1
|
Li T, Zhang R, Gong H, Tang Z, Li X, Gong Z, Challa M, Zou C, Zhang SL, Guo J, He Y. Synthesis and antibacterial evaluations of novel vancomycin analogues targeting bacteria membrane to combat Gram-negative infections. Eur J Med Chem 2025; 289:117483. [PMID: 40056801 DOI: 10.1016/j.ejmech.2025.117483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/20/2025] [Accepted: 03/04/2025] [Indexed: 03/10/2025]
Abstract
Vancomycin is primarily used to treat severe infections caused by Gram-positive bacteria and is often considered as the last-resort therapy in the life-threatening situation. However, it is inherently ineffective against Gram-negative bacteria. Herein, we report the design, synthesis, and biological evaluation of novel vancomycin analogues incorporated with lipophilic cationic groups. Through structural optimization and structure-activity relationship (SAR) studies, we identified vancomycin analogue 18b, which exhibited remarkable antibacterial activity against A. baumannii ATCC 17978, with a MIC of 8 μg/mL. In contrast, vancomycin showed no activity against this strain, even at concentration as high as 128 μg/mL. Further investigations revealed that 18b possesses rapid bactericidal properties, low toxicity, and a reduced propensity to induce bacterial resistance. The exceptional antibacterial performance of 18b is partially attributed to the presence of membrane-targeting, lipophilic piperazine cationic groups. In a mouse model infected with A. baumannii ATCC 17978, 18b exhibited excellent efficacy at a dose of 20 mg/kg, while no toxicity was observed. These findings highlight 18b as a promising candidate for further development in the fight against Gram-negative bacterial infections.
Collapse
Affiliation(s)
- Tao Li
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, Shuitu Technology Development Zone, No. 55 Daxuecheng South Rd., Shapingba, Chongqing, 401331, PR China
| | - Ruixue Zhang
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, Shuitu Technology Development Zone, No. 55 Daxuecheng South Rd., Shapingba, Chongqing, 401331, PR China
| | - Hongzhi Gong
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, Shuitu Technology Development Zone, No. 55 Daxuecheng South Rd., Shapingba, Chongqing, 401331, PR China
| | - Ziyi Tang
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, 266 Fangzheng Ave, Beibei, Chongqing, 400714, PR China; Chongqing Institute for Food and Drug Control, Chongqing, 401120, PR China
| | - Xinyu Li
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, Shuitu Technology Development Zone, No. 55 Daxuecheng South Rd., Shapingba, Chongqing, 401331, PR China
| | - Zhi Gong
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, Shuitu Technology Development Zone, No. 55 Daxuecheng South Rd., Shapingba, Chongqing, 401331, PR China
| | - Mahesh Challa
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, Shuitu Technology Development Zone, No. 55 Daxuecheng South Rd., Shapingba, Chongqing, 401331, PR China
| | - Cheng Zou
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, Shuitu Technology Development Zone, No. 55 Daxuecheng South Rd., Shapingba, Chongqing, 401331, PR China
| | - Shao-Lin Zhang
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, Shuitu Technology Development Zone, No. 55 Daxuecheng South Rd., Shapingba, Chongqing, 401331, PR China.
| | - Jian Guo
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, Shuitu Technology Development Zone, No. 55 Daxuecheng South Rd., Shapingba, Chongqing, 401331, PR China.
| | - Yun He
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, Shuitu Technology Development Zone, No. 55 Daxuecheng South Rd., Shapingba, Chongqing, 401331, PR China; BayRay Innovation Center, Shenzhen Bay Laboratory, Shenzhen, 518132, PR China.
| |
Collapse
|
2
|
Li T, Tang Z, Zhang R, Challa M, Gong H, Gong Z, Zhang SL, Guo J, He Y. Targeted vancomycin delivery via in situ albumin conjugation and acid-triggered drug release for reduced nephrotoxicity. Eur J Med Chem 2025; 291:117652. [PMID: 40262299 DOI: 10.1016/j.ejmech.2025.117652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Revised: 04/10/2025] [Accepted: 04/16/2025] [Indexed: 04/24/2025]
Abstract
Vancomycin has long been considered as the last-resort antibiotic for tacking extremely severe infections caused by methicillin-resistant Staphylococcus aureus (MRSA). However, its clinical application is limited by dose-limiting nephrotoxicity. In this study, we report a novel in situ albumin conjugation and acid sensitive prodrug strategy to selectively release vancomycin at the infection site, thereby minimizing the accumulation of vancomycin in the kidney and thus reducing its nephrotoxicity. We synthesized and evaluated four vancomycin prodrugs 13a-d and found that 13c effectively bound to plasma albumin in vitro, and released vancomycin rapidly at the infection site. Its therapeutic effect against MRSA USA300 infection was comparable to that of free vancomycin at 10 mg/kg. In vivo safety assessments demonstrated that 13c did not exhibit significant nephrotoxicity at 50 mg/kg, whereas vancomycin caused obvious nephrotoxicity at the same dose. This work represents the first example of utilizing albumin for targeted delivery of antibiotic to the bacterial infection site to mitigate the common dose-limiting nephrotoxicity of vancomycin, and this strategy may also be applicable to other aminoglycoside antibiotics with nephrotoxicity.
Collapse
Affiliation(s)
- Tao Li
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, No. 55 Daxuecheng South Rd., Shapingba, Chongqing, 401331, PR China
| | - Ziyi Tang
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, 266 Fangzheng Ave, Shuitu Technology Development Zone, Beibei, Chongqing, 400714, PR China; Chongqing Institute for Food and Drug Control, Chongqing, 401120, PR China
| | - Ruixue Zhang
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, No. 55 Daxuecheng South Rd., Shapingba, Chongqing, 401331, PR China
| | - Mahesh Challa
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, No. 55 Daxuecheng South Rd., Shapingba, Chongqing, 401331, PR China
| | - Hongzhi Gong
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, No. 55 Daxuecheng South Rd., Shapingba, Chongqing, 401331, PR China
| | - Zhi Gong
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, No. 55 Daxuecheng South Rd., Shapingba, Chongqing, 401331, PR China
| | - Shao-Lin Zhang
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, No. 55 Daxuecheng South Rd., Shapingba, Chongqing, 401331, PR China.
| | - Jian Guo
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, No. 55 Daxuecheng South Rd., Shapingba, Chongqing, 401331, PR China.
| | - Yun He
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, No. 55 Daxuecheng South Rd., Shapingba, Chongqing, 401331, PR China; Therapeutic Innovation Center, Shenzhen Bay Laboratory, Shenzhen, 518132, PR China.
| |
Collapse
|
3
|
Kumar SD, Park J, Radhakrishnan NK, Aryal YP, Jeong GH, Pyo IH, Ganbaatar B, Lee CW, Yang S, Shin Y, Subramaniyam S, Lim YJ, Kim SH, Lee S, Shin SY, Cho SJ. Novel Leech Antimicrobial Peptides, Hirunipins: Real-Time 3D Monitoring of Antimicrobial and Antibiofilm Mechanisms Using Optical Diffraction Tomography. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409803. [PMID: 39792785 PMCID: PMC11905058 DOI: 10.1002/advs.202409803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 12/13/2024] [Indexed: 01/12/2025]
Abstract
Antimicrobial peptides (AMPs) are promising agents for treating antibiotic-resistant bacterial infections. Although discovering novel AMPs is crucial for combating multidrug-resistant bacteria and biofilm-related infections, their clinical potential relies on precise, real-time evaluation of efficacy, toxicity, and mechanisms. Optical diffraction tomography (ODT), a label-free imaging technology, enables real-time visualization of bacterial morphological changes, membrane damage, and biofilm formation over time. Here, a computational analysis of the leech transcriptome using an advanced AI-based peptide screening strategy with ODT to identify potential AMPs is employed. Among the 19 potential AMPs identified, hirunipin 2 demonstrates potent antibacterial activity, low mammalian cytotoxicity, and minimal hemolytic effects. It demonstrates efficacy comparable to melittin, resistance to physiological salts and human serum, and a low likelihood of inducing bacterial resistance. Microscopy and 3D-ODT confirm its disruption of bacterial membranes and intracellular aggregation, leading to cell death. Notably, hirunipin 2 effectively inhibits biofilm formation, eradicates preformed biofilms, and synergizes with antibiotics against multidrug-resistant Acinetobacter baumannii (MDRAB) by enhancing membrane permeability. Additionally, hirunipin 2 significantly suppresses pro-inflammatory cytokine expression in LPS-stimulated macrophages, highlighting its anti-inflammatory properties. These findings highlight hirunipin 2 as a strong candidate for developing novel antibacterial, anti-inflammatory, and antibiofilm therapies, particularly against multidrug-resistant bacterial infections.
Collapse
Affiliation(s)
- S Dinesh Kumar
- Department of Cellular & Molecular Medicine, School of Medicine, Chosun University, Gwangju, 61452, Republic of Korea
| | - Jeongwon Park
- Gwangju Center, Korea Basic Science Institute (KBSI), Gwangju, 61751, Republic of Korea
- Department of Animal Science, Chonnam National University, Gwangju, 61186, South Korea
| | - Naveen Kumar Radhakrishnan
- Department of Biomedical Sciences, Graduate School, Chosun University, Gwangju, 61452, Republic of Korea
| | - Yam Prasad Aryal
- Department of Biological Sciences and Biotechnology, College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea
| | - Geon-Hwi Jeong
- Department of Biological Sciences and Biotechnology, College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea
| | - In-Hyeok Pyo
- Department of Biological Sciences and Biotechnology, College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea
| | - Byambasuren Ganbaatar
- Department of Chemistry, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Chul Won Lee
- Department of Chemistry, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Sungtae Yang
- Institute of Well-Aging Medicare & CSU G-LAMP Project Group, Chosun University, Gwangju, 61452, Republic of Korea
| | - Younhee Shin
- Research and Development Center, Insilicogen Inc, Yongin-si, Gyeonggi-do, 16954, Republic of Korea
| | | | - Yu-Jin Lim
- Research and Development Center, Insilicogen Inc, Yongin-si, Gyeonggi-do, 16954, Republic of Korea
| | - Sung-Hak Kim
- Department of Animal Science, Chonnam National University, Gwangju, 61186, South Korea
| | - Seongsoo Lee
- Gwangju Center, Korea Basic Science Institute (KBSI), Gwangju, 61751, Republic of Korea
- Department of Bio-Analysis Science, University of Science & Technology, Daejeon, 34113, Republic of Korea
- Department of Systems Biotechnology, Chung-Ang University, Anseong, 17546, Republic of Korea
- Department of Life Science, Hanyang University, Seoul, 04763, Republic of Korea
| | - Song Yub Shin
- Department of Cellular & Molecular Medicine, School of Medicine, Chosun University, Gwangju, 61452, Republic of Korea
| | - Sung-Jin Cho
- Department of Biological Sciences and Biotechnology, College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea
| |
Collapse
|
4
|
Tammam MA, Pereira F, Skellam E, Bidula S, Ganesan A, El-Demerdash A. The cytochalasans: potent fungal natural products with application from bench to bedside. Nat Prod Rep 2025. [PMID: 39989362 DOI: 10.1039/d4np00076e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Covering: 2000-2023Cytochalasans are a fascinating class of natural products that possess an intricate chemical structure with a diverse range of biological activities. They are known for their complex chemical architectures and are often isolated from various fungi. These compounds have attracted attention due to their potential pharmacological properties, including antimicrobial, antiviral, and anticancer effects. For decades, researchers have studied these molecules to better understand their mechanisms of action and to explore their potential applications in medicine and other fields. This review article aims to shed light over the period 2000-2023 on the structural diversities of 424 fungal derived cytochalasans, insights into their biosynthetic origins, pharmacokinetics and their promising therapeutic potential in drug discovery and development.
Collapse
Affiliation(s)
- Mohamed A Tammam
- Department of Biochemistry, Faculty of Agriculture, Fayoum University, Fayoum 63514, Egypt
| | - Florbela Pereira
- LAQV REQUIMTE, Department of Chemistry, NOVA School of Science and Technology, Universidade Nova de Lisboa, 2829516 Caparica, Portugal
| | - Elizabeth Skellam
- Department of Chemistry and BioDiscovery Institute, University of North Texas, 1155 Union Circle, Denton, TX, 76201, USA
| | - Stefan Bidula
- School of Chemistry, Pharmacy and Pharmacology, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK.
| | - A Ganesan
- School of Chemistry, Pharmacy and Pharmacology, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK.
| | - Amr El-Demerdash
- School of Chemistry, Pharmacy and Pharmacology, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK.
- Division of Organic Chemistry, Department of Chemistry, Faculty of Sciences, Mansoura University, Mansoura 35516, Egypt
- Department of Biochemistry and Metabolism, The John Innes Centre, Norwich Research Park, Norwich NR4 7UH, UK.
| |
Collapse
|
5
|
Yang H, Li J, Wang BL, Yang XY, Zhang Y. Discovery, synthesis, and antibacterial activity of novel myrtucommulone analogs as inhibitors of DNA gyrase and topoisomerase IV. Eur J Med Chem 2025; 283:117138. [PMID: 39662286 DOI: 10.1016/j.ejmech.2024.117138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/10/2024] [Accepted: 11/24/2024] [Indexed: 12/13/2024]
Abstract
Drug-resistant bacterial infections have emerged as a new challenge in anti-infective treatment, posing a significant threat to public health. DNA gyrase and topoisomerase IV (Topo IV) are promising targets for designing new antibiotics. Myrtus communis L. has long been used as a traditional herb for antisepsis and disinfection; however, the underlying mechanism of the antibacterial activity remains unclear. In this study, a class of novel myrtucommulone derivatives was synthesized and evaluated for DNA gyrase and Topo IV inhibitions. Analog 27 was the most potent DNA gyrase and Topo IV inhibitor. In bioactivity assays, molecule 27 exhibited a significant antibacterial effect against methicillin-resistant Staphylococcus aureus (MRSA). Additionally, it exhibited rapid bactericidal properties, low toxicity, and low inducing bacterial resistance. It demonstrated synergistic effects with ofloxacin, amikacin, cefepime, and ceftazidime, which make it a potential candidate for antimicrobial application. This work will facilitate the future development of myrtucommulone-based DNA gyrase and Topo IV inhibitors as novel antimicrobials to combat the increasing prevalence of multidrug-resistant bacteria.
Collapse
Affiliation(s)
- Hao Yang
- School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, China
| | - Jian Li
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Bo-Lin Wang
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Xin-Ya Yang
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Yu Zhang
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China.
| |
Collapse
|
6
|
Kaur N, Sahoo J, De M. Development of Nanomaterials-Based Agents for Selective Antibacterial Activity. Chembiochem 2025; 26:e202400693. [PMID: 39632741 DOI: 10.1002/cbic.202400693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 10/26/2024] [Indexed: 12/07/2024]
Abstract
Bacterial infections continue to threaten public health due to limitations in rapid and accurate diagnostic techniques. While broad-spectrum antibiotics offer empirical treatment, their overuse has fuelled the emergence of antimicrobial resistance (AMR) pathogens, posing a critical global public health challenge. In this critical scenario, nanomaterial-based antibacterial agents emerge as a promising solution to combat bacteria and inhibit their proliferation. However, selective elimination of pathogenic bacteria is paramount. This review highlights recent advancements in developing nanomaterials for selective antibacterial activity. We categorize these agents based on their mode of action, exploring how they selectively interact with bacteria and their potential antibacterial mechanisms. This review offers crucial insights for researchers exploring the potential of nanotechnology to address the growing threat of AMR.
Collapse
Affiliation(s)
- Navjot Kaur
- Department of Organic Chemistry, Indian Institute of Science, Bangalore, 560012, India
| | - Jagabandhu Sahoo
- Department of Organic Chemistry, Indian Institute of Science, Bangalore, 560012, India
| | - Mrinmoy De
- Department of Organic Chemistry, Indian Institute of Science, Bangalore, 560012, India
| |
Collapse
|
7
|
Yabbarov NG, Nikolskaya ED, Bibikov SB, Maltsev AA, Chirkina MV, Mollaeva MR, Sokol MB, Epova EY, Aliev RO, Kurochkin IN. Methods for Rapid Evaluation of Microbial Antibiotics Resistance. BIOCHEMISTRY. BIOKHIMIIA 2025; 90:S312-S341. [PMID: 40164164 DOI: 10.1134/s0006297924603678] [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: 08/22/2024] [Revised: 09/02/2024] [Accepted: 09/24/2024] [Indexed: 04/02/2025]
Abstract
Antibiotic resistance is a major challenge for public health systems worldwide. Rapid and effective identification of bacterial strains is critical for reducing the use of antibiotics and restricting the spread of antibiotic-resistant microorganisms. Various approaches have been developed in recent years for rapid bacterial identification and antibiotic susceptibility testing (AST), such as Raman spectroscopy, single cell image analysis, microfluidic techniques, mass spectrometry analysis, use of high-sensitive luminescent and fluorescent tags, impedance-based detection, and others. This review describes the methods developed for rapid bacterial identification and assessment of their antibiotic susceptibility, including general principles, specific problems, and future prospects.
Collapse
Affiliation(s)
- Nikita G Yabbarov
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Moscow, 119334, Russia.
| | - Elena D Nikolskaya
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Moscow, 119334, Russia.
| | - Sergei B Bibikov
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Moscow, 119334, Russia
| | - Aleksandr A Maltsev
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Moscow, 119334, Russia
| | - Margarita V Chirkina
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Moscow, 119334, Russia
| | - Mariia R Mollaeva
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Moscow, 119334, Russia
| | - Maria B Sokol
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Moscow, 119334, Russia
| | - Ekaterina Yu Epova
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Moscow, 119334, Russia
| | - Ruslan O Aliev
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Moscow, 119334, Russia
| | - Ilya N Kurochkin
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Moscow, 119334, Russia
| |
Collapse
|
8
|
Jiang J, Okuda S, Itoh H, Okamoto K, Nakanishi H, Suzuki M, Lu P, Nagata K. Structure-Guided Discovery of a Potent Inhibitor of the Ferric Citrate Binding Protein FecB in Vibrio Bacteria. Angew Chem Int Ed Engl 2024; 63:e202411688. [PMID: 39304960 DOI: 10.1002/anie.202411688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 09/18/2024] [Accepted: 09/20/2024] [Indexed: 09/22/2024]
Abstract
Infections caused by Gram-negative bacteria present a significant risk to human health worldwide. Novel strategies are needed to deal with the challenge caused by drug-resistant bacteria. Here, we report a new approach to combat infections by targeting iron-binding proteins to suppress bacterial growth. We investigated the function of the conserved periplasmic binding protein FecB from Vibrio alginolyticus. FecB was known to play a crucial role in the bacterial growth and to relate with biofilm formation. We then solved the crystal structures and elucidated the binding mechanism of FecB with ferric ion chelated by citrate. The results indicated that FecB binds weakly to one citrate molecule and strongly to the Fe3+-(citrate)2 complex. Based on these results, a structure-based virtual screening approach was conducted against FecB to identify small molecules that block ferric citrate uptake. Further evaluations in vivo and in vitro demonstrated that salvianolic acid C significantly suppressed bacterial growth, indicating that targeting bacterial nutrient absorption is a promising strategy for identifying potential antibacterial drugs.
Collapse
Affiliation(s)
- Jinyan Jiang
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Science, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Suguru Okuda
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Science, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Hideaki Itoh
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Science, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Ken Okamoto
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Science, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Hiromi Nakanishi
- Department of Global Agricultural Sciences, Graduate School of Agricultural and Life Science, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Michio Suzuki
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Science, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Peng Lu
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Science, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
- Future Food Laboratory, Innovation Center of Yangtze River Delta, Zhejiang University, 828, Zhongxing Road, Xitang Town, Jiashan County, Jiaxing City, Zhejiang Province, 314100, China
| | - Koji Nagata
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Science, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
- Agricultural Bioinformatics Research Unit, Graduate School of Agricultural and Life Science, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
- Research Center for Food Safety, Graduate School of Agricultural and Life Science, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| |
Collapse
|
9
|
Wu XY, Liu QY, Jiang S, Pan ZY, Dong JH, Chen BH, Li JH, Liu YS, Liu Y, He L. Copper(II) aromatic heterocyclic complexes of Gatifloxacin with multi-targeting capabilities for antibacterial therapy and combating antibiotic resistance. Bioorg Chem 2024; 153:107938. [PMID: 39520787 DOI: 10.1016/j.bioorg.2024.107938] [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: 07/18/2024] [Revised: 10/20/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024]
Abstract
In recent years, the pace of novel antibiotic development has been relatively slow, intensifying the urgency of the antibiotic resistance issue. Consequently, scientists have turned their attention to enhancing antibiotic activity by coordinating antibiotics with metal elements. This study designs and synthesizes three novel antibacterial copper complexes based on Gatifloxacin. These complexes exhibit potent antibacterial activity, notably Cu-1, with a minimum inhibitory concentration (MIC) of only 0.063 μg/mL against Staphylococcus aureus (S.aureus), demonstrating potent bacteriostatic capabilities. Further investigations unveil the antibacterial mechanisms of complex Cu-1, revealing its ability not only to suppress the activities of DNA gyrase and topoisomerases IV, but also to effectively inhibit biofilm formation and disrupt the integrity of cell membrane. This multi-targeting action contributes to mitigating the risk of bacterial resistance emergence. Additionally, synergy between Cu-1 and conventional antibiotics is confirmed through checkerboard assays, offering novel strategies for antibacterial therapy. In vivo experiments using a murine model of S.aureus infection demonstrate the significant antibacterial efficacy of Cu-1, providing robust support for its potential in treating S.aureus infections. This study demonstrates that the coordination complexes formed by copper, Gatifloxacin and suitable aromatic heterocyclic ligands exhibit multi-targeting characteristics against bacteria, offering a new direction for combating antibiotic resistance in antibacterial therapy.
Collapse
Affiliation(s)
- Xiao-Yin Wu
- Key Laboratory for Biobased Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China
| | - Qi-Yan Liu
- Key Laboratory for Biobased Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China
| | - Shan Jiang
- College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, China
| | - Zheng-Yin Pan
- College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, China
| | - Jia-Hao Dong
- Key Laboratory for Biobased Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China
| | - Bai-Hua Chen
- Key Laboratory for Biobased Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China
| | - Jin-Hao Li
- Key Laboratory for Biobased Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China
| | - Ya-Shu Liu
- Key Laboratory for Biobased Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China
| | - Yingju Liu
- Key Laboratory for Biobased Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China
| | - Liang He
- Key Laboratory for Biobased Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
10
|
Korshoj LE, Kielian T. Bacterial single-cell RNA sequencing captures biofilm transcriptional heterogeneity and differential responses to immune pressure. Nat Commun 2024; 15:10184. [PMID: 39580490 PMCID: PMC11585574 DOI: 10.1038/s41467-024-54581-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 11/14/2024] [Indexed: 11/25/2024] Open
Abstract
Biofilm formation is an important mechanism of survival and persistence for many bacterial pathogens. These multicellular communities contain subpopulations of cells that display metabolic and transcriptional diversity along with recalcitrance to antibiotics and host immune defenses. Here, we present an optimized bacterial single-cell RNA sequencing method, BaSSSh-seq, to study Staphylococcus aureus diversity during biofilm growth and transcriptional adaptations following immune cell exposure. BaSSSh-seq captures extensive transcriptional heterogeneity during biofilm compared to planktonic growth. We quantify and visualize transcriptional regulatory networks across heterogeneous biofilm subpopulations and identify gene sets that are associated with a trajectory from planktonic to biofilm growth. BaSSSh-seq also detects alterations in biofilm metabolism, stress response, and virulence induced by distinct immune cell populations. This work facilitates the exploration of biofilm dynamics at single-cell resolution, unlocking the potential for identifying biofilm adaptations to environmental signals and immune pressure.
Collapse
Affiliation(s)
- Lee E Korshoj
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE, USA.
| | - Tammy Kielian
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
11
|
Yacoub HA, Mahmoud MM, Al-Hejin AM, Abujamel TS, Tabrez S, Abd-Elmaksoud S. Effect of Nk-lysin peptides on bacterial growth, MIC, antimicrobial resistance, and viral activities. Anim Biotechnol 2024; 35:2290520. [PMID: 38100547 DOI: 10.1080/10495398.2023.2290520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2023]
Abstract
NK-lysins from chicken, bovine and human are used as antiviral and antibacterial agents. Gram-negative and gram-positive microorganisms, including Streptococcus pyogenes, Streptococcus mutans, Escherichia coli, Pseudomonas aeruginosa, Klebsiella oxytoca, Shigella sonnei, Klebsiella pneumoniae and Salmonella typhimurium, are susceptible to NK-lysin treatment. The presence of dominant TEM-1 gene was noted in all untreated and treated bacteria, while TOHO-1 gene was absent in all bacteria. Importantly, β-lactamase genes CTX-M-1, CTX-M-8, and CTX-M-9 genes were detected in untreated bacterial strains; however, none of these were found in any bacterial strains following treatment with NK-lysin peptides. NK-lysin peptides are also used to test for inhibition of infectivity, which ranged from 50 to 90% depending on NK-lysin species. Chicken, bo vine and human NK-lysin peptides are demonstrated herein to have antibacterial activity and antiviral activity against Rotavirus (strain SA-11). On the basis of the comparison between these peptides, potent antiviral activity of bovine NK-lysin against Rotavirus (strain SA-11) is particularly evident, inhibiting infection by up to 90%. However, growth was also significantly inhibited by chicken and human NK-lysin peptides, restricted by 80 and 50%, respectively. This study provided a novel treatment using NK-lysin peptides to inhibit expression of β-lactamase genes in β-lactam antibiotic-resistant bacterial infections.
Collapse
Affiliation(s)
- Haitham A Yacoub
- Cell Biology Department, Biotechnology Research Institute, National Research Centre, Cairo, Egypt
| | - Maged Mostafa Mahmoud
- Regerenative Medicine Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia (SA)
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia (SA)
| | - Ahmed M Al-Hejin
- Biological Sciences Department, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Turki S Abujamel
- Regerenative Medicine Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia (SA)
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Shams Tabrez
- Regerenative Medicine Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia (SA)
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia (SA)
| | - Sherif Abd-Elmaksoud
- Environmental Virology Laboratory, Water Pollution Research Department, Environmental Research Institute, National Research Centre, Cairo, Egypt
| |
Collapse
|
12
|
Tang Q, Zhang H, Chandarajoti K, Jiao Z, Nie L, Lv S, Zuo J, Zhou W, Han X. Design and synthesis of coumarin-based amphoteric antimicrobials with biofilm interference and immunoregulation effects. RSC Med Chem 2024:d4md00721b. [PMID: 39802671 PMCID: PMC11712211 DOI: 10.1039/d4md00721b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 10/16/2024] [Indexed: 01/16/2025] Open
Abstract
Bacterial infections pose a threat to human and animal health, and the formation of biofilm exacerbates the microbial threat. New antimicrobial agents to address this challenge are much needed. In this study, several new amphoteric compounds derived from the natural product coumarin were designed and synthesized by mimicking the structure and function of antimicrobial peptides. Strong inhibitory effect of 8b was observed on S. aureus 29213 and five isolated clinically positive strains, with an MIC value of 1-4 μg mL-1, accompanied by the potential advantages of rapid sterilization and no drug resistance. The in vivo activity of 8b was supported by good antibacterial and anti-inflammatory effects in a mouse wound infection model. More importantly, good immunomodulatory effects, inhibition of biofilm formation, and biofilm clearance were detected in the treatment using 8b, which makes it a potential candidate antibacterial for controlling S. aureus infections forming biofilm.
Collapse
Affiliation(s)
- Qun Tang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences 200241 Shanghai China
- Key laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Research Institute, Chinese Academy of Agricultural Sciences Shanghai 200241 China
| | - Haiyang Zhang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences 200241 Shanghai China
| | - Kasemsiri Chandarajoti
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Prince of Songkla University Hat Yai Songkhla 90112 Thailand
- Drug Delivery System Excellence Center, Faculty of Pharmaceutical Sciences, Prince of Songkla University Hat Yai Songkhla 90112 Thailand
| | - Zirui Jiao
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences 200241 Shanghai China
- Key laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Research Institute, Chinese Academy of Agricultural Sciences Shanghai 200241 China
| | - Lianhua Nie
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences 200241 Shanghai China
- College of Engineering, Shanxi Agricultural University Taigu 030801 China
| | - Sai Lv
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences 200241 Shanghai China
- Key laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Research Institute, Chinese Academy of Agricultural Sciences Shanghai 200241 China
| | - Jiakun Zuo
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences 200241 Shanghai China
| | - Wen Zhou
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences 200241 Shanghai China
- Key laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Research Institute, Chinese Academy of Agricultural Sciences Shanghai 200241 China
| | - Xiangan Han
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences 200241 Shanghai China
| |
Collapse
|
13
|
Ma C, Wei R, Yu R, Lei L, Pan X, Hu HY, Feng B, Liu Z. Design, synthesis of griseofamine A derivatives and development of potent antibacterial agents against MRSA. Eur J Med Chem 2024; 276:116703. [PMID: 39059183 DOI: 10.1016/j.ejmech.2024.116703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 07/19/2024] [Accepted: 07/19/2024] [Indexed: 07/28/2024]
Abstract
The prevalence of methicillin-resistant Staphylococcus aureus (MRSA), one of the most important multidrug-resistant bacteria in clinic, has become a serious global health issue. In this study, we designed and synthesized a series of griseofamine A derivatives and evaluated their antibacterial profiles. In vitro assays found that compound 9o10 showed a remarkable improvement of antibacterial activity toward MRSA (MIC = 0.0625 μg/mL), compared with griseofamine A (MIC = 8 μg/mL) and vancomycin (MIC = 0.5 μg/mL) with low hemolysis and cytotoxicity. Its rapid bactericidal property was also confirmed by time-kill curve assay. Furthermore, compound 9o10 displayed weak drug resistance frequency. In in vivo experiment, compound 9o10 exhibited more potent antibacterial efficacy than vancomycin and excellent biosafety (LD50 > 2 g/kg). Preliminary mechanism study revealed compound 9o10 might involve antibacterial mechanisms contributing to membrane damage. Taken together, compound 9o10 possessed excellent inhibitory activity against MRSA in vitro and in vivo with low toxicity and drug resistance frequency, making it a promising hit compound for further development against MRSA infections.
Collapse
Affiliation(s)
- Caiyun Ma
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100050, PR China
| | - Rao Wei
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100050, PR China
| | - Rui Yu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100050, PR China
| | - Ling Lei
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100050, PR China
| | - Xuan Pan
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100050, PR China.
| | - Hai-Yu Hu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100050, PR China.
| | - Bo Feng
- Department of Pharmacy, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, PR China.
| | - Zhanzhu Liu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100050, PR China.
| |
Collapse
|
14
|
Umesh, Chandran VC, Saha P, Nath D, Bera S, Bhattacharya S, Pal A. A hydrogel based on Fe(II)-GMP demonstrates tunable emission, self-healing mechanical strength and Fenton chemistry-mediated notable antibacterial properties. NANOSCALE 2024; 16:13050-13060. [PMID: 38899974 DOI: 10.1039/d4nr01011f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Supramolecular hydrogels serve as an excellent platform to enable in situ reactive oxygen species (ROS) generation while maintaining controlled localized conditions, thereby mitigating cytotoxicity. Herein, we demonstrate hydrogel formation using guanosine-5'-monophosphate (GMP) with tetra(4-carboxylphenyl) ethylene (1) to exhibit aggregation-induced emission (AIE) and tunable mechanical strength in the presence of divalent metal ions such as Ca2+, Mg2+, and Fe2+. The addition of divalent metal ions leads to structural transformation in the metallogels (M-1GMP). Furthermore, the incorporation of Fe2+ ions into the hydrogel (Fe-1GMP) promotes the Fenton reaction that could be upregulated upon adding ascorbic acid (AA), demonstrating antibacterial efficacy via ROS generation. In vitro studies on AA-loaded Fe-1GMP demonstrate excellent bacterial killing efficacy against E. coli, S. aureus and vancomycin-resistant enterococci (VRE) strains. Finally, in vivo studies involving topical administration of Fe-1GMP to Balb/c mice with skin infections further suggest the potential antibacterial efficacy of the hydrogel. Taken together, the hydrogel with its unique combination of mechanical tunability, ROS generation capability and antibacterial efficacy can be used for biomedical applications, particularly in wound healing and infection control.
Collapse
Affiliation(s)
- Umesh
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector 81, Mohali, Punjab 140306, India.
| | - Vysakh C Chandran
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector 81, Mohali, Punjab 140306, India.
| | - Pranay Saha
- School of Applied & Interdisciplinary Sciences, Indian Association for the Cultivation of Science, Kolkata, 700032, India.
| | - Debasish Nath
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector 81, Mohali, Punjab 140306, India.
| | - Sayan Bera
- School of Applied & Interdisciplinary Sciences, Indian Association for the Cultivation of Science, Kolkata, 700032, India.
| | - Santanu Bhattacharya
- School of Applied & Interdisciplinary Sciences, Indian Association for the Cultivation of Science, Kolkata, 700032, India.
- Department of Chemistry, Indian Institute of Science Education and Research Tirupati, Yerpedu Mandal, Tirupati District, Andhra Pradesh, 517619, India
| | - Asish Pal
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector 81, Mohali, Punjab 140306, India.
| |
Collapse
|
15
|
Korshoj LE, Kielian T. Bacterial single-cell RNA sequencing captures biofilm transcriptional heterogeneity and differential responses to immune pressure. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.28.601229. [PMID: 38979200 PMCID: PMC11230364 DOI: 10.1101/2024.06.28.601229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Biofilm formation is an important mechanism of survival and persistence for many bacterial pathogens. These multicellular communities contain subpopulations of cells that display vast metabolic and transcriptional diversity along with high recalcitrance to antibiotics and host immune defenses. Investigating the complex heterogeneity within biofilm has been hindered by the lack of a sensitive and high-throughput method to assess stochastic transcriptional activity and regulation between bacterial subpopulations, which requires single-cell resolution. We have developed an optimized bacterial single-cell RNA sequencing method, BaSSSh-seq, to study Staphylococcus aureus diversity during biofilm growth and transcriptional adaptations following immune cell exposure. We validated the ability of BaSSSh-seq to capture extensive transcriptional heterogeneity during biofilm compared to planktonic growth. Application of new computational tools revealed transcriptional regulatory networks across the heterogeneous biofilm subpopulations and identification of gene sets that were associated with a trajectory from planktonic to biofilm growth. BaSSSh-seq also detected alterations in biofilm metabolism, stress response, and virulence that were tailored to distinct immune cell populations. This work provides an innovative platform to explore biofilm dynamics at single-cell resolution, unlocking the potential for identifying biofilm adaptations to environmental signals and immune pressure.
Collapse
|
16
|
Huang YJ, Yang MH, Lin LY, Liu J, Zang YP, Lin J, Chen WM. Exploring the Localization of Siderophore-Mediated Cargo Delivery in Gram-Negative Bacteria Using 3-Hydroxypyridin-4(1 H)-one-Fluorescein Probes. ACS Infect Dis 2024; 10:2303-2317. [PMID: 38725130 DOI: 10.1021/acsinfecdis.4c00287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
The design of siderophore-antibiotic conjugates is a promising strategy to overcome drug resistance in negative bacteria. However, accumulating studies have shown that only those antibiotics acting on the cell wall or cell membrane multiply their antibacterial effects when coupled with siderophores, while antibiotics acting on targets in the cytoplasm of bacteria do not show an obvious enhancement of their antibacterial effects when coupled with siderophores. To explore the causes of this phenomenon, we synthesized several conjugate probes using 3-hydroxypyridin-4(1H)-ones as siderophores and replacing the antibiotic cargo with 5-carboxyfluorescein (5-FAM) or malachite green (MG) cargo. By monitoring changes in the fluorescence intensity of FAM conjugate 20 in bacteria, the translocation of the conjugate across the outer membranes of Gram-negative pathogens was confirmed. Further, the use of the fluorogen activating protein(FAP)/MG system revealed that 3-hydroxypyridin-4(1H)-one-MG conjugate 26 was ultimately distributed mainly in the periplasm rather than being translocated into the cytosol of Escherichia coli and Pseudomonas aeruginosa PAO1. Additional mechanistic studies suggested that the uptake of the conjugate involved the siderophore-dependent iron transport pathway and the 3-hydroxypyridin-4(1H)-ones siderophore receptor-dependent mechanism. Meanwhile, we demonstrated that the conjugation of 3-hydroxypyridin-4(1H)-ones to the fluorescein 5-FAM can reduce the possibility of the conjugates crossing the membrane layers of mammalian Vero cells by passive diffusion, and the advantages of the mono-3-hydroxypyridin-4(1H)-ones as a delivery vehicle in the design of conjugates compared to the tri-3-hydroxypyridin-4(1H)-ones. Overall, this work reveals the localization rules of 3-hydroxypyridin-4(1H)-ones as siderophores to deliver the cargo into Gram-negative bacteria. It provides a theoretical basis for the subsequent design of siderophore-antibiotic conjugates, especially based on 3-hydroxypyridin-4(1H)-ones as siderophores.
Collapse
Affiliation(s)
- Yong-Jun Huang
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 511400, China
| | - Ming-Han Yang
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 511400, China
| | - Ling-Yin Lin
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 511400, China
| | - Jun Liu
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 511400, China
| | - Yi-Peng Zang
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 511400, China
| | - Jing Lin
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 511400, China
| | - Wei-Min Chen
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 511400, China
| |
Collapse
|
17
|
Khranovska N, Skachkova O, Gorbach O, Semchuk I, Shvets Y, Komarov I. ANTICANCER IMMUNOGENIC POTENTIAL OF ONCOLYTIC PEPTIDES: RECENT ADVANCES AND NEW PROSPECTS. Exp Oncol 2024; 46:3-12. [PMID: 38852058 DOI: 10.15407/exp-oncology.2024.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Indexed: 06/10/2024]
Abstract
Oncolytic peptides are derived from natural host defense peptides/antimicrobial peptides produced in a wide variety of life forms. Over the past two decades, they have attracted much attention in both basic research and clinical applications. Oncolytic peptides were expected to act primarily on tumor cells and also trigger the immunogenic cell death. Their ability in the tumor microenvironment remodeling and potentiating the anticancer immunity has long been ignored. Despite the promising results, clinical application of oncolytic peptides is still hindered by their unsatisfactory bioactivity and toxicity to normal cells. To ensure safer therapy, various approaches are being developed. The idea of the Ukrainian research group was to equip peptide molecules with a "molecular photoswitch" - a diarylethene fragment capable of photoisomerization, allowing for the localized photoactivation of peptides within tumors reducing side effects. Such oncolytic peptides that may induce the membrane lysis-mediated cancer cell death and subsequent anticancer immune responses in combination with the low toxicity to normal cells have provided a new paradigm for cancer therapy. This review gives an overview of the broad effects and perspectives of oncolytic peptides in anticancer immunity highlighting the potential issues related to the use of oncolytic peptides in cancer immunotherapy. We summarize the current status of research on peptide-based tumor immunotherapy in combination with other therapies including immune checkpoint inhibitors, chemotherapy, and targeted therapy.
Collapse
Affiliation(s)
- N Khranovska
- Nonprofit organization "National Cancer Institute", Kyiv, Ukraine
| | - O Skachkova
- Nonprofit organization "National Cancer Institute", Kyiv, Ukraine
| | - O Gorbach
- Nonprofit organization "National Cancer Institute", Kyiv, Ukraine
| | - I Semchuk
- Nonprofit organization "National Cancer Institute", Kyiv, Ukraine
| | - Yu Shvets
- Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - I Komarov
- Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| |
Collapse
|
18
|
Salem ME, Abdelhamid IA, Elwahy AH, Ragheb MA, Alqahtani AS, Zaki ME, Algethami FK, Mahmoud HK. Novel hybrid thiazoles, bis-thiazoles linked to azo-sulfamethoxazole: Synthesis, docking, and antimicrobial activity. Heliyon 2024; 10:e31082. [PMID: 38813143 PMCID: PMC11133767 DOI: 10.1016/j.heliyon.2024.e31082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 05/03/2024] [Accepted: 05/09/2024] [Indexed: 05/31/2024] Open
Abstract
The reaction of sulfamethoxazolehydrazonoyl chloride with thiosemicarbazones, bis-thiosemicarbazones, or 4-amino-3-mercapto-1,2,4-triazole in dioxane in the presence of triethylamine as a basic catalyst at reflux resulted in the regioselective synthesis of thiazoles and bis-thiazoles linked to azo-sulfamethoxazole as novel hybrid molecules. The structures of the new compounds were confirmed using a range of spectra. Each compound's antibacterial properties were evaluated using the agar well-diffusion technique, and most of them demonstrated significant potency. In silico investigations revealed that the described compounds had strong interactions with the binding sites of MurE ligase, tyrosyl-tRNA synthetase, and dihydropteroate synthase, demonstrating inhibitory activity.
Collapse
Affiliation(s)
- Mostafa E. Salem
- Department of Chemistry, College of Science, Imam Mohammad Ibn Saud Islamic University(IMSIU), P.O. Box, 90950, Riyadh, 11623, Saudi Arabia
- Department of Chemistry, Faculty of Science, Cairo University, Giza, 12613, Egypt
| | - Ismail A. Abdelhamid
- Department of Chemistry, Faculty of Science, Cairo University, Giza, 12613, Egypt
| | - Ahmed H.M. Elwahy
- Department of Chemistry, Faculty of Science, Cairo University, Giza, 12613, Egypt
| | - Mohamed A. Ragheb
- Department of Chemistry (Biochemistry Division), Faculty of Science, Cairo University, Giza, 12613, Egypt
| | - Arwa sultan Alqahtani
- Department of Chemistry, College of Science, Imam Mohammad Ibn Saud Islamic University(IMSIU), P.O. Box, 90950, Riyadh, 11623, Saudi Arabia
| | - Magdi E.A. Zaki
- Department of Chemistry, College of Science, Imam Mohammad Ibn Saud Islamic University(IMSIU), P.O. Box, 90950, Riyadh, 11623, Saudi Arabia
| | - Faisal K. Algethami
- Department of Chemistry, College of Science, Imam Mohammad Ibn Saud Islamic University(IMSIU), P.O. Box, 90950, Riyadh, 11623, Saudi Arabia
| | - Huda Kamel Mahmoud
- Department of Chemistry, Faculty of Science, Cairo University, Giza, 12613, Egypt
| |
Collapse
|
19
|
Lin S, Chen Y, Sun Y, Yu G, Liao X, Yang Q. Evaluation of multi-target iridium(iii)-based metallodrugs in combating antimicrobial resistance and infections caused by Staphylococcus aureus. RSC Adv 2024; 14:16194-16206. [PMID: 38769952 PMCID: PMC11103350 DOI: 10.1039/d4ra02152e] [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: 03/21/2024] [Accepted: 04/28/2024] [Indexed: 05/22/2024] Open
Abstract
The rapid emergence and spread of multidrug-resistant bacteria pose a serious challenge to human life and health, necessitating the development of novel antibacterial agents. Herein, to address this challenge, three iridium-based antibacterial agents were prepared and their antimicrobial activity were explored. Importantly, the three complexes all showed robust potency against S. aureus with MIC values in the range of 1.9-7.9 μg mL-1. Notably, the most active complex Ir3 also exhibited relative stability in mammalian fluids and a significant antibacterial effect on clinically isolated drug-resistant bacteria. Mechanism studies further demonstrated that the complex Ir3 can kill S. aureus by disrupting the integrity of the bacterial membrane and inducing ROS production. This multi-target advantage allows Ir3 to not only effectively combat bacterial resistance but also efficiently clear the bacterial biofilm. In addition, when used together, complex Ir3 could enhance the antibacterial potency of some clinical antibiotics against S. aureus. Moreover, both G. mellonella wax worms and mouse infection model demonstrated that Ir3 has low toxicity and robust anti-infective efficacy in vivo. Overall, complex Ir3 can serve as a new antibacterial agent for combating Gram-positive bacterial infections.
Collapse
Affiliation(s)
- Shijie Lin
- Department of Pharmacy, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University) Haikou 570311 China
| | - Yushou Chen
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science&Technology Normal University Nanchang 330013 China
| | - Yajuan Sun
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science&Technology Normal University Nanchang 330013 China
| | - Guangying Yu
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science&Technology Normal University Nanchang 330013 China
| | - Xiangwen Liao
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science&Technology Normal University Nanchang 330013 China
| | - Qiang Yang
- Department of Clinical Pharmacy, Hainan Cancer Hospital Haikou 570100 China
| |
Collapse
|
20
|
Domínguez-Martín EM, Ntungwe E, Isca VMS, Princiotto S, Díaz-Lanza AM, André V, Ríjo P. Antibiotic Coordination Frameworks against Antibiotic Resistance: How to Involve Students through Experimental Practices in the Search for Solutions to Public Health Problems. JOURNAL OF CHEMICAL EDUCATION 2024; 101:2045-2051. [PMID: 38764939 PMCID: PMC11097387 DOI: 10.1021/acs.jchemed.3c01125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 02/16/2024] [Accepted: 02/20/2024] [Indexed: 05/21/2024]
Abstract
For decades, multiple varieties of antibiotics have been successfully used for therapeutic purposes. Nevertheless, antibiotic resistance is currently one of the major threats to global health. This work presents an innovative laboratory practice carried out in an inorganic medicinal chemistry course within the Degrees of Pharmacy and Biochemistry for undergraduate students. This experiment includes three classes of 2 h each. The first class consisted of the mechanochemical synthesis of an antibiotic coordination framework (ACF) using a known antibiotic (nalidixic acid) and zinc as the ligand. The prepared Zn-nalidixic acid ACF (Zn-ACF) was obtained in up to 82% yield with high purity. On the second day, the synthesized Zn-ACF was characterized by Fourier-transform infrared spectroscopy (FTIR) and powder X-ray diffraction (PXRD). Finally, during the last class, the antimicrobial activity was tested against Escherichia coli by the well diffusion method. The students verified the higher antimicrobial activity of Zn-ACF compared to nalidixic acid, proving that small changes in the chemical structure can result in great biological differences. In the end, the students presented their results in a poster format, encouraging the development of their soft skills and scientific results communication and dissemination. In the future, it is expected that such a laboratory experiment at the interface between medicinal chemistry, microbiology, analytical techniques, public health, and pharmacology will lead to the development and implementation of some service-learning practices and will serve as a model to look at for other courses and institutions.
Collapse
Affiliation(s)
- Eva María Domínguez-Martín
- CBIOS
− Universidade Lusófona’s Research Center for
Biosciences & Health Technologies, Campo Grande 376, 1749-024 Lisbon, Portugal
- Universidad
de Alcalá de Henares, Facultad de Farmacia, Departamento de Ciencias Biomédicas
(Área de Farmacología); Nuevos Agentes Antitumorales,
Acción Tóxica Sobre Células Leucémicas, Ctra. Madrid-Barcelona km. 33,600, 28805 Alcalá de Henares, Madrid, España
| | - Epole Ntungwe
- CBIOS
− Universidade Lusófona’s Research Center for
Biosciences & Health Technologies, Campo Grande 376, 1749-024 Lisbon, Portugal
- Universidad
de Alcalá de Henares, Facultad de Farmacia, Departamento de Ciencias Biomédicas
(Área de Farmacología); Nuevos Agentes Antitumorales,
Acción Tóxica Sobre Células Leucémicas, Ctra. Madrid-Barcelona km. 33,600, 28805 Alcalá de Henares, Madrid, España
| | - Vera M. S. Isca
- CBIOS
− Universidade Lusófona’s Research Center for
Biosciences & Health Technologies, Campo Grande 376, 1749-024 Lisbon, Portugal
- Instituto
de Investigação do Medicamento (iMed.ULisboa), Faculdade
de Farmácia, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Salvatore Princiotto
- Department
of Food, Environmental and Nutritional Sciences (DeFENS), University of Milan, Via Celoria 2, Milan 20133, Italy
| | - Ana María Díaz-Lanza
- Universidad
de Alcalá de Henares, Facultad de Farmacia, Departamento de Ciencias Biomédicas
(Área de Farmacología); Nuevos Agentes Antitumorales,
Acción Tóxica Sobre Células Leucémicas, Ctra. Madrid-Barcelona km. 33,600, 28805 Alcalá de Henares, Madrid, España
| | - Vânia André
- Centro
de Química Estrutural, Institute of Molecular Sciences, Instituto
Superior Técnico, Universidade de
Lisboa, Avenida Rovisco
Pais, 1049-001 Lisbon, Portugal
| | - Patrícia Ríjo
- CBIOS
− Universidade Lusófona’s Research Center for
Biosciences & Health Technologies, Campo Grande 376, 1749-024 Lisbon, Portugal
- Instituto
de Investigação do Medicamento (iMed.ULisboa), Faculdade
de Farmácia, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| |
Collapse
|
21
|
Yang R, Xue Z, Li X, Xu T, Zhong Y, Hu S, Qin S, Guo Y. Novel natural osthole-inspired amphiphiles as membrane targeting antibacterials against methicillin-resistant Staphylococcus aureus (MRSA). Eur J Med Chem 2024; 271:116449. [PMID: 38691893 DOI: 10.1016/j.ejmech.2024.116449] [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: 03/19/2024] [Revised: 04/23/2024] [Accepted: 04/23/2024] [Indexed: 05/03/2024]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is a widespread pathogen causing clinical infections and is multi-resistant to many antibiotics, making it urgent need to develop novel antibacterials to combat MRSA. Herein, we designed and prepared a series of novel osthole amphiphiles 6a-6ad by mimicking the structures and function of antimicrobial peptides (AMPs). Antibacterial assays showed that osthole amphiphile 6aa strongly inhibited S. aureus and 10 clinical MRSA isolates with MIC values of 1-2 μg/mL, comparable to that of the commercial antibiotic vancomycin. Additionally, 6aa had the advantages of rapid bacteria killing without readily developing drug resistance, low toxicity, good membrane selectivity, and good plasma stability. Mechanistic studies indicated that 6aa possesses good membrane-targeting ability to bind to phosphatidylglycerol (PG) on the bacterial cell membranes, thereby disrupting the cell membranes and causing an increase in intracellular ROS as well as leakage of proteins and DNA, and accelerating bacterial death. Notably, in vivo activity results revealed that 6aa exhibits strong anti-MRSA efficacy than vancomycin as well as a substantial reduction in MRSA-induced proinflammatory cytokines, including TNF-α and IL-6. Given the impressive in vitro and in vivo anti-MRSA efficacy of 6aa, which makes it a potential candidate against MRSA infections.
Collapse
Affiliation(s)
- Ruige Yang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan Province, China; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan Province, China.
| | - Zihan Xue
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| | - Xinhui Li
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| | - Ting Xu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan Province, China
| | - Yan Zhong
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan Province, China
| | - Songlin Hu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan Province, China
| | - Shangshang Qin
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| | - Yong Guo
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan Province, China; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan Province, China.
| |
Collapse
|
22
|
Huang YJ, Zang YP, Peng LJ, Yang MH, Lin J, Chen WM. Cajaninstilbene acid derivatives conjugated with siderophores of 3-hydroxypyridin-4(1H)-ones as novel antibacterial agents against Gram-negative bacteria based on the Trojan horse strategy. Eur J Med Chem 2024; 269:116339. [PMID: 38537513 DOI: 10.1016/j.ejmech.2024.116339] [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: 02/03/2024] [Revised: 03/14/2024] [Accepted: 03/15/2024] [Indexed: 04/07/2024]
Abstract
The low permeability of the outer membrane of Gram-negative bacteria is a serious obstacle to the development of new antibiotics against them. Conjugation of antibiotic with siderophore based on the "Trojan horse strategy" is a promising strategy to overcome the outer membrane obstacle. In this study, series of antibacterial agents were designed and synthesized by conjugating the 3-hydroxypyridin-4(1H)-one based siderophores with cajaninstilbene acid (CSA) derivative 4 which shows good activity against Gram-positive bacteria by targeting their cell membranes but is ineffective against Gram-negative bacteria. Compared to the inactive parent compound 4, the conjugates 45c or 45d exhibits significant improvement in activity against Gram-negative bacteria, including Escherichia coli, Klebsiella pneumoniae and especially P. aeruginosa (minimum inhibitory concentrations, MICs = 7.8-31.25 μM). The antibacterial activity of the conjugates is attributed to the CSA derivative moiety, and the action mechanism is by disruption of bacterial cell membranes. Further studies on the uptake mechanisms showed that the bacterial siderophore-dependent iron transport system was involved in the uptake of the conjugates. In addition, the conjugates 45c and 45d showed a lower cytotoxic effects in vivo and in vitro and a positive therapeutic effect in the treatment of C. elegans infected by P. aeruginosa. Overall, our work describes a new class and a promising 3-hydroxypyridin-4(1H)-one-CSA derivative conjugates for further development as antibacterial agents against Gram-negative bacteria.
Collapse
Affiliation(s)
- Yong-Jun Huang
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou, 511400, China
| | - Yi-Peng Zang
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou, 511400, China
| | - Li-Jun Peng
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou, 511400, China
| | - Ming-Han Yang
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou, 511400, China
| | - Jing Lin
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou, 511400, China.
| | - Wei-Min Chen
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou, 511400, China.
| |
Collapse
|
23
|
He H, Huang X, Wen C, Liu C, Jiang B, Huang Y, Su Y, Li W. A novel defensin-like peptide C-13326 possesses protective effect against multidrug-resistant Aeromonas schubertii in hybrid snakehead (Channa maculate ♀ × Channa argus ♂). JOURNAL OF FISH DISEASES 2024; 47:e13922. [PMID: 38204197 DOI: 10.1111/jfd.13922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 12/27/2023] [Accepted: 12/29/2023] [Indexed: 01/12/2024]
Abstract
The purpose of this study was to investigate whether a defensin-like antimicrobial peptide (C-13326 peptide) identified in Hermetia illucens could possess protective effect against multidrug-resistant Aeromonas schubertii in hybrid snakehead (Channa maculate ♀ × Channa argus ♂). The cDNA of C-13326 peptide comprised 243 nucleotides encoding 80 amino acids, with six conserved cysteine residues and the classical CSαβ structure. The recombinant expression plasmid pPIC9K-C-13326 was constructed and transformed into GS115 Pichia pastoris, and the C-13326 peptide was expressed by induction with 1% methanol. The crude extract of C-13326 peptide was precipitated by ammonium sulfate, assayed by Braford method, detected by tricine-SDS-PAGE, evaluated by BandScan software and identified by liquid chromatography-mass spectrometry. The C-13326 peptide was shown to have inhibitory activity against the growth of multidrug-resistant A. schubertii DM210910 by using the minimum growth inhibitory concentration and Oxford cup method. In addition, scanning electron microscopy analysis suggested that C-13326 peptide inhibited the growth of A. schubertii DM210910 by damaging the bacterial cell membrane. To explore the role of peptide C-13326 in vivo, hybrid snakehead was fed with peptide C-13326 as feed additives for 7 days. The results revealed that C-13326 peptide could significantly down-regulate the expression levels of IL-1β, IL-8, IL-12 and TNF-α (p < .05), and significantly improved the survival rate of hybrid snakehead after challenging with A. schubertii DM210910. Therefore, the C-13326 peptide is a promising antimicrobial agent for A. schubertii treatment in aquaculture.
Collapse
Affiliation(s)
- Huanrong He
- Innovative Institute of Animal Healthy Breeding, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Science and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, China
| | - Xinzhi Huang
- Innovative Institute of Animal Healthy Breeding, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Science and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, China
| | - Caiyi Wen
- Innovative Institute of Animal Healthy Breeding, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Science and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, China
| | - Chun Liu
- Innovative Institute of Animal Healthy Breeding, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Science and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, China
| | - Biao Jiang
- Innovative Institute of Animal Healthy Breeding, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Science and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, China
| | - Yanhua Huang
- Innovative Institute of Animal Healthy Breeding, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Science and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, China
| | - Youlu Su
- Innovative Institute of Animal Healthy Breeding, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Science and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, China
| | - Wei Li
- Innovative Institute of Animal Healthy Breeding, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Science and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, China
| |
Collapse
|
24
|
Homayonia S, Ling CC. Epoxide-Mediated Trans-Thioglycosylation and Application to the Synthesis of Oligosaccharides Related to the Capsular Polysaccharides of C. jejuni HS:4. Chemistry 2024; 30:e202303753. [PMID: 38215247 DOI: 10.1002/chem.202303753] [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: 11/10/2023] [Revised: 01/10/2024] [Accepted: 01/10/2024] [Indexed: 01/14/2024]
Abstract
The enzyme-resistant thioglycosides are highly valuable immunogens because of their enhanced metabolic stability. We report the first synthesis of a family of thiooligosaccharides related to the capsular polysaccharides (CPS) of Campylobacter jejuni HS:4 for potential use in conjugate vaccines. The native CPS structures of the pathogen consist of a challenging repeating disaccharide formed with β(1→4)-linked 6-deoxy-β-D-ido-heptopyranoside and N-acetyl-D-glucosamine; the rare 6-deoxy-ido-heptopyranosyl backbone and β-anomeric configuration of the former monosaccharide makes the synthesis of this family of antigens very challenging. So far, no synthesis of the thioanalogs of the CPS antigens have been reported. The unprecedented synthesis presented in this work is built on an elegant approach by using β-glycosylthiolate as a glycosyl donor to open the 2,3-epoxide functionality of pre-designed 6-deoxy-β-D-talo-heptopyranosides. Our results illustrated that this key trans-thioglycosylation can be designed in a modular and regio and stereo-selective manner. Built on the success of this novel approach, we succeeded the synthesis of a family of thiooligosaccharides including a thiohexasaccharide which is considered to be the desired antigen length and complexity for immunizations. We also report the first direct conversion of base-stable but acid-labile 2-trimethylsilylethyl glycosides to glycosyl-1-thioacetates in a one-pot manner.
Collapse
Affiliation(s)
- Saba Homayonia
- Department of Chemistry, University of Calgary, 2500 University Drive NW, Calgary, Alberta, T2N 1N4, Canada
| | - Chang-Chun Ling
- Department of Chemistry, University of Calgary, 2500 University Drive NW, Calgary, Alberta, T2N 1N4, Canada
| |
Collapse
|
25
|
Niode NJ, Kepel BJ, Hessel SS, Kairupan TS, Tallei TE. Rhynchophorus ferrugineus larvae: A novel source for combating broad-spectrum bacterial and fungal infections. Vet World 2024; 17:156-170. [PMID: 38406375 PMCID: PMC10884581 DOI: 10.14202/vetworld.2024.156-170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 12/21/2023] [Indexed: 02/27/2024] Open
Abstract
Antimicrobial resistance is a growing concern due to the growth of antibiotic-resistant microorganisms, which makes it difficult to treat infection. Due to its broad-spectrum antimicrobial properties against a diverse array of bacteria, both Gram-positive and Gram-negative bacteria, and fungi, Rhynchophorus ferrugineus larval antimicrobial peptides (AMPs) have demonstrated potential as antimicrobial agents for the treatment of microbial infections and prevention of antibiotic resistance. This study emphasizes the unexplored mechanisms of action of R. ferrugineus larvae against microorganisms. Among the most widely discussed mechanisms is the effect of AMPs in larvae in response to a threat or infection. Modulation of immune-related genes in the intestine and phagocytic capacity of its hemocytes may also affect the antimicrobial activity of R. ferrugineus larvae, with an increase in phenoloxidase activity possibly correlated with microbial clearance and survival rates of larvae. The safety and toxicity of R. ferrugineus larvae extracts, as well as their long-term efficacy, are also addressed in this paper. The implications of future research are explored in this paper, and it is certain that R. ferrugineus larvae have the potential to be developed as a broad-spectrum antimicrobial agent with proper investigation.
Collapse
Affiliation(s)
- Nurdjannah Jane Niode
- Department of Dermatology and Venereology, Faculty of Medicine, Sam Ratulangi University, Prof. Dr. R. D. Kandou Hospital Manado, Manado 95115, North Sulawesi, Indonesia
| | - Billy Johnson Kepel
- Department of Chemistry, Faculty of Medicine, Sam Ratulangi University, Manado 95115, North Sulawesi, Indonesia
| | - Sofia Safitri Hessel
- Department of Biotechnology, Indonesia Biodiversity and Biogeography Research Institute (INABIG), Bandung 40132, West Java, Indonesia
| | - Tara Sefanya Kairupan
- Department of Dermatology and Venereology, Faculty of Medicine, Sam Ratulangi University, Prof. Dr. R. D. Kandou Hospital Manado, Manado 95115, North Sulawesi, Indonesia
| | - Trina Ekawati Tallei
- Department of Biology, Faculty of Mathematics and Natural Sciences, Sam Ratulangi University, Manado 95115, North Sulawesi, Indonesia
- Department of Biology, Faculty of Medicine, Sam Ratulangi University, Manado 95115, North Sulawesi, Indonesia
| |
Collapse
|
26
|
Zhu YY, Wang ZJ, Zhu M, Zhou ZS, Hu BY, Wei MZ, Zhao YL, Dai Z, Luo XD. A dual mechanism with H 2S inhibition and membrane damage of morusin from Morus alba Linn. against MDR-MRSA. Bioorg Med Chem 2024; 97:117544. [PMID: 38071943 DOI: 10.1016/j.bmc.2023.117544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 11/15/2023] [Accepted: 12/04/2023] [Indexed: 12/30/2023]
Abstract
It's urgent to discover new antibiotics along with the increasing emergence and dissemination of multidrug resistant (MDR) bacterial pathogens. In the present investigation, morusin exhibited rapid bactericidal activity against methicillin-resistant Staphylococcus aureus (MRSA) and vancomycin-resistant Enterococcus (VRE) by targeting the phospholipid of bacterial inner membrane, increasing membrane rigidity and disrupting bacterial homeostasis together with the membrane permeability, which caused fundamental metabolic disorders. Furthermore, morusin can also accumulate ROS, suppress H2S production, and aggravate oxidative damage in bacteria. Importantly, morusin also inhibited the spread of wounds and reduced the bacterial burden in the mouse model of skin infection caused by MRSA. It's a chance to meet the challenge of existing antibiotic resistance and avoid the development of bacterial resistance, given the multiple targets of morusin.
Collapse
Affiliation(s)
- Yan-Yan Zhu
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650500, People's Republic of China
| | - Zhao-Jie Wang
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650500, People's Republic of China
| | - Meng Zhu
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650500, People's Republic of China
| | - Zhong-Shun Zhou
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650500, People's Republic of China
| | - Bin-Yuan Hu
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650500, People's Republic of China
| | - Mei-Zhen Wei
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650500, People's Republic of China
| | - Yun-Li Zhao
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650500, People's Republic of China
| | - Zhi Dai
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650500, People's Republic of China
| | - Xiao-Dong Luo
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650500, People's Republic of China; State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, People's Republic of China.
| |
Collapse
|
27
|
Tariq M, Shivalkar S, Hasan H, Sahoo AK, Sk MP. Manganese Doping in Biomass Derived Carbon Dots Amplifies White Light-Induced Antibacterial Activity. ACS OMEGA 2023; 8:49460-49466. [PMID: 38162730 PMCID: PMC10753716 DOI: 10.1021/acsomega.3c08586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 11/21/2023] [Accepted: 11/23/2023] [Indexed: 01/03/2024]
Abstract
The prevalence of antibiotic-resistant bacterial infections demands effective alternative therapeutics of antibiotics, whereas biocompatible zero-dimensional nanomaterials are an excellent option due to their small size. In this study, we report the one-step hydrothermal approach that was used to synthesize luminescent manganese doped carbon dots (Mn-Cdots) with an efficient quantum yield of 9.2% by employing green Psidium guajava L. (Guava) leaf as the precursor. High-resolution microscopy TEM was used to investigate the average particle size of Mn-Cdots, which was found to be 2.9 ± 0.045 nm. The structural properties and elemental composition of Mn-Cdots were analyzed by FTIR, XRD, EPR, and XPS spectroscopy, and the optical properties of Mn-Cdots were examined by UV-visible and fluorescent spectroscopy. Light-mediated antibacterial activity of Mn-Cdots was investigated by Gram-negative bacteria E. coli under white, blue, and yellow light. The doping effect of a minute quantity of Mn in Mn-Cdots increased the level of ROS generation in the presence of white lights compared to Cdots. Thus, Mn-Cdots might act as potent antibacterial agents.
Collapse
Affiliation(s)
- Mohammad Tariq
- Department
of Chemistry, Aligarh Muslim University, Aligarh, 202002 Uttar Pradesh, India
| | - Saurabh Shivalkar
- Department
of Applied Sciences, Indian Institute of
Information Technology Allahabad, Prayagraj, 211012 Uttar Pradesh, India
| | - Hammad Hasan
- Department
of Chemistry, Aligarh Muslim University, Aligarh, 202002 Uttar Pradesh, India
| | - Amaresh Kumar Sahoo
- Department
of Applied Sciences, Indian Institute of
Information Technology Allahabad, Prayagraj, 211012 Uttar Pradesh, India
| | - Md Palashuddin Sk
- Department
of Chemistry, Aligarh Muslim University, Aligarh, 202002 Uttar Pradesh, India
| |
Collapse
|
28
|
Alzahrani HA. Quinoline-2-one derivatives as promising antibacterial agents against multidrug-resistant Gram-positive bacterial strains. Braz J Microbiol 2023; 54:2799-2805. [PMID: 37831330 PMCID: PMC10689604 DOI: 10.1007/s42770-023-01132-w] [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: 03/15/2023] [Accepted: 09/15/2023] [Indexed: 10/14/2023] Open
Abstract
This study describes the discovery of a variety of quinoline2-one derivatives with significant antibacterial action vs a spectrum of multidrug-resistant Gram-positive bacterial strains, especially methicillin-resistant Staphylococcus aureus (MRSA). Compounds 6c, 6l, and 6o exhibited significant antibacterial activity versus the Gram-positive bacterial pathogens evaluated. In comparison to the reference daptomycin, compound 6c demonstrated the most effective activity among the assessed derivatives, with MIC concentrations of 0.75 μg/mL versus MRSA and VRE and 2.50 μg/mL against MRSE. We also reported on these compounds' biofilm and dihydrofolate reductase inhibitory activities. Compound 6c showed the greatest antibiofilm action in a dose-dependent way and a substantial decrease of biofilm development in the MRSA ACL51 strain at concentrations of 0.5, 0.25, and 0.12 MIC, with reductions of 79%, 55%, and 38%, consecutively, whereas the corresponding values for vancomycin were 20%, 12%, and 9%. These findings imply that these quinoline compounds could be used to develop a new category of antibiotic representatives to prevent Gram-positive drug-resistant bacterial strains.
Collapse
Affiliation(s)
- Hayat Ali Alzahrani
- Medical Laboratory Technology Department, Applied Medical Science College, Northern Border University, Arar, Saudi Arabia.
| |
Collapse
|
29
|
Mai Y, Wang Z, Zhou Y, Wang G, Chen J, Lin Y, Ji P, Zhang W, Jing Q, Chen L, Chen Z, Lin H, Jiang L, Yuan C, Xu P, Huang M. From disinfectants to antibiotics: Enhanced biosafety of quaternary ammonium compounds by chemical modification. JOURNAL OF HAZARDOUS MATERIALS 2023; 460:132454. [PMID: 37703742 DOI: 10.1016/j.jhazmat.2023.132454] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/30/2023] [Accepted: 08/30/2023] [Indexed: 09/15/2023]
Abstract
The excessive use of quaternary ammonium compounds (QACs) following the COVID-19 pandemic has raised substantial concerns regarding their biosafety. Overuse of QACs has been associated with chronic biological adverse effects, including genotoxicity or carcinogenicity. In particular, inadvertent intravascular administration or oral ingestion of QACs can lead to fatal acute toxicity. To enhance the biosafety and antimicrobial efficacy of QACs, this study reports a new series of QACs, termed as PACs, with the alkyl chain of benzalkonium substituted by a phthalocyanine moiety. Firstly, the rigid phthalocyanine moiety enhances the selectivity of QACs to bacteria over human cells and reduces alkyl chain's entropic penalty of binding to bacterial membranes. Furthermore, phthalocyanine neutralizes hemolysis and cytotoxicity of QACs by binding with albumin in plasma. Our experimental results demonstrate that PACs inherit the optical properties of phthalocyanine and validate the broad-spectrum antibacterial activity of PACs in vitro. Moreover, the intravascular administration of the most potent PAC, PAC1a, significantly reduced bacterial burden and ameliorated inflammation level in a bacteria-induced septic mouse model. This study presents a new strategy to improve the antimicrobial efficacy and biosafety of QACs, thus expanding their range of applications to the treatment of systemic infections.
Collapse
Affiliation(s)
- Yuhan Mai
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, PR China
| | - Zhiyou Wang
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, PR China
| | - Yang Zhou
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, PR China
| | - Guodong Wang
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, PR China
| | - Jingyi Chen
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, PR China
| | - Yuxin Lin
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, PR China
| | - Panpan Ji
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350108, PR China
| | - Wei Zhang
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350108, PR China
| | - Qian Jing
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, PR China
| | - Liyun Chen
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, PR China
| | - Zheng Chen
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, PR China
| | - Haili Lin
- Department of Pharmacy, The Peoples Hospital of Fujian Province, Fuzhou, Fujian 350004, PR China
| | - Longguang Jiang
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, PR China
| | - Cai Yuan
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350108, PR China; Fujian Key Laboratory of Marine Enzyme Engineering, Fuzhou University, Fuzhou, Fujian 350108, PR China
| | - Peng Xu
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350108, PR China; Fujian Key Laboratory of Marine Enzyme Engineering, Fuzhou University, Fuzhou, Fujian 350108, PR China.
| | - Mingdong Huang
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, PR China.
| |
Collapse
|
30
|
Luo B, Cai J, Xiong Y, Ding X, Li X, Li S, Xu C, Vasil'kov AY, Bai Y, Wang X. Quaternized chitosan coated copper sulfide nanozyme with peroxidase-like activity for synergistic antibacteria and promoting infected wound healing. Int J Biol Macromol 2023; 246:125651. [PMID: 37399873 DOI: 10.1016/j.ijbiomac.2023.125651] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 06/15/2023] [Accepted: 06/29/2023] [Indexed: 07/05/2023]
Abstract
Bacterial infection can hinder the infected wound healing process. Because of the growing drug-resistance bacteria, there is an urgent desire to develop alternative antibacterial strategies to the antibiotics. Herein, the quaternized chitosan coated CuS (CuS-QCS) nanozyme with peroxidase (POD)-like activity was developed through a facile biomineralized approach for synergistic efficient antibacterial therapy and wound healing. The CuS-QCS killed bacteria by the electrostatic bonding of positive charged QCS with bacteria and releasing Cu2+ to damage bacterial membrane. And importantly, CuS-QCS nanozyme exhibited higher intrinsic POD-like activity, which converted H2O2 with low concentration into highly toxic hydroxyl radical (OH) for the elimination of bacteria by oxidative stress. Through cooperation of POD-like activity, Cu2+ and QCS, CuS-QCS nanozyme exhibited excellent antibacterial efficacy of approximate 99.9 % against E. coli and S. aureus in vitro. In addition, the QCS-CuS was successfully used to promote the healing of S. aureus infected wound with good biocompatibility. This synergistic nanoplatform presented here shows great potential applications in the field of wound infection management.
Collapse
Affiliation(s)
- Bichong Luo
- State Key Laboratory of Pulp and Paper Engineering, South China University of Technology, 381 Wushan Road, Guangzhou 510640, China
| | - Jihai Cai
- State Key Laboratory of Pulp and Paper Engineering, South China University of Technology, 381 Wushan Road, Guangzhou 510640, China
| | - Yutong Xiong
- State Key Laboratory of Pulp and Paper Engineering, South China University of Technology, 381 Wushan Road, Guangzhou 510640, China
| | - Xu Ding
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, The First Clinical Medical College of Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing 210023, China
| | - Xiaoyun Li
- State Key Laboratory of Pulp and Paper Engineering, South China University of Technology, 381 Wushan Road, Guangzhou 510640, China
| | - Shanshan Li
- State Key Laboratory of Pulp and Paper Engineering, South China University of Technology, 381 Wushan Road, Guangzhou 510640, China
| | - Changliang Xu
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, The First Clinical Medical College of Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing 210023, China
| | - Alexander Yu Vasil'kov
- A. N. Nesmeyanov Institute of Organoelement Compounds, Russian Academy of Sciences, 28 Vavilov st., Moscow 119991, Russia
| | - Yun Bai
- Jiangsu Province People's Hospital (Nanjing Medical University First Affiliated Hospital), 300 Guangzhou Road, Nanjing 210029, China.
| | - Xiaoying Wang
- State Key Laboratory of Pulp and Paper Engineering, South China University of Technology, 381 Wushan Road, Guangzhou 510640, China.
| |
Collapse
|
31
|
Guliy OI, Zaitsev BD, Borodina IA. Electroacoustic Biosensor Systems for Evaluating Antibiotic Action on Microbial Cells. SENSORS (BASEL, SWITZERLAND) 2023; 23:6292. [PMID: 37514587 PMCID: PMC10383298 DOI: 10.3390/s23146292] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/07/2023] [Accepted: 07/09/2023] [Indexed: 07/30/2023]
Abstract
Antibiotics are widely used to treat infectious diseases. This leads to the presence of antibiotics and their metabolic products in the ecosystem, especially in aquatic environments. In many countries, the growth of pathogen resistance to antibiotics is considered a threat to national security. Therefore, methods for determining the sensitivity/resistance of bacteria to antimicrobial drugs are important. This review discusses the mechanisms of the formation of antibacterial resistance and the various methods and sensor systems available for analyzing antibiotic effects on bacteria. Particular attention is paid to acoustic biosensors with active immobilized layers and to sensors that analyze antibiotics directly in liquids. It is shown that sensors of the second type allow analysis to be done within a short period, which is important for timely treatment.
Collapse
Affiliation(s)
- Olga I Guliy
- Institute of Biochemistry and Physiology of Plants and Microorganisms-Subdivision of the Federal State Budgetary Research Institution Saratov Federal Scientific Centre of the Russian Academy of Sciences (IBPPM RAS), Saratov 410049, Russia
| | - Boris D Zaitsev
- Kotelnikov Institute of Radio Engineering and Electronics, Russian Academy of Sciences, Saratov Branch, Saratov 410019, Russia
| | - Irina A Borodina
- Kotelnikov Institute of Radio Engineering and Electronics, Russian Academy of Sciences, Saratov Branch, Saratov 410019, Russia
| |
Collapse
|
32
|
Yuan D, Liu S, Li S, Liu R, Zhu X. Design, Synthesis and Biological Evaluation of 7-Substituted-1,3-diaminopyrrol[3,2-f]quinazolines as Potential Antibacterial Agents. ChemMedChem 2023; 18:e202300078. [PMID: 37017005 DOI: 10.1002/cmdc.202300078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/29/2023] [Accepted: 04/04/2023] [Indexed: 04/06/2023]
Abstract
The evolution of drug-resistant bacteria poses a serious threat to public health; hence, it is imperative to develop new and efficient antibiotics. Irresistin-16 (IRS-16) is a dual-target antibacterial candidate that affects folate biosynthesis and membrane integrity and exhibits potent lethality against various bacteria. In this study, a series of 1,3-diamino-7H-pyrrol[3,2-f]quinazoline (DAPQ) derivatives based on IRS-16 was designed and synthesized to identify outstanding antibacterial candidates. The most promising compound, 7-(4-(4-methylpiperazin-1-yl) benzyl)-7H-pyrrol[3,2-f] quinazoline-1,3-diamine (18 e), displayed excellent antibacterial activity against both gram-positive and gram-negative bacteria (minimum inhibitory concentrations=1-4 μg/mL), improved water solubility, poor hemolytic activity and low cytotoxicity. Compound 18 e exhibited rapid bactericidal properties and prevented bacterial resistance in laboratory simulations. These results provide a basis for the development of new DAPQ-based compounds to combat emerging bacterial resistance.
Collapse
Affiliation(s)
- Duo Yuan
- School of Engineering, China Pharmaceutical University, Nanjing, 210009, China
| | - Shangde Liu
- School of Pharmacy, Tsinghua University, Beijing, 100084, China
| | - Shanshan Li
- School of Engineering, China Pharmaceutical University, Nanjing, 210009, China
| | - Rongrong Liu
- School of Engineering, China Pharmaceutical University, Nanjing, 210009, China
| | - Xiong Zhu
- School of Engineering, China Pharmaceutical University, Nanjing, 210009, China
| |
Collapse
|
33
|
Huang YJ, Zhong XL, Zang YP, Yang MH, Lin J, Chen WM. 3-Hydroxy-pyridin-4(1H)-ones as siderophores mediated delivery of isobavachalcone enhances antibacterial activity against pathogenic Pseudomonas aeruginosa. Eur J Med Chem 2023; 257:115454. [PMID: 37210837 DOI: 10.1016/j.ejmech.2023.115454] [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: 03/16/2023] [Revised: 04/24/2023] [Accepted: 05/03/2023] [Indexed: 05/23/2023]
Abstract
The natural prenylated chalcone isobavachalcone (IBC) shows good antibacterial activity against Gram-positive bacteria but is ineffective against Gram-negative bacteria, most likely due to the outer membrane barrier of Gram-negative bacteria. The Trojan horse strategy has been shown to be an effective strategy to overcome the reduction in the permeability of the outer membrane of Gram-negative bacteria. In this study, eight different 3-hydroxy-pyridin-4(1H)-one-isobavachalcone conjugates were designed and synthesized based on the siderophore Trojan horse strategy. The conjugates exhibited 8- to 32-fold lower minimum inhibitory concentrations (MICs) and 32- to 177-fold lower half-inhibitory concentrations (IC50s) against Pseudomonas aeruginosa PAO1 as well as clinical multidrug-resistant (MDR) strains compared to the parent IBC under iron limitation. Further studies showed that the antibacterial activity of the conjugates was regulated by the bacterial iron uptake pathway under different iron concentration conditions. Studies on the antibacterial mechanism of conjugate 1b showed that it exerts antibacterial activity by disrupting cytoplasmic membrane integrity and inhibiting cell metabolism. Finally, conjugate 1b showed a lower cytotoxic effects on Vero cells than IBC and a positive therapeutic effect in the treatment of bacterial infections caused by Gram-negative bacteria PAO1. Overall, this work demonstrates that IBC can be delivered to Gram-negative bacteria when combined with 3-hydroxy-pyridin-4(1H)-ones as siderophores and provides a scientific basis for the development of effective antibacterial agents against Gram-negative bacteria.
Collapse
Affiliation(s)
- Yong-Jun Huang
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou, 511400, China
| | - Xiao-Lin Zhong
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou, 511400, China
| | - Yi-Peng Zang
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou, 511400, China
| | - Ming-Han Yang
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou, 511400, China
| | - Jing Lin
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou, 511400, China.
| | - Wei-Min Chen
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou, 511400, China.
| |
Collapse
|
34
|
Ayon NJ. High-Throughput Screening of Natural Product and Synthetic Molecule Libraries for Antibacterial Drug Discovery. Metabolites 2023; 13:625. [PMID: 37233666 PMCID: PMC10220967 DOI: 10.3390/metabo13050625] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 04/29/2023] [Accepted: 05/01/2023] [Indexed: 05/27/2023] Open
Abstract
Due to the continued emergence of resistance and a lack of new and promising antibiotics, bacterial infection has become a major public threat. High-throughput screening (HTS) allows rapid screening of a large collection of molecules for bioactivity testing and holds promise in antibacterial drug discovery. More than 50% of the antibiotics that are currently available on the market are derived from natural products. However, with the easily discoverable antibiotics being found, finding new antibiotics from natural sources has seen limited success. Finding new natural sources for antibacterial activity testing has also proven to be challenging. In addition to exploring new sources of natural products and synthetic biology, omics technology helped to study the biosynthetic machinery of existing natural sources enabling the construction of unnatural synthesizers of bioactive molecules and the identification of molecular targets of antibacterial agents. On the other hand, newer and smarter strategies have been continuously pursued to screen synthetic molecule libraries for new antibiotics and new druggable targets. Biomimetic conditions are explored to mimic the real infection model to better study the ligand-target interaction to enable the designing of more effective antibacterial drugs. This narrative review describes various traditional and contemporaneous approaches of high-throughput screening of natural products and synthetic molecule libraries for antibacterial drug discovery. It further discusses critical factors for HTS assay design, makes a general recommendation, and discusses possible alternatives to traditional HTS of natural products and synthetic molecule libraries for antibacterial drug discovery.
Collapse
Affiliation(s)
- Navid J Ayon
- Chemistry of Life Processes Institute, Northwestern University, Evanston, IL 60208, USA
| |
Collapse
|
35
|
Jian Y, Peng Y, Zhou W, Xu Y, Li C, Wang X, Zhou Q. Ru(II) Complexes with Enaminone Structures for Rapid Sterilization of Staphylococcus aureus and MRSA with Little Accumulation of Drug Resistance. ChemMedChem 2023; 18:e202300065. [PMID: 36751034 DOI: 10.1002/cmdc.202300065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 02/07/2023] [Indexed: 02/09/2023]
Abstract
Drug-resistant bacteria, such as methicillin-resistant Staphylococcus aureus (MRSA), pose a serious threat to human life. Therefore, there is urgent need to develop antibiotics with new chemical structures and antibacterial mechanisms, especially those that elicit little drug resistance after long-term use. Herein we synthesized three novel ruthenium complexes (Ru1-Ru3) containing the enaminone structures for the first time. At a concentration of 5 μM, Ru1-Ru3 can lead to a CFU reduction of about 5 log units towards S. aureus and MRSA. Interestingly, Ru3 displayed rapid bactericidal effects and could decrease the CFU numbers of both pathogens by 5 log units within 40 min. The control compounds (Ru4 and Ru5) without the enaminone structures displayed very poor antibacterial activity under the same conditions. Moreover, S. aureus did not show apparent drug resistance towards Ru3 after 20 passages incubation with a sublethal concentration. These results highlight the critical role of enaminone structures for antibacterial applications.
Collapse
Affiliation(s)
- Yao Jian
- Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, P.R. China.,University of Chinese Academy of Sciences, Beijing, 100049, P.R. China
| | - Yatong Peng
- Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, P.R. China.,University of Chinese Academy of Sciences, Beijing, 100049, P.R. China
| | - Wanpeng Zhou
- Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, P.R. China.,University of Chinese Academy of Sciences, Beijing, 100049, P.R. China
| | - Yunli Xu
- Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, P.R. China.,University of Chinese Academy of Sciences, Beijing, 100049, P.R. China
| | - Chao Li
- Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, P.R. China
| | - Xuesong Wang
- Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, P.R. China.,University of Chinese Academy of Sciences, Beijing, 100049, P.R. China
| | - Qianxiong Zhou
- Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, P.R. China
| |
Collapse
|
36
|
Li S, Wang M, Chen S, Ampomah-Wireko M, Gao C, Xia Z, Nininahazwe L, Qin S, Zhang E. Development of biaromatic core-linked antimicrobial peptide mimics: Substituent position significantly affects antibacterial activity and hemolytic toxicity. Eur J Med Chem 2023; 247:115029. [PMID: 36549113 DOI: 10.1016/j.ejmech.2022.115029] [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: 11/01/2022] [Revised: 12/04/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022]
Abstract
The development of bacterial resistance to the majority of clinically significant antimicrobials has made it more difficult to treat bacterial infections with conventional antibiotics. As part of ongoing research on antimicrobial peptide mimetics, a series of quaternary ammonium cationic compounds with various linkers were designed and synthesized, with some demonstrating high antibacterial activity against Gram-negative and Gram-positive bacteria. The structure-activity relationship study revealed that the spatial position of substituents had a significant impact on antibacterial activity and hemolytic toxicity. The best compound, 3e, has good antibacterial activity against Staphylococcus aureus [minimum inhibitory concentration (MIC = 1 μg/mL)] and the least hemolytic toxicity [hemolytic concentration (HC50 = 905 μg/mL)], is stable in mammalian body fluids, and rarely induces bacterial resistance. The mechanism study revealed that the membrane action mode may be its potential bactericidal mechanism, and it can effectively cause the accumulation of intracellular reactive oxygen species (ROS) for killing bacteria. Importantly, 3e can effectively reduce the load of methicillin-resistant Staphylococcus aureus (MRSA) in mouse skin and has a higher in vivo bactericidal efficiency than vancomycin. These findings highlight the significance of divergent linkers in quaternary ammonium cations as antimicrobial peptide mimics and the potential of these cations to treat bacterial infections.
Collapse
Affiliation(s)
- Sen Li
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Meng Wang
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Shengcong Chen
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Maxwell Ampomah-Wireko
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Chen Gao
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Ziwei Xia
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Lauraine Nininahazwe
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Shangshang Qin
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou, 450001, PR China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province, Zhengzhou, 450001, PR China.
| | - En Zhang
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou, 450001, PR China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province, Zhengzhou, 450001, PR China.
| |
Collapse
|
37
|
Li H, Hsieh K, Wong PK, Mach KE, Liao JC, Wang TH. Single-cell pathogen diagnostics for combating antibiotic resistance. NATURE REVIEWS. METHODS PRIMERS 2023; 3:6. [PMID: 39917628 PMCID: PMC11800871 DOI: 10.1038/s43586-022-00190-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 12/06/2022] [Indexed: 02/09/2025]
Abstract
Bacterial infections and antimicrobial resistance are a major cause for morbidity and mortality worldwide. Antimicrobial resistance often arises from antimicrobial misuse, where physicians empirically treat suspected bacterial infections with broad-spectrum antibiotics until standard culture-based diagnostic tests can be completed. There has been a tremendous effort to develop rapid diagnostics in support of the transition from empirical treatment of bacterial infections towards a more precise and personalized approach. Single-cell pathogen diagnostics hold particular promise, enabling unprecedented quantitative precision and rapid turnaround times. This Primer provides a guide for assessing, designing, implementing and applying single-cell pathogen diagnostics. First, single-cell pathogen diagnostic platforms are introduced based on three essential capabilities: cell isolation, detection assay and output measurement. Representative results, common analysis methods and key applications are highlighted, with an emphasis on initial screening of bacterial infection, bacterial species identification and antimicrobial susceptibility testing. Finally, the limitations of existing platforms are discussed, with perspectives offered and an outlook towards clinical deployment. This Primer hopes to inspire and propel new platforms that can realize the vision of precise and personalized bacterial infection treatments in the near future.
Collapse
Affiliation(s)
- Hui Li
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, USA
- Present address: School of Electrical, Computer and Biomedical Engineering, Southern Illinois University, Carbondale, IL, USA
- These authors contributed equally: Hui Li, Kuangwen Hsieh
| | - Kuangwen Hsieh
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, USA
- These authors contributed equally: Hui Li, Kuangwen Hsieh
| | - Pak Kin Wong
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, USA
| | - Kathleen E. Mach
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | - Joseph C. Liao
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | - Tza-Huei Wang
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
38
|
Khan FA, Yaqoob S, Ali S, Tanveer N, Wang Y, Ashraf S, Hasan KA, Khalifa SAM, Shou Q, Ul-Haq Z, Jiang ZH, El-Seedi HR. Designing Functionally Substituted Pyridine-Carbohydrazides for Potent Antibacterial and Devouring Antifungal Effect on Multidrug Resistant (MDR) Strains. MOLECULES (BASEL, SWITZERLAND) 2022; 28:molecules28010212. [PMID: 36615406 PMCID: PMC9822510 DOI: 10.3390/molecules28010212] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/21/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022]
Abstract
The emergence of multidrug-resistant (MDR) pathogens and the gradual depletion of available antibiotics have exacerbated the need for novel antimicrobial agents with minimal toxicity. Herein, we report functionally substituted pyridine carbohydrazide with remarkable antimicrobial effect on multi-drug resistant strains. In the series, compound 6 had potent activity against four MDR strains of Candida spp., with minimum inhibitory concentration (MIC) values being in the range of 16-24 µg/mL and percentage inhibition up to 92.57%, which was exceptional when compared to broad-spectrum antifungal drug fluconazole (MIC = 20 µg/mL, 81.88% inhibition). Substitution of the octyl chain in 6 with a shorter butyl chain resulted in a significant anti-bacterial effect of 4 against Pseudomonas aeruginosa (ATCC 27853), the MIC value being 2-fold superior to the standard combination of ampicillin/cloxacillin. Time-kill kinetics assays were used to discern the efficacy and pharmacodynamics of the potent compounds. Further, hemolysis tests confirmed that both compounds had better safety profiles than the standard drugs. Besides, molecular docking simulations were used to further explore their mode of interaction with target proteins. Overall results suggest that these compounds have the potential to become promising antimicrobial drugs against MDR strains.
Collapse
Affiliation(s)
- Farooq-Ahmad Khan
- Third World Center for Science and Technology, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
- Correspondence: (F.-A.K.); (K.A.H.); (H.R.E.-S.)
| | - Sana Yaqoob
- Third World Center for Science and Technology, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Shujaat Ali
- Third World Center for Science and Technology, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Nimra Tanveer
- Third World Center for Science and Technology, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Yan Wang
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Sajda Ashraf
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Khwaja Ali Hasan
- Molecular and Structural Biology Research Laboratory, Department of Biochemistry, University of Karachi, Karachi 75270, Pakistan
- Correspondence: (F.-A.K.); (K.A.H.); (H.R.E.-S.)
| | - Shaden A. M. Khalifa
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, S-106 91 Stockholm, Sweden
| | - Qiyang Shou
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Zaheer Ul-Haq
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Zi-Hua Jiang
- Department of Chemistry, Lakehead University, 955 Oliver Road, Thunder Bay, Ontario P7B 5E1, Canada
| | - Hesham R. El-Seedi
- International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang 212013, China
- Pharmacognosy Group, Department of Pharmaceutical Biosciences, BMC, Uppsala University, Box 591, SE-751 24 Uppsala, Sweden
- International Joint Research Laboratory of Intelligent Agriculture and Agri-Products Processing (Jiangsu University), Jiangsu Education Department, Nanjing 210024, China
- Correspondence: (F.-A.K.); (K.A.H.); (H.R.E.-S.)
| |
Collapse
|
39
|
Yang R, Hou E, Cheng W, Yan X, Zhang T, Li S, Yao H, Liu J, Guo Y. Membrane-Targeting Neolignan-Antimicrobial Peptide Mimic Conjugates to Combat Methicillin-Resistant Staphylococcus aureus (MRSA) Infections. J Med Chem 2022; 65:16879-16892. [PMID: 36512751 DOI: 10.1021/acs.jmedchem.2c01674] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Infections caused by methicillin-resistant Staphylococcus aureus (MRSA) continue to endanger public health. Here, we report the synthesis of neolignan isomagnolone (I) and its isomer II, and the preparation of a series of novel neolignan-antimicrobial peptide (AMP) mimic conjugates. Notably, conjugates III5 and III15 exhibit potent anti-MRSA activity in vitro and in vivo, comparable to that of vancomycin, a current effective treatment for MRSA. Moreover, III5 and III15 display not only fast-killing kinetics and low resistance frequency but also low toxicity as well as effects on bacterial biofilms. Mechanism studies reveal that III5 and III15 exhibit rapid bactericidal effects through binding to the phosphatidylglycerol (PG) and cardiolipin (CL) of the bacterial membrane, thereby disrupting the cell membranes and allowing increased reactive oxygen species (ROS) as well as protein and DNA leakage. The results indicate that these neolignan-AMP mimic conjugates could be promising antimicrobial candidates for combating MRSA infections.
Collapse
Affiliation(s)
- Ruige Yang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Enhua Hou
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Wanqing Cheng
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Xiaoting Yan
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Tingting Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Shihong Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China
| | - Hong Yao
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China
| | - Jifeng Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Yong Guo
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China
| |
Collapse
|
40
|
Sorinolu AJ, Godakhindi V, Siano P, Vivero-Escoto JL, Munir M. Influence of silver ion release on the inactivation of antibiotic resistant bacteria using light-activated silver nanoparticles. MATERIALS ADVANCES 2022; 3:9090-9102. [PMID: 36545324 PMCID: PMC9743134 DOI: 10.1039/d2ma00711h] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 11/07/2022] [Indexed: 06/17/2023]
Abstract
The widespread increase in antibiotic resistance (AR), in an extensive range of microorganisms, demands the development of alternative antimicrobials with novel non-specific low-mutation bacterial targets. Silver nanoparticles (AgNPs) and photosensitizers (PSs) are promising antimicrobial agents with broad-spectrum activity and low tendency for antimicrobial resistance development. Herein, we investigated the light-mediated oxidation of AgNPs for accelerated release of Ag+ in the antibacterial synergy of PS-AgNP conjugates using protoporphyrin IX (PpIX) as a PS. Also, the influence of polyethyleneimine (PEI) coated AgNPs in promoting antibacterial activity was examined. We synthesized, characterized and tested the antimicrobial effect of three nanoparticles: AgNPs, PpIX-AgNPs, and PEI-PpIX-AgNPs against a methicillin-resistant Staphylococcus aureus strain (MRSA) and a wild-type multidrug resistant (MDR) E. coli. PpIX-AgNPs were the most effective material achieving >7 log inactivation of MRSA and MDR E. coli. The order of bacterial log inactivation was PpIX-AgNPs > PEI-PpIX-AgNPs > AgNPs. This order correlates with the trend of Ag+ concentration released by the NPs (PpIX-AgNPs > PEI-PpIX-AgNPs > AgNPs). Our study confirms a synergistic effect between PpIX and AgNPs in the inactivation of AR pathogens with about 10-fold increase in inactivation of ARB relative to AgNPs only. The concentration of Ag+ released from NPs determined the log inactivation of MRSA and MDR E. coli more than either the phototoxic effect or the electrostatic interaction promoted by surface charge of nanoparticles with bacteria cells. All NPs showed negligible cytotoxicity to mammalian cells at the bacterial inhibitory concentration after 24 h exposure. These observations confirm the crucial role of optimized Ag+ release for enhanced performance of AgNP-based antimicrobials against AR pathogens.
Collapse
Affiliation(s)
- Adeola Julian Sorinolu
- Department of Civil and Environmental Engineering, University of North Carolina at Charlotte Charlotte NC 28223 USA +1 (704)-687-1623
| | - Varsha Godakhindi
- Department of Chemistry, University of North Carolina at Charlotte Charlotte NC 28223 USA +1 (704)-687-5239
- Nanoscale Science Program, University of North Carolina at Charlotte Charlotte NC 28223 USA
| | - Paolo Siano
- Department of Chemistry, University of North Carolina at Charlotte Charlotte NC 28223 USA +1 (704)-687-5239
| | - Juan L Vivero-Escoto
- Department of Chemistry, University of North Carolina at Charlotte Charlotte NC 28223 USA +1 (704)-687-5239
- Nanoscale Science Program, University of North Carolina at Charlotte Charlotte NC 28223 USA
| | - Mariya Munir
- Department of Civil and Environmental Engineering, University of North Carolina at Charlotte Charlotte NC 28223 USA +1 (704)-687-1623
| |
Collapse
|
41
|
Thum MD, Lu Q, Stockmaster KT, Haridas D, Fears KP, Balow RB, Lundin JG. 3D‐printable cyclic peptide loaded microporous polymers for antimicrobial wound dressing materials. POLYM ADVAN TECHNOL 2022. [DOI: 10.1002/pat.5948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Matthew D. Thum
- Chemistry Division U.S. Naval Research Laboratory Washington, DC USA
| | - Qin Lu
- Chemistry Division U.S. Naval Research Laboratory Washington, DC USA
| | | | - Dhanya Haridas
- Chemistry Division U.S. Naval Research Laboratory Washington, DC USA
| | - Kenan P. Fears
- Chemistry Division U.S. Naval Research Laboratory Washington, DC USA
| | - Robert B. Balow
- Chemistry Division U.S. Naval Research Laboratory Washington, DC USA
| | - Jeffrey G. Lundin
- Chemistry Division U.S. Naval Research Laboratory Washington, DC USA
| |
Collapse
|
42
|
Hu J, Ding Y, Tao B, Yuan Z, Yang Y, Xu K, Li X, liu P, Cai K. Surface modification of titanium substrate via combining photothermal therapy and quorum-sensing-inhibition strategy for improving osseointegration and treating biofilm-associated bacterial infection. Bioact Mater 2022; 18:228-241. [PMID: 35387171 PMCID: PMC8961458 DOI: 10.1016/j.bioactmat.2022.03.011] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 01/19/2022] [Accepted: 03/07/2022] [Indexed: 12/11/2022] Open
Abstract
Insufficient osseointegration and biofilm-associated bacterial infection are important challenges for clinical application of titanium (Ti)-based implants. Here, we constructed mesoporous polydopamine (MPDA) nanoparticles (NPs) loaded with luteolin (LUT, a quorum sensing inhibitor), which were further coated with the shell of calcium phosphate (CaP) to construct MPDA-LUT@CaP nanosystem. Then, MPDA-LUT@CaP NPs were immobilized on the surface of Ti implants. Under acidic environment of bacterial biofilm-infection, the CaP shell of MPDA-LUT@CaP NPs was rapidly degraded and released LUT, Ca2+ and PO4 3- from the surface of Ti implant. LUT could effectively inhibit and disperse biofilm. Furthermore, under near-infrared irradiation (NIR), the thermotherapy induced by the photothermal conversion effect of MPDA destroyed the integrity of the bacterial membrane, and synergistically led to protein leakage and a decrease in ATP levels. Combined with photothermal therapy (PTT) and quorum-sensing-inhibition strategy, the surface-functionalized Ti substrate had an antibacterial rate of over 95.59% against Staphylococcus aureus and the elimination rate of the formed biofilm was as high as 90.3%, so as to achieve low temperature and efficient treatment of bacterial biofilm infection. More importantly, the modified Ti implant accelerated the growth of cell and the healing process of bone tissue due to the released Ca2+ and PO4 3-. In summary, this work combined PTT with quorum-sensing-inhibition strategy provides a new idea for surface functionalization of implant for achieving effective antibacterial and osseointegration capabilities.
Collapse
Affiliation(s)
- Jingwei Hu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Yao Ding
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Bailong Tao
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Zhang Yuan
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Yulu Yang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Kun Xu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Xuan Li
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Peng liu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Kaiyong Cai
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| |
Collapse
|
43
|
El-Zemity SR, Badawy ME, Esmaiel KE, Badr MM. Synthesis, antioxidant, antimicrobial, and molecular docking studies of some N-cinnamyl phenylacetamide and N-(3,7-dimethylocta-2,6-dien-1-yl) phenylacetamide derivatives. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.133411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
44
|
Antimicrobial Activity of Lactones. Antibiotics (Basel) 2022; 11:antibiotics11101327. [PMID: 36289985 PMCID: PMC9598898 DOI: 10.3390/antibiotics11101327] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 09/23/2022] [Accepted: 09/27/2022] [Indexed: 11/17/2022] Open
Abstract
The development of bacterial resistance to antibiotics and the consequent lack of effective therapy is one of the biggest problems in modern medicine. A consequence of these processes is an urgent need to continuously design and develop novel antimicrobial agents. Among the compounds showing antimicrobial potential, lactones are a group to explore. For centuries, their antimicrobial activities have been used in folk medicine. Currently, novel lactone compounds are continuously described in the literature. Some of those structures exhibit high antimicrobial potential and some are an inspiration for design and synthesis of future drugs. This paper describes recent developments on antimicrobial lactones with smaller ring sizes, up to seven membered ε-lactones. Their isolation from natural sources, chemical synthesis, synergistic activity with antibiotics, and effects on quorum sensing are presented herein.
Collapse
|
45
|
Postek W, Pacocha N, Garstecki P. Microfluidics for antibiotic susceptibility testing. LAB ON A CHIP 2022; 22:3637-3662. [PMID: 36069631 DOI: 10.1039/d2lc00394e] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The rise of antibiotic resistance is a threat to global health. Rapid and comprehensive analysis of infectious strains is critical to reducing the global use of antibiotics, as informed antibiotic use could slow down the emergence of resistant strains worldwide. Multiple platforms for antibiotic susceptibility testing (AST) have been developed with the use of microfluidic solutions. Here we describe microfluidic systems that have been proposed to aid AST. We identify the key contributions in overcoming outstanding challenges associated with the required degree of multiplexing, reduction of detection time, scalability, ease of use, and capacity for commercialization. We introduce the reader to microfluidics in general, and we analyze the challenges and opportunities related to the field of microfluidic AST.
Collapse
Affiliation(s)
- Witold Postek
- Institute of Physical Chemistry of the Polish Academy of Sciences, ul. Kasprzaka 44/52, 01-224 Warszawa, Poland.
- Broad Institute of MIT and Harvard, Merkin Building, 415 Main St, Cambridge, MA 02142, USA.
| | - Natalia Pacocha
- Institute of Physical Chemistry of the Polish Academy of Sciences, ul. Kasprzaka 44/52, 01-224 Warszawa, Poland.
| | - Piotr Garstecki
- Institute of Physical Chemistry of the Polish Academy of Sciences, ul. Kasprzaka 44/52, 01-224 Warszawa, Poland.
| |
Collapse
|
46
|
Zhou YX, Cao XY, Peng C. Antimicrobial activity of natural products against MDR bacteria: A scientometric visualization analysis. Front Pharmacol 2022; 13:1000974. [PMID: 36225591 PMCID: PMC9548655 DOI: 10.3389/fphar.2022.1000974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 09/13/2022] [Indexed: 11/13/2022] Open
Abstract
Objective: A growing number of studies have demonstrated the antimicrobial activity of natural products against multidrug-resistant bacteria. This study aimed to apply scientometric method to explore the current status and future trends in this field. Methods: All relevant original articles and reviews for the period 1997–2021 were retrieved from the Web of Science Core Collection database. VOSviewer, a scientometric software, and an online bibliometric analysis platform were used to conduct visualization study. Results: A total of 1,267 papers were included, including 1,005 original articles and 262 reviews. The United States and India made the largest contribution in this field. The University of Dschang from Cameroon produced the most publications. Coutinho HDM, Kuete V, and Gibbons S were key researchers, as they published a great many articles and were co-cited in numerous publications. Frontiers in Microbiology and Antimicrobial Agents and Chemotherapy were the most influential journals with the highest number of publications and co-citations, respectively. “Medicinal plants”, “methicillin-resistant Staphylococcus aureus”, “biofilm”, “minimum inhibitory concentration”, and “efflux pumps” were the most frequently used keywords, so these terms are presumed to be the current hot topics. All the included keywords could be roughly divided into four major themes, of which the theme of “natural product development approach” had attracted much attention in recent years. Furthermore, “Pseudomonas aeruginosa”, “nanoparticles”, “green synthesis”, “antimicrobial peptides”, “antibiofilm”, “biosynthetic gene clusters”, and “molecular dynamics simulation” had the latest average appearance year, indicating that these topics may become the research hot spots in the coming years. Conclusion: This study performed a scientometric analysis of the antibacterial activity of natural products against multidrug-resistant bacteria from a holistic perspective. It is hoped to provide novel and useful data for scientific research, and help researchers to explore this field more intuitively and effectively.
Collapse
Affiliation(s)
- Yan-Xi Zhou
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Library, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiao-Yu Cao
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Cheng Peng
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Cheng Peng,
| |
Collapse
|
47
|
New potent ciprofloxacin-uracil conjugates as DNA gyrase and topoisomerase IV inhibitors against methicillin-resistant Staphylococcus aureus. Bioorg Med Chem 2022; 73:117004. [PMID: 36148773 DOI: 10.1016/j.bmc.2022.117004] [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: 06/19/2022] [Revised: 09/07/2022] [Accepted: 09/08/2022] [Indexed: 11/20/2022]
Abstract
A series of ciprofloxacin-uracil conjugates 5a-t were synthesized and identified by 1H NMR, 13C NMR, mass spectroscopy and elemental analyses. The antibacterial results revealed that the new derivatives exhibited better activity against Gram-positive than the Gram-negative strains; most of the target compounds exhibited good activities against S. aureus ATCC 6538. Compounds 5b and 5g possess the highest activities with MICs of 1.25 and 2.37 µM, respectively, which are more potent than the parent drug ciprofloxacin, MIC, 7.58 µM. In addition, they also exhibited potent activities against MRSA AUMC 261 with MICs, 0.031 and 0.046 µM respectively, higher than ciprofloxacin with MIC, 0.57 µM. Moreover, compounds 5b and 5g showed potent inhibitory activities against DNA gyrase (IC50 = 1.72 and 5.72 µM) and topoisomerase IV (4.36 and 7.77 µM) compared to ciprofloxacin with IC50 values 0.66 and 8.16 µM, respectively. The molecular docking study revealed that compounds 5b and 5g may formed stable interaction with the active sites of DNA gyrase and topoisomerase IV similar to ciprofloxacin. Hence, 5b and 5g are considered promising antibacterial candidated against MRSA AUMC 261 strains that requires further optimization.
Collapse
|
48
|
Wang X, Fu HY, He W, Xiang YT, Yang ZC, Kuang Y, Yang SX. Synthesis and Antibacterial Activity Evaluation of Biphenyl and Dibenzofuran Derivatives as Potential Antimicrobial Agents against Antibiotic-Resistant Bacteria. Curr Issues Mol Biol 2022; 44:4087-4099. [PMID: 36135192 PMCID: PMC9497828 DOI: 10.3390/cimb44090280] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 08/31/2022] [Accepted: 09/01/2022] [Indexed: 11/16/2022] Open
Abstract
The escalating prevalence of antibiotic-resistant bacteria has led to a serious global public health problem; therefore, there is an urgent need for the development of structurally innovative antibacterial agents. In our study, a series of biphenyl and dibenzofuran derivatives were designed and synthesized by Suzuki-coupling and demethylation reactions in moderate to excellent yields (51–94% yield). Eleven compounds exhibited potent antibacterial activities against the prevalent antibiotic-resistant Gram-positive and Gram-negative pathogens, among which compounds 4′-(trifluoromethyl)-[1,1′-biphenyl]-3,4,5-triol (6i) and 5-(9H-carbazol-2-yl) benzene-1,2,3-triol (6m) showed the most potent inhibitory activities against methicillin-resistant Staphylococcus aureus and multidrug-resistant Enterococcus faecalis with MIC (minimum inhibitory concentration) values as low as 3.13 and 6.25 μg/mL, respectively. Compounds 3′,5′-dimethyl-[1,1′-biphenyl]-3,4,4′,5-tetraol (6e), 4′-fluoro-[1,1′-biphenyl]-3,4,5-triol (6g), and 4′-(trifluoromethyl)-[1,1′-biphenyl]-3,4,5-triol (6i) showed comparable inhibitory activities with ciprofloxacin to Gram-negative bacterium carbapenems-resistant Acinetobacter baumannii. Study of the structure–activity relationship indicated that a strong electron-withdrawing group on the A ring and hydroxyl groups on the B ring of biphenyls were beneficial to their antibacterial activities, and for benzo-heterocycles, N-heterocycle exhibited optimal antibacterial activity. These results can provide novel structures of antibacterial drugs chemically different from currently known antibiotics and broaden prospects for the development of effective antibiotics against antibiotic-resistant bacteria.
Collapse
Affiliation(s)
| | | | | | | | | | - Yi Kuang
- Correspondence: (Y.K.); (S.-X.Y.)
| | | |
Collapse
|
49
|
Wang S, Zhao C, Xue B, Li C, Zhang X, Yang X, Li Y, Yang Y, Shen Z, Wang J, Qiu Z. Nanoalumina triggers the antibiotic persistence of Escherichia coli through quorum sensing regulators lrsF and qseB. JOURNAL OF HAZARDOUS MATERIALS 2022; 436:129198. [PMID: 35739728 DOI: 10.1016/j.jhazmat.2022.129198] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 05/13/2022] [Accepted: 05/19/2022] [Indexed: 06/15/2023]
Abstract
Nanomaterials with bactericidal effects might provide novel strategies against bacteria. However, some bacteria can survive despite the exposure to nanomaterials, which challenges the safety of antibacterial nanomaterials. Here, we used a high dose of antibiotics to kill the E. coli. that survived under different concentrations of nanoalumina treatment to screen persisters, and found that nanoalumina could significantly trigger persisters formation. Treatment with 50 mg/L nanoalumina for 4 h resulted in the formation of (0.084 ± 0.005) % persisters. Both reactive oxygen species (ROS) and toxin-antitoxin (TA) system were involved in persisters formation. Interestingly, RT-PCR analysis and knockout of the five genes related to ROS and TA confirmed that only hipB was associated with the formation of persisters, suggesting the involvement of other mechanisms. We further identified 73 differentially expressed genes by transcriptome sequencing and analyzed them with bioinformatics tools. We selected six candidate genes and verified that five of them closely related to quorum sensing (QS) that were involved in persisters formation, and further validated that the coexpression of QS factors lrsF and qseB was a novel pathway for persisters. Our findings provided a better understanding on the emergence of bacterial persistence and the microbial behavior under nanomaterials exposure.
Collapse
Affiliation(s)
- Shang Wang
- Department of Hygienic Toxicology And Environmental Hygiene, Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; Key Laboratory of Risk Assessment and Control for Environment & Food Safety, Tianjin 300050, China
| | - Chen Zhao
- Department of Hygienic Toxicology And Environmental Hygiene, Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; Key Laboratory of Risk Assessment and Control for Environment & Food Safety, Tianjin 300050, China
| | - Bin Xue
- Department of Hygienic Toxicology And Environmental Hygiene, Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Chenyu Li
- Department of Hygienic Toxicology And Environmental Hygiene, Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Xi Zhang
- Department of Hygienic Toxicology And Environmental Hygiene, Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Xiaobo Yang
- Department of Hygienic Toxicology And Environmental Hygiene, Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Yan Li
- Department of Hygienic Toxicology And Environmental Hygiene, Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Yanping Yang
- Department of Hygienic Toxicology And Environmental Hygiene, Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Zhiqiang Shen
- Department of Hygienic Toxicology And Environmental Hygiene, Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; Key Laboratory of Risk Assessment and Control for Environment & Food Safety, Tianjin 300050, China
| | - Jingfeng Wang
- Department of Hygienic Toxicology And Environmental Hygiene, Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; Key Laboratory of Risk Assessment and Control for Environment & Food Safety, Tianjin 300050, China
| | - Zhigang Qiu
- Department of Hygienic Toxicology And Environmental Hygiene, Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China.
| |
Collapse
|
50
|
Karanja CW, Naganna N, Abutaleb NS, Dayal N, Onyedibe KI, Aryal U, Seleem MN, Sintim HO. Isoquinoline Antimicrobial Agent: Activity against Intracellular Bacteria and Effect on Global Bacterial Proteome. Molecules 2022; 27:5085. [PMID: 36014324 PMCID: PMC9416421 DOI: 10.3390/molecules27165085] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/01/2022] [Accepted: 08/02/2022] [Indexed: 11/16/2022] Open
Abstract
A new class of alkynyl isoquinoline antibacterial compounds, synthesized via Sonogashira coupling, with strong bactericidal activity against a plethora of Gram-positive bacteria including methicillin- and vancomycin-resistant Staphylococcus aureus (S. aureus) strains is presented. HSN584 and HSN739, representative compounds in this class, reduce methicillin-resistant S. aureus (MRSA) load in macrophages, whilst vancomycin, a drug of choice for MRSA infections, was unable to clear intracellular MRSA. Additionally, both HSN584 and HSN739 exhibited a low propensity to develop resistance. We utilized comparative global proteomics and macromolecule biosynthesis assays to gain insight into the alkynyl isoquinoline mechanism of action. Our preliminary data show that HSN584 perturb S. aureus cell wall and nucleic acid biosynthesis. The alkynyl isoquinoline moiety is a new scaffold for the development of potent antibacterial agents against fatal multidrug-resistant Gram-positive bacteria.
Collapse
Affiliation(s)
- Caroline W. Karanja
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, IN 47907, USA
| | - Nimishetti Naganna
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, IN 47907, USA
| | - Nader S. Abutaleb
- Department of Comparative Pathobiology, Purdue University College of Veterinary Medicine, 625 Harrison Street, West Lafayette, IN 47907, USA
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, 1410 Prices Fork Rd, Blacksburg, VA 24061, USA
| | - Neetu Dayal
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, IN 47907, USA
| | - Kenneth I. Onyedibe
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, IN 47907, USA
- Purdue Institute of Inflammation, Immunology and Infectious Disease, West Lafayette, IN 47907, USA
| | - Uma Aryal
- Department of Comparative Pathobiology, Purdue University College of Veterinary Medicine, 625 Harrison Street, West Lafayette, IN 47907, USA
- Purdue Proteomics Facility, Bindley Bioscience Center, Purdue University, West Lafayette, IN 47907, USA
| | - Mohamed N. Seleem
- Department of Comparative Pathobiology, Purdue University College of Veterinary Medicine, 625 Harrison Street, West Lafayette, IN 47907, USA
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, 1410 Prices Fork Rd, Blacksburg, VA 24061, USA
| | - Herman O. Sintim
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, IN 47907, USA
- Purdue Institute of Inflammation, Immunology and Infectious Disease, West Lafayette, IN 47907, USA
- Institute for Drug Discovery, Purdue University, 720 Clinic Drive, West Lafayette, IN 47907, USA
| |
Collapse
|