018-present) of quinolone hybrids with potential antibacterial activity against ESKAPE pathogens, together with the structure-activity relationships and mechanisms of action to facilitate further rational design of more effective candidates.
Collapse
Affiliation(s)
- Jingyue Gao
- Key Laboratory for Experimental Teratology of the Ministry of Education and Center for Experimental Nuclear Medicine, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Haodong Hou
- Key Laboratory for Experimental Teratology of the Ministry of Education and Center for Experimental Nuclear Medicine, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Feng Gao
- Key Laboratory for Experimental Teratology of the Ministry of Education and Center for Experimental Nuclear Medicine, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.
| |
Collapse
|
32
|
Chen H, Wu J, Xiong Q, Li X, Huang X. Efficient capture of fluoroquinolones in urine and milk samples with multi-monolith fibers solid phase microextraction based on hybrid metal-organic framework/monolith material. Microchem J 2023. [DOI: 10.1016/j.microc.2023.108575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
|
33
|
Kapusta O, Jarosz A, Stadnik K, Giannakoudakis DA, Barczyński B, Barczak M. Antimicrobial Natural Hydrogels in Biomedicine: Properties, Applications, and Challenges-A Concise Review. Int J Mol Sci 2023; 24:2191. [PMID: 36768513 PMCID: PMC9917233 DOI: 10.3390/ijms24032191] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/18/2023] [Accepted: 01/19/2023] [Indexed: 01/24/2023] Open
Abstract
Natural hydrogels are widely used as biomedical materials in many areas, including drug delivery, tissue scaffolds, and particularly wound dressings, where they can act as an antimicrobial factor lowering the risk of microbial infections, which are serious health problems, especially with respect to wound healing. In this review article, a number of promising strategies in the development of hydrogels with biocidal properties, particularly those originating from natural polymers, are briefly summarized and concisely discussed. Common strategies to design and fabricate hydrogels with intrinsic or stimuli-triggered antibacterial activity are exemplified, and the mechanisms lying behind these properties are also discussed. Finally, practical antibacterial applications are also considered while discussing the current challenges and perspectives.
Collapse
Affiliation(s)
- Oliwia Kapusta
- Institute of Chemical Sciences, Faculty of Chemistry, Maria Curie-Sklodowska University, 20031 Lublin, Poland
| | - Anna Jarosz
- Institute of Chemical Sciences, Faculty of Chemistry, Maria Curie-Sklodowska University, 20031 Lublin, Poland
| | - Katarzyna Stadnik
- Institute of Chemical Sciences, Faculty of Chemistry, Maria Curie-Sklodowska University, 20031 Lublin, Poland
| | | | - Bartłomiej Barczyński
- 1st Department of Oncological Gynecology and Gynecology, Medical University in Lublin, 20-059 Lublin, Poland
| | - Mariusz Barczak
- Institute of Chemical Sciences, Faculty of Chemistry, Maria Curie-Sklodowska University, 20031 Lublin, Poland
| |
Collapse
|
34
|
Wood SJ, Kuzel TM, Shafikhani SH. Pseudomonas aeruginosa: Infections, Animal Modeling, and Therapeutics. Cells 2023; 12:199. [PMID: 36611992 PMCID: PMC9818774 DOI: 10.3390/cells12010199] [Citation(s) in RCA: 79] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 01/05/2023] Open
Abstract
Pseudomonas aeruginosa is an important Gram-negative opportunistic pathogen which causes many severe acute and chronic infections with high morbidity, and mortality rates as high as 40%. What makes P. aeruginosa a particularly challenging pathogen is its high intrinsic and acquired resistance to many of the available antibiotics. In this review, we review the important acute and chronic infections caused by this pathogen. We next discuss various animal models which have been developed to evaluate P. aeruginosa pathogenesis and assess therapeutics against this pathogen. Next, we review current treatments (antibiotics and vaccines) and provide an overview of their efficacies and their limitations. Finally, we highlight exciting literature on novel antibiotic-free strategies to control P. aeruginosa infections.
Collapse
Affiliation(s)
- Stephen J. Wood
- Department of Medicine, Division of Hematology, Oncology, & Cell Therapy, Rush University Medical Center, Chicago, IL 60612, USA
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL 60612, USA
| | - Timothy M. Kuzel
- Department of Medicine, Division of Hematology, Oncology, & Cell Therapy, Rush University Medical Center, Chicago, IL 60612, USA
- Cancer Center, Rush University Medical Center, Chicago, IL 60612, USA
| | - Sasha H. Shafikhani
- Department of Medicine, Division of Hematology, Oncology, & Cell Therapy, Rush University Medical Center, Chicago, IL 60612, USA
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL 60612, USA
- Cancer Center, Rush University Medical Center, Chicago, IL 60612, USA
| |
Collapse
|
35
|
Qian R, Xu Z, Hu X, Liu S, Mai Y, Tan X, Su X, Jiang M, Tang W, Tian W, Xie L. Ag/Ag 2O with NIR-Triggered Antibacterial Activities: Photocatalytic Sterilization Enhanced by Low-Temperature Photothermal Effect. Int J Nanomedicine 2023; 18:1507-1520. [PMID: 36998603 PMCID: PMC10046159 DOI: 10.2147/ijn.s400511] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 03/14/2023] [Indexed: 04/01/2023] Open
Abstract
Purpose A synergistic antibacterial system employing photocatalytic performance and low-temperature photothermal effect (LT-PTT) with the potential for infectious skin wound healing promotion was developed. Methods Ag/Ag2O was synthesized with a two-step method, and its physicochemical properties were characterized. After its photocatalytic performance and photothermal effect were evaluated under 0.5 W/cm2 808 nm NIR laser irradiation, its antibacterial activities in both planktonic and biofilm forms were then studied in vitro targeting Staphylococcus Aureus (S. aureus), and the biocompatibility was tested with L-929 cell lines afterward. Finally, the animal model of dorsal skin wound infection was established on Sprague-Dawley rats and was used to assess infectious wound healing promotion of Ag/Ag2O in vivo. Results Ag/Ag2O showed boosted photocatalytic performance and local temperature accumulation compared with Ag2O when exposed to 0.5 W/cm2 808 nm NIR irradiation, which therefore endowed Ag/Ag2O with the ability to kill pathogens rapidly and cleavage bacterial biofilm in vitro. Furthermore, after treatment with Ag/Ag2O and 0.5 W/cm2 808 nm NIR irradiation, infectious wounds of rats realized skin tissue regeneration from a histochemical level. Conclusion By exhibiting excellent NIR-triggered photocatalytic sterilization ability enhanced by low-temperature photothermal effect, Ag/Ag2O was promising to be a novel, photo-responsive antibacterial agent.
Collapse
Affiliation(s)
- Ruojing Qian
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, People’s Republic of China
- Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu, People’s Republic of China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, People’s Republic of China
| | - Zhaoyu Xu
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, People’s Republic of China
| | - Xingyu Hu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, People’s Republic of China
- Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu, People’s Republic of China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, People’s Republic of China
| | - Suru Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, People’s Republic of China
- Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu, People’s Republic of China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, People’s Republic of China
| | - Yao Mai
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, People’s Republic of China
- Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu, People’s Republic of China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, People’s Republic of China
| | - Xinzhi Tan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, People’s Republic of China
- Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu, People’s Republic of China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, People’s Republic of China
| | - Xiaofan Su
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, People’s Republic of China
- Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu, People’s Republic of China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, People’s Republic of China
| | - Mingyan Jiang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, People’s Republic of China
- Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu, People’s Republic of China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, People’s Republic of China
| | - Wei Tang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, People’s Republic of China
- Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu, People’s Republic of China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, People’s Republic of China
| | - Weidong Tian
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, People’s Republic of China
- Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu, People’s Republic of China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, People’s Republic of China
- Correspondence: Weidong Tian; Li Xie, State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China, Tel +86-28-85502156; +86-28-85503499, Email ;
| | - Li Xie
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, People’s Republic of China
- Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu, People’s Republic of China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, People’s Republic of China
| |
Collapse
|