1
|
Birkelbach J, Seyfert CE, Walesch S, Müller R. Harnessing Gram-negative bacteria for novel anti-Gram-negative antibiotics. Microb Biotechnol 2024; 17:e70032. [PMID: 39487848 PMCID: PMC11531245 DOI: 10.1111/1751-7915.70032] [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: 07/25/2024] [Accepted: 10/01/2024] [Indexed: 11/04/2024] Open
Abstract
Natural products have proven themselves as a valuable resource for antibiotics. However, in view of increasing antimicrobial resistance, there is an urgent need for new, structurally diverse agents that have the potential to overcome resistance and treat Gram-negative pathogens in particular. Historically, the search for new antibiotics was strongly focussed on the very successful Actinobacteria. On the other hand, other producer strains have been under-sampled and their potential for the production of bioactive natural products has been underestimated. In this mini-review, we highlight prominent examples of novel anti-Gram negative natural products produced by Gram-negative bacteria that are currently in lead optimisation or preclinical development. Furthermore, we will provide insights into the considerations and strategies behind the discovery of these agents and their putative applications.
Collapse
Affiliation(s)
- Joy Birkelbach
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS)Helmholtz Centre for Infection Research (HZI) and Saarland University Department of PharmacySaarbrückenGermany
- German Centre for Infection Research (DZIF), Partner Site Hannover‐BraunschweigBraunschweigGermany
| | - Carsten E. Seyfert
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS)Helmholtz Centre for Infection Research (HZI) and Saarland University Department of PharmacySaarbrückenGermany
- German Centre for Infection Research (DZIF), Partner Site Hannover‐BraunschweigBraunschweigGermany
| | - Sebastian Walesch
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS)Helmholtz Centre for Infection Research (HZI) and Saarland University Department of PharmacySaarbrückenGermany
- German Centre for Infection Research (DZIF), Partner Site Hannover‐BraunschweigBraunschweigGermany
| | - Rolf Müller
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS)Helmholtz Centre for Infection Research (HZI) and Saarland University Department of PharmacySaarbrückenGermany
- German Centre for Infection Research (DZIF), Partner Site Hannover‐BraunschweigBraunschweigGermany
| |
Collapse
|
2
|
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
|
3
|
Chu X, Wang L, Zhu Y, Feng Z, Guan Q, Song L, Luo Z. A unique cell division protein critical for the assembly of the bacterial divisome. eLife 2024; 12:RP87922. [PMID: 39361022 PMCID: PMC11449484 DOI: 10.7554/elife.87922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2024] Open
Abstract
Identification of unique essential bacterial genes is important for not only the understanding of their cell biology but also the development of new antimicrobials. Here, we report a previously unrecognized core component of the Acinetobacter baumannii divisome. Our results reveal that the protein, termed Aeg1 interacts with multiple cell division proteins, including FtsN, which is required for components of the divisome to localize to the midcell. We demonstrate that the FtsAE202K and FtsBE65A mutants effectively bypassed the need of Aeg1 by A. baumannii, as did the activation variants FtsWM254I and FtsWS274G. Our results suggest that Aeg1 is a cell division protein that arrives at the division site to initiate cell division by recruiting FtsN, which activates FtsQLB and FtsA to induce the septal peptidoglycan synthase FtsWI. The discovery of the new essential cell division protein has provided a new target for the development of antibacterial agents.
Collapse
Affiliation(s)
- Xiao Chu
- Department of Respiratory Medicine, Infectious Diseases and Pathogen Biology Center State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, The First Hospital of Jilin University, Changchun, China
| | - Lidong Wang
- Department of Respiratory Medicine, Infectious Diseases and Pathogen Biology Center State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, The First Hospital of Jilin University, Changchun, China
| | - Yiheng Zhu
- Department of Respiratory Medicine, Infectious Diseases and Pathogen Biology Center State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, The First Hospital of Jilin University, Changchun, China
| | - Zhengshan Feng
- Department of Respiratory Medicine, Infectious Diseases and Pathogen Biology Center State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, The First Hospital of Jilin University, Changchun, China
| | - Qingtian Guan
- Bioinformatics Laboratory, The First Hospital of Jilin University, Changchun, China
| | - Lei Song
- Department of Respiratory Medicine, Infectious Diseases and Pathogen Biology Center State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, The First Hospital of Jilin University, Changchun, China
| | - Zhaoqing Luo
- Department of Respiratory Medicine, Infectious Diseases and Pathogen Biology Center State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
4
|
Chen K, Zhang L, Ding Y, Sun Z, Meng J, Luo R, Zhou X, Liu L, Yang S. Activity-based protein profiling in drug/pesticide discovery: Recent advances in target identification of antibacterial compounds. Bioorg Chem 2024; 151:107655. [PMID: 39032407 DOI: 10.1016/j.bioorg.2024.107655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 06/18/2024] [Accepted: 07/14/2024] [Indexed: 07/23/2024]
Abstract
Given the escalating incidence of bacterial diseases and the challenge posed by pathogenic bacterial resistance, it is imperative to identify appropriate methodologies for conducting proteomic investigations on bacteria, and thereby promoting the target-based drug/pesticide discovery. Interestingly, a novel technology termed "activity-based protein profiling" (ABPP) has been developed to identify the target proteins of active molecules. However, few studies have summarized advancements in ABPP for identifying the target proteins in antibacterial-active compounds. In order to accelerate the discovery and development of new drug/agrochemical discovery, we provide a concise overview of ABPP and its recent applications in antibacterial agent discovery. Diversiform cases were cited to demonstrate the potential of ABPP for target identification though highlighting the design strategies and summarizing the reported target protein of antibacterial compounds. Overall, this review is an excellent reference for probe design towards antibacterial compounds, and offers a new perspective of ABPP in bactericide development.
Collapse
Affiliation(s)
- Kunlun Chen
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China
| | - Ling Zhang
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China
| | - Yue Ding
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China
| | - Zhaoju Sun
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China
| | - Jiao Meng
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China
| | - Rongshuang Luo
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China
| | - Xiang Zhou
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China.
| | - Liwei Liu
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China
| | - Song Yang
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China.
| |
Collapse
|
5
|
Zhang DJ, Yuan ZQ, Yue YX, Zhang M, Wu WJ, Yang CG, Qiu WW. Synthesis and antibacterial activities of heterocyclic ring-fused 20(S)-protopanaxadiol derivatives. Bioorg Med Chem 2024; 112:117901. [PMID: 39232465 DOI: 10.1016/j.bmc.2024.117901] [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: 07/11/2024] [Revised: 08/23/2024] [Accepted: 08/25/2024] [Indexed: 09/06/2024]
Abstract
Multidrug-resistant (MDR) bacterial infections are becoming a life-threatening issue in public health; therefore, it is urgent to develop novel antibacterial agents for treating infections caused by MDR bacteria. The 20(S)-protopanaxadiol (PPD) derivative 9 was identified as a novel antibacterial hit compound in screening of our small synthetic natural product-like (NPL) library. A series of novel PPD derivatives with heterocyclic rings fused at the C-2 and C-3 positions of the A-ring were synthesized and their antibacterial activities against Staphylococcus aureus (S. aureus) Newman strain and MDR S. aureus strains (USA300, NRS-1, NRS-70, NRS-100, NRS-108, NRS-271, XJ017, and XJ036) were evaluated. Among these compounds, quinoxaline derivative 56 (SH617) exhibited the highest activity with MICs of 0.5-4 μg/mL against the S. aureus Newman strain and the eight MDR S. aureus strains. Its antibacterial activity was comparable to that of the positive control, vancomycin. In the zebrafish, 56 revealed no obvious toxicity even at a high administered dose. In vivo, following a lethal infection induced by USA300 strains in zebrafish, 56 exhibited significantly increased survival rates in a dose-dependent manner.
Collapse
Affiliation(s)
- De-Jie Zhang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China
| | - Zi-Qi Yuan
- State Key Laboratory of Drug Research, Centre for Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yan-Xin Yue
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China
| | - Min Zhang
- Department of Laboratory Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200123, China
| | - Wen-Juan Wu
- Department of Laboratory Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200123, China
| | - Cai-Guang Yang
- State Key Laboratory of Drug Research, Centre for Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China; School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, China.
| | - Wen-Wei Qiu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China.
| |
Collapse
|
6
|
Wu J, Guo D. Systematic analysis of traditional Chinese medicine prescriptions provides new insights into drug combination therapy for pox. JOURNAL OF ETHNOPHARMACOLOGY 2024; 337:118842. [PMID: 39306210 DOI: 10.1016/j.jep.2024.118842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 08/09/2024] [Accepted: 09/17/2024] [Indexed: 09/28/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The decline in cross-protection provided by the smallpox vaccine increases the risk of infection from other poxviruses. While drug combinations are a promising management, they remain underdeveloped for poxviruses. Prior to the development of the smallpox vaccine, China had long relied on herbal medicine to combat pox and accumulated a wealth of knowledge regarding different herb combinations and symptoms related to pox. The information was documented in the form of prescriptions. AIM OF THE STUDY The extensive data of prescriptions offer the potential for uncovering commonalities underlying these prescriptions, thereby providing valuable insights into the development of drug combinations against pox. MATERIALS AND METHODS The 2344 prescriptions were collected from the LTM-TCM database and 12 traditional Chinese medicine books. Firstly, the relative frequency of citation was utilized to identify the most used herbs among these prescriptions. TCMSP and LTM-TCM databases were employed to gather information about active compounds and their targets. GeneCards and DisGeNET databases were utilized to determine the associated targets for smallpox, cowpox, chickenpox, and mpox. Subsequently, network pharmacology analysis was conducted to investigate potential pathway information related to the most used herbs. A comparison of active compounds from these herbs resulted in the identification of 29 high-frequency compounds. The functions of these compounds were elucidated through gene overlap analysis, docking, and literature review. Finally, we summarized pox-related symptoms and used fidelity levels to distinguish specific herbs for corresponding symptoms. RESULTS Based on 2344 traditional pox-related prescriptions, we identified 19 most used herbs and 64 associated bio-functional modules for poxvirus treatment, with the most significant one being immunoregulation primarily involving CD4+ regulation. We also identified 29 leads that possess anti-inflammatory, antimicrobial, and antiviral properties. These herbs and leads hold the potential for pox treatment. Additionally, docking analysis suggested that these leads could inhibit poxvirus DNA synthesis, RNA capping machinery processes, and mature poxvirus particle formation, as well as immunosuppressors. The clinical features of mpox in 2022 were found to align well with our description of symptoms related to the pox. CONCLUSION Through the analysis of 2344 prescriptions for pox treatment, we obtained a comprehensive library of the most used herbs and high-frequency compounds, along with their potential functional spectrum. These libraries served as raw resources for drug combination development, while the identified symptom patterns and specific herbs greatly enhanced our insight into diverse treatments for pox patients.
Collapse
Affiliation(s)
- Jiawei Wu
- State Key Laboratory of Agrobiotechnology and School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Dianjing Guo
- State Key Laboratory of Agrobiotechnology and School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China.
| |
Collapse
|
7
|
Ge B, Hu C, Qian Y, Tang Y, Zhang Q, Jiang S, Mu Z, Zhang M. Sinensetin interferes with Staphylococcus aureus infections by targeting staphylocoagulase and improves infection survival rates in mouse model of pneumonia. J Appl Microbiol 2024; 135:lxae235. [PMID: 39284774 DOI: 10.1093/jambio/lxae235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 07/30/2024] [Accepted: 09/14/2024] [Indexed: 09/20/2024]
Abstract
AIMS Coagulase (Coa), a crucial virulence factor of Staphylococcus aureus (S. aureus), is considered a vital target for anti-virulence strategies. The research aimed to discover a natural compound capable of inhibiting S. aureus infection by targeting the virulence factor Coa. METHODS AND RESULTS The study showed that sinensetin at a concentration of 128 μg mL-1 effectively inhibited both Coa-induced coagulation and biofilm formation in S. aureus. However, western blot results indicated that sinensetin did not impact the expression of Coa protein, suggesting that sinensetin may directly target Coa to counteract the virulence of S. aureus. Thermal shift assay results demonstrated that sinensetin enhanced the thermal stability of Coa, supporting the theory of direct binding. Molecular docking and point mutation experiments identified two key binding sites for sinensetin to Coa as R73A-Coa and R204A-Coa. In vivo studies on mice revealed that sinensetin not only reduced lung tissue damage caused by S. aureus infection, but also decreased inflammatory factors in the lung lavage fluid. Furthermore, combining sinensetin with oxacillin improved the survival rates of the Galleria mellonella and mice. CONCLUSIONS Sinensetin is a promising natural compound that acts as a direct inhibitor of Coa against S. aureus infections.
Collapse
Affiliation(s)
- Bin Ge
- Changchun University of Chinese Medicine, Changchun 130117, China
| | - Chunjie Hu
- Affiliated Hospital of Changchun University of Chinese Medicine, Changchun 130021, China
| | - Yimin Qian
- Changchun University of Chinese Medicine, Changchun 130117, China
| | - Yating Tang
- Changchun University of Chinese Medicine, Changchun 130117, China
| | - Qiuyue Zhang
- Affiliated Hospital of Changchun University of Chinese Medicine, Changchun 130021, China
| | - Shuang Jiang
- Changchun University of Chinese Medicine, Changchun 130117, China
| | - Zongyi Mu
- Affiliated Hospital of Changchun University of Chinese Medicine, Changchun 130021, China
| | - Maoyun Zhang
- Changchun University of Chinese Medicine, Changchun 130117, China
| |
Collapse
|
8
|
Lee AJ, Hammond J, Sheridan J, Swift S, Munkacsi AB, Villas-Boas SG. Antifungal Activity of Disalt of Epipyrone A from Epicoccum nigrum Likely via Disrupted Fatty Acid Elongation and Sphingolipid Biosynthesis. J Fungi (Basel) 2024; 10:597. [PMID: 39330357 PMCID: PMC11433475 DOI: 10.3390/jof10090597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 08/06/2024] [Accepted: 08/09/2024] [Indexed: 09/28/2024] Open
Abstract
Multidrug-resistant fungal pathogens and antifungal drug toxicity have challenged our current ability to fight fungal infections. Therefore, there is a strong global demand for novel antifungal molecules with the distinct mode of action and specificity to service the medical and agricultural sectors. Polyenes are a class of antifungal drugs with the broadest spectrum of activity among the current antifungal drugs. Epipyrone A, a water-soluble antifungal molecule with a unique, linear polyene structure, was isolated from the fungus Epiccocum nigrum. Since small changes in a compound structure can significantly alter its cell target and mode of action, we present here a study on the antifungal mode of action of the disalt of epipyrone A (DEA) using chemical-genetic profiling, fluorescence microscopy, and metabolomics. Our results suggest the disruption of sphingolipid/fatty acid biosynthesis to be the primary mode of action of DEA, followed by the intracellular accumulation of toxic phenolic compounds, in particular p-toluic acid (4-methylbenzoic acid). Although membrane ergosterol is known to be the main cell target for polyene antifungal drugs, we found little evidence to support that is the case for DEA. Sphingolipids, on the other hand, are known for their important roles in fungal cell physiology, and their biosynthesis has been recognized as a potential fungal-specific cell target for the development of new antifungal drugs.
Collapse
Affiliation(s)
- Alex J Lee
- School of Biological Sciences, University of Auckland, Auckland 1010, New Zealand
| | - Joseph Hammond
- School of Biological Sciences, Victoria University of Wellington, P.O. Box 600, Wellington 6140, New Zealand
| | - Jeffrey Sheridan
- School of Biological Sciences, Victoria University of Wellington, P.O. Box 600, Wellington 6140, New Zealand
| | - Simon Swift
- Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand
| | - Andrew B Munkacsi
- School of Biological Sciences, Victoria University of Wellington, P.O. Box 600, Wellington 6140, New Zealand
| | - Silas G Villas-Boas
- School of Biological Sciences, University of Auckland, Auckland 1010, New Zealand
- Luxembourg Institute of Science and Technology, Environmental Research and Innovation Department, L-4362 Esch-sur-Alzette, Luxembourg
| |
Collapse
|
9
|
Bellucci MC, Romani C, Sani M, Volonterio A. Dual Antibiotic Approach: Synthesis and Antibacterial Activity of Antibiotic-Antimicrobial Peptide Conjugates. Antibiotics (Basel) 2024; 13:783. [PMID: 39200083 PMCID: PMC11352213 DOI: 10.3390/antibiotics13080783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/16/2024] [Accepted: 08/18/2024] [Indexed: 09/01/2024] Open
Abstract
In recent years, bacterial resistance to conventional antibiotics has become a major concern in the medical field. The global misuse of antibiotics in clinics, personal use, and agriculture has accelerated this resistance, making infections increasingly difficult to treat and rendering new antibiotics ineffective more quickly. Finding new antibiotics is challenging due to the complexity of bacterial mechanisms, high costs and low financial incentives for the development of new molecular scaffolds, and stringent regulatory requirements. Additionally, innovation has slowed, with many new antibiotics being modifications of existing drugs rather than entirely new classes. Antimicrobial peptides (AMPs) are a valid alternative to small-molecule antibiotics offering several advantages, including broad-spectrum activity and a lower likelihood of inducing resistance due to their multifaceted mechanisms of action. However, AMPs face challenges such as stability issues in physiological conditions, potential toxicity to human cells, high production costs, and difficulties in large-scale manufacturing. A reliable strategy to overcome the drawbacks associated with the use of small-molecule antibiotics and AMPs is combination therapy, namely the simultaneous co-administration of two or more antibiotics or the synthesis of covalently linked conjugates. This review aims to provide a comprehensive overview of the literature on the development of antibiotic-AMP conjugates, with a particular emphasis on critically analyzing the design and synthetic strategies employed in their creation. In addition to the synthesis, the review will also explore the reported antibacterial activity of these conjugates and, where available, examine any data concerning their cytotoxicity.
Collapse
Affiliation(s)
- Maria Cristina Bellucci
- Department of Food, Environmental, and Nutritional Sciences, Università degli Studi di Milano, Via Celoria 2, 20131 Milano, Italy;
| | - Carola Romani
- Department of Chemistry, Materials, and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Via Mancinelli 7, 20131 Milano, Italy;
| | - Monica Sani
- Consiglio Nazionale delle Ricerche, Istituto di Scienze e Tecnologie Chimica “G. Natta” (SCITEC), Via Mario Bianco 9, 20131 Milano, Italy;
| | - Alessandro Volonterio
- Department of Chemistry, Materials, and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Via Mancinelli 7, 20131 Milano, Italy;
| |
Collapse
|
10
|
Park J, Kim D, Son YJ, Ciufolini MA, Clovis S, Han M, Kim LH, Shin SJ, Hwang HJ. Chemical optimization and derivatization of micrococcin p2 to target multiple bacterial infections: new antibiotics from thiopeptides. World J Microbiol Biotechnol 2024; 40:307. [PMID: 39162916 DOI: 10.1007/s11274-024-04109-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 08/09/2024] [Indexed: 08/21/2024]
Abstract
Antimicrobial resistance poses a significant threat to humanity, and the development of new antibiotics is urgently needed. Our research has focused on thiopeptide antibiotics such as micrococcin P2 (MP2) and derivatives thereof as new anti-infective agents. Thiopeptides are sulfur-rich, structurally complex substances that exhibit potent activity against Gram-positive pathogens and Mycobacteria species, including clinically resistant strains. The clinical development of thiopeptides has been hampered by the lack of efficient synthetic platforms to conduct detailed structure-activity relationship studies of these natural products. The present contribution touches upon efficient synthetic routes to MP2 that laid the groundwork for clinical translation. The medicinal chemistry campaign on MP2 has been guided by computational molecular dynamic simulations and parallel investigations to improve drug-like properties, such as enhancing the aqueous solubility and optimizing antibacterial activity. Such endeavors have enabled identification of promising lead compounds, AJ-037 and AJ-206, against Mycobacterium avium complex (MAC). Extensive in vitro studies revealed that these compounds exert potent activity against MAC species, a subspecies of non-tuberculous mycobacteria (NTM) that proliferate inside macrophages. Two additional pre-clinical candidates have been identified: AJ-024, for the treatment of Clostridioides difficile infections, and AJ-147, for methicillin-resistant Staphylococcus aureus impetigo. Both compounds compare quite favorably with current first-line treatments. In particular, the ability of AJ-147 to downregulate pro-inflammatory cytokines adds a valuable dimension to its clinical use. In light of above, these new thiopeptide derivatives are well-poised for further clinical development.
Collapse
Affiliation(s)
- Jiyun Park
- Department of Microbiology, Institute for Immunology and Immunological Disease, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Dahyun Kim
- A&J Science Co., Ltd, 80 Chumbok Ro, Dong Gu, Daegu, 41061, Republic of Korea
| | - Young-Jin Son
- A&J Science Co., Ltd, 80 Chumbok Ro, Dong Gu, Daegu, 41061, Republic of Korea
| | - Marco A Ciufolini
- Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, BC, V6K 1Z1, Canada
| | - Shyaka Clovis
- A&J Science Co., Ltd, 80 Chumbok Ro, Dong Gu, Daegu, 41061, Republic of Korea
| | - Minwoo Han
- New Drug Development Center, Daegu Gyeongbuk Medical Innovation Foundation (K-MEDI hub), 80 Chumbok Ro, Dong Gu, Daegu, 41061, Republic of Korea
| | - Lee-Han Kim
- Department of Microbiology, Institute for Immunology and Immunological Disease, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Sung Jae Shin
- Department of Microbiology, Institute for Immunology and Immunological Disease, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
| | - Hee-Jong Hwang
- A&J Science Co., Ltd, 80 Chumbok Ro, Dong Gu, Daegu, 41061, Republic of Korea.
| |
Collapse
|
11
|
Melo ECD, Pinheiro RDS, Costa BS, Lima RMTD, Dias ACS, de Jesus Aguiar Dos Santos T, Nascimento MLLBD, Sousa JMDCE, Islam MT, Melo Cavalcante AADC, El-Nashar HAS, El-Shazly M, Oliveira Filho JWGD. Allium cepa as a Toxicogenetic Investigational Tool for Plant Extracts: A Systematic Review. Chem Biodivers 2024:e202401406. [PMID: 39103292 DOI: 10.1002/cbdv.202401406] [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: 06/12/2024] [Revised: 08/03/2024] [Accepted: 08/05/2024] [Indexed: 08/07/2024]
Abstract
Toxicological studies are important to investigate the genotoxic effects of various substances. Allium cepa can be used as test model for this purpose. This review summarizes the scope and applications for this A. cepa test model. For this, an up-to-date (April 2023) literature search was made in the Science Direct, PubMed, and Web of Science databases to find published evidence on studies performed using A. cepa as a test model. Out of 3,748 studies, 74 fit the inclusion criteria. The results showed that the use of the test model A. cepa contributed considerably to measuring the toxicological potential of plant extracts, proving the efficacy of the test as a potent bioindicator of toxic effects. In addition, 27 studies used more than one test system to verify the toxicological potential of extracts and fractions. Studies have shown that the A. cepa model has the potential to replace other test systems that make use of animals and cell cultures, besides having other advantages such as low cost, ease of execution, and good conditions for the observation of chromosomes. In conclusion, the A. cepa test can be considered one of the potential biomonitoring systems in toxicological studies of crude extracts.
Collapse
Affiliation(s)
- Erisson Cruz de Melo
- Laboratory of Genetics and Molecular Biology - Federal Institute of Piauí (IFPI), 64001-270, Teresina, Piauí, Brazil
| | - Randyson da Silva Pinheiro
- Laboratory of Genetics and Molecular Biology - Federal Institute of Piauí (IFPI), 64001-270, Teresina, Piauí, Brazil
| | - Bruno Sousa Costa
- Laboratory of Genetics and Molecular Biology - Federal Institute of Piauí (IFPI), 64001-270, Teresina, Piauí, Brazil
| | - Rosália Maria Tôrres de Lima
- Laboratory of Genetics and Molecular Biology - Federal Institute of Piauí (IFPI), 64001-270, Teresina, Piauí, Brazil
- Northeast Biotechnology Network (RENORBIO), Postgraduate Program in Biotechnology, Federal University of Piauí - IFPI, 64049-550, Teresina, Piauí, Brazil
- Laboratory of Genetics and Toxicology-LAPGENIC, Post-Graduate Program in Pharmaceutical Science, Federal University of Piauí, 64049-550, Teresina, Piauí, Brazil
| | - Ana Carolina Soares Dias
- Laboratory of Genetics and Molecular Biology, Federal University of Maranhão, Bacanga, 65080-805, São Luís, Maranhão, Brazil
| | | | - Maria Luisa Lima Barreto do Nascimento
- Laboratory of Genetics and Toxicology-LAPGENIC, Post-Graduate Program in Pharmaceutical Science, Federal University of Piauí, 64049-550, Teresina, Piauí, Brazil
- Postgraduate Program in Pharmaceutical Sciences, Federal University of Piauí, 64049-550, Teresina, Piauí, Brazil
| | - João Marcelo de Castro E Sousa
- Northeast Biotechnology Network (RENORBIO), Postgraduate Program in Biotechnology, Federal University of Piauí - IFPI, 64049-550, Teresina, Piauí, Brazil
- Laboratory of Genetics and Toxicology-LAPGENIC, Post-Graduate Program in Pharmaceutical Science, Federal University of Piauí, 64049-550, Teresina, Piauí, Brazil
- Postgraduate Program in Pharmaceutical Sciences, Federal University of Piauí, 64049-550, Teresina, Piauí, Brazil
| | - Muhammad Torequl Islam
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, 8100, Gopalganj, Bangladesh
- Pharmacy Discipline, Khulna University, 9208, Khulna, Bangladesh
- Bioinformatics and Drug Innovation Laboratory, BioLuster Research Center, 8100, Gopalganj, Bangladesh
| | - Ana Amélia de Carvalho Melo Cavalcante
- Northeast Biotechnology Network (RENORBIO), Postgraduate Program in Biotechnology, Federal University of Piauí - IFPI, 64049-550, Teresina, Piauí, Brazil
- Laboratory of Genetics and Toxicology-LAPGENIC, Post-Graduate Program in Pharmaceutical Science, Federal University of Piauí, 64049-550, Teresina, Piauí, Brazil
| | - Heba A S El-Nashar
- Department of Pharmacognosy, Faculty of Pharmacy, Ain Shams University, 11566, Abbassia, Cairo, Egypt
| | - Mohamed El-Shazly
- Department of Pharmacognosy, Faculty of Pharmacy, Ain Shams University, 11566, Abbassia, Cairo, Egypt
| | - José Williams Gomes de Oliveira Filho
- Laboratory of Genetics and Molecular Biology - Federal Institute of Piauí (IFPI), 64001-270, Teresina, Piauí, Brazil
- Northeast Biotechnology Network (RENORBIO), Postgraduate Program in Biotechnology, Federal University of Piauí - IFPI, 64049-550, Teresina, Piauí, Brazil
- Laboratory of Genetics and Toxicology-LAPGENIC, Post-Graduate Program in Pharmaceutical Science, Federal University of Piauí, 64049-550, Teresina, Piauí, Brazil
| |
Collapse
|
12
|
Lam YC, Hamchand R, Mucci NC, Kauffman SJ, Dudkina N, Reagle EV, Casanova-Torres ÁM, DeCuyper J, Chen H, Song D, Thomas MG, Palm NW, Goodrich-Blair H, Crawford JM. The Xenorhabdus nematophila LrhA transcriptional regulator modulates production of γ-keto- N-acyl amides with inhibitory activity against mutualistic host nematode egg hatching. Appl Environ Microbiol 2024; 90:e0052824. [PMID: 38916293 PMCID: PMC11267870 DOI: 10.1128/aem.00528-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 06/01/2024] [Indexed: 06/26/2024] Open
Abstract
Xenorhabdus nematophila is a symbiotic Gammaproteobacterium that produces diverse natural products that facilitate mutualistic and pathogenic interactions in their nematode and insect hosts, respectively. The interplay between X. nematophila secondary metabolism and symbiosis stage is tuned by various global regulators. An example of such a regulator is the LysR-type protein transcription factor LrhA, which regulates amino acid metabolism and is necessary for virulence in insects and normal nematode progeny production. Here, we utilized comparative metabolomics and molecular networking to identify small molecule factors regulated by LrhA and characterized a rare γ-ketoacid (GKA) and two new N-acyl amides, GKA-Arg (1) and GKA-Pro (2) which harbor a γ-keto acyl appendage. A lrhA null mutant produced elevated levels of compound 1 and reduced levels of compound 2 relative to wild type. N-acyl amides 1 and 2 were shown to be selective agonists for the human G-protein-coupled receptors (GPCRs) C3AR1 and CHRM2, respectively. The CHRM2 agonist 2 deleteriously affected the hatch rate and length of Steinernema nematodes. This work further highlights the utility of exploiting regulators of host-bacteria interactions for the identification of the bioactive small molecule signals that they control. IMPORTANCE Xenorhabdus bacteria are of interest due to their symbiotic relationship with Steinernema nematodes and their ability to produce a variety of natural bioactive compounds. Despite their importance, the regulatory hierarchy connecting specific natural products and their regulators is poorly understood. In this study, comparative metabolomic profiling was utilized to identify the secondary metabolites modulated by the X. nematophila global regulator LrhA. This analysis led to the discovery of three metabolites, including an N-acyl amide that inhibited the egg hatching rate and length of Steinernema carpocapsae nematodes. These findings support the notion that X. nematophila LrhA influences the symbiosis between X. nematophila and S. carpocapsae through N-acyl amide signaling. A deeper understanding of the regulatory hierarchy of these natural products could contribute to a better comprehension of the symbiotic relationship between X. nematophila and S. carpocapsae.
Collapse
Affiliation(s)
- Yick Chong Lam
- Department of Chemistry, Yale University, New Haven, Connecticut, USA
- Institute of Biomolecular Design & Discovery, Yale University, West Haven, Connecticut, USA
| | - Randy Hamchand
- Department of Chemistry, Yale University, New Haven, Connecticut, USA
- Institute of Biomolecular Design & Discovery, Yale University, West Haven, Connecticut, USA
| | - Nicholas C. Mucci
- Department of Microbiology, University of Tennessee, Knoxville, Tennessee, USA
| | - Sarah J. Kauffman
- Department of Microbiology, University of Tennessee, Knoxville, Tennessee, USA
| | - Natavan Dudkina
- Department of Chemistry, Yale University, New Haven, Connecticut, USA
- Institute of Biomolecular Design & Discovery, Yale University, West Haven, Connecticut, USA
| | - Emily V. Reagle
- Institute of Biomolecular Design & Discovery, Yale University, West Haven, Connecticut, USA
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | - Jessica DeCuyper
- Department of Microbiology, University of Tennessee, Knoxville, Tennessee, USA
| | - Haiwei Chen
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Deguang Song
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Michael G. Thomas
- Department of Bacteriology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Noah W. Palm
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Heidi Goodrich-Blair
- Department of Microbiology, University of Tennessee, Knoxville, Tennessee, USA
- Department of Bacteriology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Jason M. Crawford
- Department of Chemistry, Yale University, New Haven, Connecticut, USA
- Institute of Biomolecular Design & Discovery, Yale University, West Haven, Connecticut, USA
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
13
|
Sun N, Zhi Z, Xiao T, Deng X, He T, Dong W, Feng S, Chen S, Wong WL, Yuan W. The study of honokiol as a natural product-based antimicrobial agent and its potential interaction with FtsZ protein. Front Microbiol 2024; 15:1361508. [PMID: 39104591 PMCID: PMC11298477 DOI: 10.3389/fmicb.2024.1361508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 07/10/2024] [Indexed: 08/07/2024] Open
Abstract
Multidrug resistant bacteria have been a global health threat currently and frontline clinical treatments for these infections are very limited. To develop potent antibacterial agents with new bactericidal mechanisms is thus needed urgently to address this critical antibiotic resistance challenge. Natural products are a treasure of small molecules with high bioactive and low toxicity. In the present study, we demonstrated that a natural compound, honokiol, showed potent antibacterial activity against a number of Gram-positive bacteria including MRSA and VRE. Moreover, honokiol in combination with clinically used β-lactam antibiotics exhibits strong synergistic antimicrobial effects against drug-resistant S. aureus strains. Biochemical studies further reveal that honokiol may disrupt the GTPase activity, FtsZ polymerization, cell division. These biological impacts induced by honokiol may ultimately cause bacterial cell death. The in vivo antibacterial activity of honokiol against S. aureus infection was also verified with a biological model of G. mellonella larvae. The in vivo results support that honokiol is low toxic against the larvae and effectively increases the survival rate of the larvae infected with S. aureus. These findings demonstrate the potential of honokiol for further structural advancement as a new class of antibacterial agents with high potency against multidrug-resistant bacteria.
Collapse
Affiliation(s)
- Ning Sun
- Guangzhou 11th People's Hospital, Guangzhou Cadre and Talent Health Management Center, Guangzhou, China
| | - Ziling Zhi
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, King Med School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, China
| | - Ting Xiao
- Guangzhou 11th People's Hospital, Guangzhou Cadre and Talent Health Management Center, Guangzhou, China
| | - Xin Deng
- Guangzhou 11th People's Hospital, Guangzhou Cadre and Talent Health Management Center, Guangzhou, China
| | - Tenghui He
- Guangzhou 11th People's Hospital, Guangzhou Cadre and Talent Health Management Center, Guangzhou, China
| | - Wanyang Dong
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, King Med School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, China
| | - Shuyi Feng
- Guangzhou 11th People's Hospital, Guangzhou Cadre and Talent Health Management Center, Guangzhou, China
| | - Sisi Chen
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, King Med School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, China
| | - Wing-Leung Wong
- State Key Laboratory of Chemical Biology and Drug Discovery, and Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Wenchang Yuan
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, King Med School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
14
|
Song H, Cheng Z, Qin R, Chen Z, Wang T, Wang Y, Jiang H, Du Y, Wu F. Iron/Molybdenum Sulfide Nanozyme Cocatalytic Fenton Reaction for Photothermal/Chemodynamic Efficient Wound Healing. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:14346-14354. [PMID: 38953474 DOI: 10.1021/acs.langmuir.4c00922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
The issue of bacterial infectious diseases remains a significant concern worldwide, particularly due to the misuse of antibiotics, which has caused the emergence of antibiotic-resistant strains. Fortunately, the rapid development of nanomaterials has propelled significant progress in antimicrobial therapy, offering promising solutions. Among them, the utilization of nanoenzyme-based chemodynamic therapy (CDT) has become a highly hopeful approach to combating bacterial infectious diseases. Nevertheless, the application of CDT appears to be facing certain constraints for its low efficiency in the Fenton reaction at the infected site. In this study, we have successfully synthesized a versatile nanozyme, which was a composite of molybdenum sulfide (MoS2) and iron sulfide (FeS2), through the hydrothermal method. The results showed that iron/molybdenum sulfide nanozymes (Fe/Mo SNZs) with desirable peroxidase (POD) mimic activity can generate cytotoxic reactive oxygen species (ROS) by successfully triggering the Fenton reaction. The presence of MoS2 significantly accelerates the conversion of Fe2+/Fe3+ through a cocatalytic reaction that involves the participation of redox pairs of Mo4+/Mo6+, thereby enhancing the efficiency of CDT. Additionally, based on the excellent photothermal performance of Fe/Mo SNZs, a near-infrared (NIR) laser was used to induce localized temperature elevation for photothermal therapy (PTT) and enhance the POD-like nanoenzymatic activity. Notably, both in vitro and in vivo results demonstrated that Fe/Mo SNZs with good broad-spectrum antibacterial properties can help eradicate Gram-negative bacteria like Escherichia coli and Gram-positive bacteria like Staphylococcus aureus. The most exciting thing is that the synergistic PTT/CDT exhibited astonishing antibacterial ability and can achieve complete elimination of bacteria, which promoted wound healing after infection. Overall, this study presents a synergistic PTT/CDT strategy to address antibiotic resistance, providing avenues and directions for enhancing the efficacy of wound healing treatments and offering promising prospects for further clinical use in the near future.
Collapse
Affiliation(s)
- Huiping Song
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education; International Joint Laboratory for Drug Target of Critical Illnesses; School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Zheng Cheng
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University; State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases; Jiangsu Province Engineering Research Centre of Stomatological Translational Medicine, Nanjing Medical University, Nanjing ,Jiangsu 210029, China
| | - Ran Qin
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University; State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases; Jiangsu Province Engineering Research Centre of Stomatological Translational Medicine, Nanjing Medical University, Nanjing ,Jiangsu 210029, China
| | - Ziyu Chen
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education; International Joint Laboratory for Drug Target of Critical Illnesses; School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Tianxiao Wang
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education; International Joint Laboratory for Drug Target of Critical Illnesses; School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Yuli Wang
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University; State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases; Jiangsu Province Engineering Research Centre of Stomatological Translational Medicine, Nanjing Medical University, Nanjing ,Jiangsu 210029, China
| | - Huijun Jiang
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education; International Joint Laboratory for Drug Target of Critical Illnesses; School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Yifei Du
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University; State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases; Jiangsu Province Engineering Research Centre of Stomatological Translational Medicine, Nanjing Medical University, Nanjing ,Jiangsu 210029, China
| | - Fan Wu
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education; International Joint Laboratory for Drug Target of Critical Illnesses; School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| |
Collapse
|
15
|
Nuhamunada M, Mohite OS, Phaneuf P, Palsson B, Weber T. BGCFlow: systematic pangenome workflow for the analysis of biosynthetic gene clusters across large genomic datasets. Nucleic Acids Res 2024; 52:5478-5495. [PMID: 38686794 PMCID: PMC11162802 DOI: 10.1093/nar/gkae314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 03/22/2024] [Accepted: 04/11/2024] [Indexed: 05/02/2024] Open
Abstract
Genome mining is revolutionizing natural products discovery efforts. The rapid increase in available genomes demands comprehensive computational platforms to effectively extract biosynthetic knowledge encoded across bacterial pangenomes. Here, we present BGCFlow, a novel systematic workflow integrating analytics for large-scale genome mining of bacterial pangenomes. BGCFlow incorporates several genome analytics and mining tools grouped into five common stages of analysis such as: (i) data selection, (ii) functional annotation, (iii) phylogenetic analysis, (iv) genome mining, and (v) comparative analysis. Furthermore, BGCFlow provides easy configuration of different projects, parallel distribution, scheduled job monitoring, an interactive database to visualize tables, exploratory Jupyter Notebooks, and customized reports. Here, we demonstrate the application of BGCFlow by investigating the phylogenetic distribution of various biosynthetic gene clusters detected across 42 genomes of the Saccharopolyspora genus, known to produce industrially important secondary/specialized metabolites. The BGCFlow-guided analysis predicted more accurate dereplication of BGCs and guided the targeted comparative analysis of selected RiPPs. The scalable, interoperable, adaptable, re-entrant, and reproducible nature of the BGCFlow will provide an effective novel way to extract the biosynthetic knowledge from the ever-growing genomic datasets of biotechnologically relevant bacterial species.
Collapse
Affiliation(s)
- Matin Nuhamunada
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby 2800, Denmark
| | - Omkar S Mohite
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby 2800, Denmark
| | - Patrick V Phaneuf
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby 2800, Denmark
| | - Bernhard O Palsson
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby 2800, Denmark
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, USA
| | - Tilmann Weber
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby 2800, Denmark
| |
Collapse
|
16
|
Berida TI, Adekunle YA, Dada-Adegbola H, Kdimy A, Roy S, Sarker SD. Plant antibacterials: The challenges and opportunities. Heliyon 2024; 10:e31145. [PMID: 38803958 PMCID: PMC11128932 DOI: 10.1016/j.heliyon.2024.e31145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 05/09/2024] [Accepted: 05/10/2024] [Indexed: 05/29/2024] Open
Abstract
Nature possesses an inexhaustible reservoir of agents that could serve as alternatives to combat the growing threat of antimicrobial resistance (AMR). While some of the most effective drugs for treating bacterial infections originate from natural sources, they have predominantly been derived from fungal and bacterial species. However, a substantial body of literature is available on the promising antibacterial properties of plant-derived compounds. In this comprehensive review, we address the major challenges associated with the discovery and development of plant-derived antimicrobial compounds, which have acted as obstacles preventing their clinical use. These challenges encompass limited sourcing, the risk of agent rediscovery, suboptimal drug metabolism, and pharmacokinetics (DMPK) properties, as well as a lack of knowledge regarding molecular targets and mechanisms of action, among other pertinent issues. Our review underscores the significance of these challenges and their implications in the quest for the discovery and development of effective plant-derived antimicrobial agents. Through a critical examination of the current state of research, we give valuable insights that will advance our understanding of these classes of compounds, offering potential solutions to the global crisis of AMR. © 2017 Elsevier Inc. All rights reserved.
Collapse
Affiliation(s)
- Tomayo I. Berida
- Department of BioMolecular Sciences, Division of Pharmacognosy, University of Mississippi, University, MS, 38677, USA
| | - Yemi A. Adekunle
- Department of Pharmaceutical and Medicinal Chemistry, College of Pharmacy, Afe Babalola University, Ado-Ekiti, Nigeria
- Centre for Natural Products Discovery (CNPD), School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, James Parsons Building, Byrom Street, Liverpool, L3 3AF, United Kingdom
| | - Hannah Dada-Adegbola
- Department of Medical Microbiology and Parasitology, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Ayoub Kdimy
- LS3MN2E, CERNE2D, Faculty of Science, Mohammed V University in Rabat, Rabat, 10056, Morocco
| | - Sudeshna Roy
- Department of BioMolecular Sciences, Division of Pharmacognosy, University of Mississippi, University, MS, 38677, USA
| | - Satyajit D. Sarker
- Centre for Natural Products Discovery (CNPD), School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, James Parsons Building, Byrom Street, Liverpool, L3 3AF, United Kingdom
| |
Collapse
|
17
|
Yang J, Xu L, Zhou Y, Cui M, Liu D, Wang J, Wang Y, Deng X. Repurposing harmaline as a novel approach to reverse tmexCD1-toprJ1-mediated tigecycline resistance against klebsiella pneumoniae infections. Microb Cell Fact 2024; 23:152. [PMID: 38790017 PMCID: PMC11127330 DOI: 10.1186/s12934-024-02410-4] [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: 02/17/2024] [Accepted: 04/29/2024] [Indexed: 05/26/2024] Open
Abstract
BACKGROUND A novel plasmid-mediated resistance-nodulation-division (RND) efflux pump gene cluster tmexCD1-toprJ1 in Klebsiella pneumoniae tremendously threatens the use of convenient therapeutic options in the post-antibiotic era, including the "last-resort" antibiotic tigecycline. RESULTS In this work, the natural alkaloid harmaline was found to potentiate tigecycline efficacy (4- to 32-fold) against tmexCD1-toprJ1-positive K. pneumoniae, which also thwarted the evolution of tigecycline resistance. Galleria mellonella and mouse infection models in vivo further revealed that harmaline is a promising candidate to reverse tigecycline resistance. Inspiringly, harmaline works synergistically with tigecycline by undermining tmexCD1-toprJ1-mediated multidrug resistance efflux pump function via interactions with TMexCD1-TOprJ1 active residues and dissipation of the proton motive force (PMF), and triggers a vicious cycle of disrupting cell membrane integrity and metabolic homeostasis imbalance. CONCLUSION These results reveal the potential of harmaline as a novel tigecycline adjuvant to combat hypervirulent K. pneumoniae infections.
Collapse
Affiliation(s)
- Jindian Yang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Lei Xu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Yonglin Zhou
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, 130062, China
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western China, School of Life Sciences, Ningxia University, Yinchuan, China
| | - Minhe Cui
- Jilin Province Mushuo Livestock Farming Co., Ltd., Jilin, China
| | - Dejun Liu
- Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jianfeng Wang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, 130062, China.
| | - Yang Wang
- Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China.
| | - Xuming Deng
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, 130062, China.
| |
Collapse
|
18
|
Hofer W, Deschner F, Jézéquel G, Pessanha de Carvalho L, Abdel-Wadood N, Pätzold L, Bernecker S, Morgenstern B, Kany AM, Große M, Stadler M, Bischoff M, Hirsch AKH, Held J, Herrmann J, Müller R. Functionalization of Chlorotonils: Dehalogenil as Promising Lead Compound for In Vivo Application. Angew Chem Int Ed Engl 2024; 63:e202319765. [PMID: 38502093 DOI: 10.1002/anie.202319765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 02/15/2024] [Accepted: 02/22/2024] [Indexed: 03/20/2024]
Abstract
The natural product chlorotonil displays high potency against multidrug-resistant Gram-positive bacteria and Plasmodium falciparum. Yet, its scaffold is characterized by low solubility and oral bioavailability, but progress was recently made to enhance these properties. Applying late-stage functionalization, we aimed to further optimize the molecule. Previously unknown reactions including a sulfur-mediated dehalogenation were revealed. Dehalogenil, the product of this reaction, was identified as the most promising compound so far, as this new derivative displayed improved solubility and in vivo efficacy while retaining excellent antimicrobial activity. We confirmed superb activity against multidrug-resistant clinical isolates of Staphylococcus aureus and Enterococcus spp. and mature transmission stages of Plasmodium falciparum. We also demonstrated favorable in vivo toxicity, pharmacokinetics and efficacy in infection models with S. aureus. Taken together, these results identify dehalogenil as an advanced lead molecule.
Collapse
Affiliation(s)
- Walter Hofer
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI) and Department of Pharmacy at Saarland University Campus Building E8.1, 66123, Saarbrücken, Germany
- German Centre for Infection Research (DZIF), Braunschweig, 38124, Germany
| | - Felix Deschner
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI) and Department of Pharmacy at Saarland University Campus Building E8.1, 66123, Saarbrücken, Germany
- German Centre for Infection Research (DZIF), Braunschweig, 38124, Germany
| | - Gwenaëlle Jézéquel
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI) and Department of Pharmacy at Saarland University Campus Building E8.1, 66123, Saarbrücken, Germany
- German Centre for Infection Research (DZIF), Braunschweig, 38124, Germany
| | - Laìs Pessanha de Carvalho
- German Centre for Infection Research (DZIF), Braunschweig, 38124, Germany
- Institute of Tropical Medicine, Eberhard Karls University Tübingen, Wilhelmstraße 27, 72074, Tübingen, Germany
| | - Noran Abdel-Wadood
- Institute for Medical Microbiology and Hygiene, Saarland University, 66421, Homburg, Germany
- Institute of Anatomy and Cell Biology /, Saarland University, 66421, Homburg, Germany
| | - Linda Pätzold
- Institute for Medical Microbiology and Hygiene, Saarland University, 66421, Homburg, Germany
| | - Steffen Bernecker
- Microbial Drugs, Helmholtz Centre for Infection Research (HZI), Inhoffenstraße 7, 38124, Braunschweig, Germany
| | - Bernd Morgenstern
- Inorganic Solid State Chemistry, Saarland University Campus, 66123, Saarbrücken, Germany
| | - Andreas M Kany
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI) and Department of Pharmacy at Saarland University Campus Building E8.1, 66123, Saarbrücken, Germany
- German Centre for Infection Research (DZIF), Braunschweig, 38124, Germany
| | - Miriam Große
- German Centre for Infection Research (DZIF), Braunschweig, 38124, Germany
- Microbial Drugs, Helmholtz Centre for Infection Research (HZI), Inhoffenstraße 7, 38124, Braunschweig, Germany
| | - Marc Stadler
- German Centre for Infection Research (DZIF), Braunschweig, 38124, Germany
- Microbial Drugs, Helmholtz Centre for Infection Research (HZI), Inhoffenstraße 7, 38124, Braunschweig, Germany
| | - Markus Bischoff
- Institute for Medical Microbiology and Hygiene, Saarland University, 66421, Homburg, Germany
| | - Anna K H Hirsch
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI) and Department of Pharmacy at Saarland University Campus Building E8.1, 66123, Saarbrücken, Germany
- German Centre for Infection Research (DZIF), Braunschweig, 38124, Germany
- Helmholtz International Lab for Anti-Infectives, Saarbrücken, 66123, Germany
| | - Jana Held
- German Centre for Infection Research (DZIF), Braunschweig, 38124, Germany
- Institute of Tropical Medicine, Eberhard Karls University Tübingen, Wilhelmstraße 27, 72074, Tübingen, Germany
- Centre de Recherches Médicales de Lambaréné, Lambaréné, BP 242, BP 242, Gabon
| | - Jennifer Herrmann
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI) and Department of Pharmacy at Saarland University Campus Building E8.1, 66123, Saarbrücken, Germany
- German Centre for Infection Research (DZIF), Braunschweig, 38124, Germany
| | - Rolf Müller
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI) and Department of Pharmacy at Saarland University Campus Building E8.1, 66123, Saarbrücken, Germany
- German Centre for Infection Research (DZIF), Braunschweig, 38124, Germany
- Helmholtz International Lab for Anti-Infectives, Saarbrücken, 66123, Germany
| |
Collapse
|
19
|
Slingerland C, Martin NI. Recent Advances in the Development of Polymyxin Antibiotics: 2010-2023. ACS Infect Dis 2024; 10:1056-1079. [PMID: 38470446 PMCID: PMC11019560 DOI: 10.1021/acsinfecdis.3c00630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 03/13/2024]
Abstract
The polymyxins are nonribosomal lipopeptides produced by Paenibacillus polymyxa and are potent antibiotics with activity specifically directed against Gram-negative bacteria. While the clinical use of polymyxins has historically been limited due to their toxicity, their use is on the rise given the lack of alternative treatment options for infections due to multidrug resistant Gram-negative pathogens. The Gram-negative specificity of the polymyxins is due to their ability to target lipid A, the membrane embedded LPS anchor that decorates the cell surface of Gram-negative bacteria. Notably, the mechanisms responsible for polymyxin toxicity, and in particular their nephrotoxicity, are only partially understood with most insights coming from studies carried out in the past decade. In parallel, many synthetic and semisynthetic polymyxin analogues have been developed in recent years in an attempt to mitigate the nephrotoxicity of the natural products. Despite these efforts, to date, no polymyxin analogues have gained clinical approval. This may soon change, however, as at the moment there are three novel polymyxin analogues in clinical trials. In this context, this review provides an update of the most recent insights with regard to the structure-activity relationships and nephrotoxicity of new polymyxin variants reported since 2010. We also discuss advances in the synthetic methods used to generate new polymyxin analogues, both via total synthesis and semisynthesis.
Collapse
Affiliation(s)
- Cornelis
J. Slingerland
- Biological
Chemistry Group, Institute of Biology Leiden, Leiden University, Sylviusweg 72, 2333 BE Leiden, The Netherlands
| | - Nathaniel I. Martin
- Biological
Chemistry Group, Institute of Biology Leiden, Leiden University, Sylviusweg 72, 2333 BE Leiden, The Netherlands
| |
Collapse
|
20
|
de Crécy-Lagard V, Hutinet G, Cediel-Becerra JDD, Yuan Y, Zallot R, Chevrette MG, Ratnayake RMMN, Jaroch M, Quaiyum S, Bruner S. Biosynthesis and function of 7-deazaguanine derivatives in bacteria and phages. Microbiol Mol Biol Rev 2024; 88:e0019923. [PMID: 38421302 PMCID: PMC10966956 DOI: 10.1128/mmbr.00199-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024] Open
Abstract
SUMMARYDeazaguanine modifications play multifaceted roles in the molecular biology of DNA and tRNA, shaping diverse yet essential biological processes, including the nuanced fine-tuning of translation efficiency and the intricate modulation of codon-anticodon interactions. Beyond their roles in translation, deazaguanine modifications contribute to cellular stress resistance, self-nonself discrimination mechanisms, and host evasion defenses, directly modulating the adaptability of living organisms. Deazaguanine moieties extend beyond nucleic acid modifications, manifesting in the structural diversity of biologically active natural products. Their roles in fundamental cellular processes and their presence in biologically active natural products underscore their versatility and pivotal contributions to the intricate web of molecular interactions within living organisms. Here, we discuss the current understanding of the biosynthesis and multifaceted functions of deazaguanines, shedding light on their diverse and dynamic roles in the molecular landscape of life.
Collapse
Affiliation(s)
- Valérie de Crécy-Lagard
- Department of Microbiology and Cell Science, University of Florida, Gainesville, Florida, USA
- University of Florida Genetics Institute, Gainesville, Florida, USA
| | - Geoffrey Hutinet
- Department of Biology, Haverford College, Haverford, Pennsylvania, USA
| | | | - Yifeng Yuan
- Department of Microbiology and Cell Science, University of Florida, Gainesville, Florida, USA
| | - Rémi Zallot
- Department of Life Sciences, Manchester Metropolitan University, Manchester, United Kingdom
| | - Marc G. Chevrette
- Department of Microbiology and Cell Science, University of Florida, Gainesville, Florida, USA
| | | | - Marshall Jaroch
- Department of Microbiology and Cell Science, University of Florida, Gainesville, Florida, USA
| | - Samia Quaiyum
- Department of Microbiology and Cell Science, University of Florida, Gainesville, Florida, USA
| | - Steven Bruner
- Department of Chemistry, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
21
|
Meyer KJ, Nodwell JR. Streptomyces extracellular vesicles are a broad and permissive antimicrobial packaging and delivery system. J Bacteriol 2024; 206:e0032523. [PMID: 38353531 PMCID: PMC10955852 DOI: 10.1128/jb.00325-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 01/29/2024] [Indexed: 03/22/2024] Open
Abstract
Streptomyces are the primary source of bioactive specialized metabolites used in research and medicine, including many antimicrobials. These are presumed to be secreted and function as freely soluble compounds. However, increasing evidence suggests that extracellular vesicles are an alternative secretion system. We assessed environmental and lab-adapted Streptomyces (sporulating filamentous actinomycetes) and found frequent production of antimicrobial vesicles. The molecular cargo included actinomycins, anthracyclines, candicidin, and actinorhodin, reflecting both diverse chemical properties and diverse antibacterial and antifungal activity. The levels of packaged antimicrobials correlated with the level of inhibitory activity of the vesicles, and a strain knocked out for the production of anthracyclines produced vesicles that lacked antimicrobial activity. We demonstrated that antimicrobial containing vesicles achieve direct delivery of the cargo to other microbes. Notably, this delivery via membrane fusion occurred to a broad range of microbes, including pathogenic bacteria and yeast. Vesicle encapsulation offers a broad and permissive packaging and delivery system for antimicrobial specialized metabolites, with important implications for ecology and translation.IMPORTANCEExtracellular vesicle encapsulation changes our picture of how antimicrobial metabolites function in the environment and provides an alternative translational approach for the delivery of antimicrobials. We find many Streptomyces strains are capable of releasing antimicrobial vesicles, and at least four distinct classes of compounds can be packaged, suggesting this is widespread in nature. This is a striking departure from the primary paradigm of the secretion and action of specialized metabolites as soluble compounds. Importantly, the vesicles deliver antimicrobial metabolites directly to other microbes via membrane fusion, including pathogenic bacteria and yeast. This suggests future applications in which lipid-encapsulated natural product antibiotics and antifungals could be used to solve some of the most pressing problems in drug resistance.
Collapse
Affiliation(s)
- Kirsten J. Meyer
- Department of Biochemistry, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Justin R. Nodwell
- Department of Biochemistry, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
22
|
Shen T, Guo J, Han Z, Zhang G, Liu Q, Si X, Wang D, Wu S, Xia J. AutoMolDesigner for Antibiotic Discovery: An AI-Based Open-Source Software for Automated Design of Small-Molecule Antibiotics. J Chem Inf Model 2024; 64:575-583. [PMID: 38265916 DOI: 10.1021/acs.jcim.3c01562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2024]
Abstract
Discovery of small-molecule antibiotics with novel chemotypes serves as one of the essential strategies to address antibiotic resistance. Although a considerable number of computational tools committed to molecular design have been reported, there is a deficit in holistic and efficient tools specifically developed for small-molecule antibiotic discovery. To address this issue, we report AutoMolDesigner, a computational modeling software dedicated to small-molecule antibiotic design. It is a generalized framework comprising two functional modules, i.e., generative-deep-learning-enabled molecular generation and automated machine-learning-based antibacterial activity/property prediction, wherein individually trained models and curated datasets are out-of-the-box for whole-cell-based antibiotic screening and design. It is open-source, thus allowing for the incorporation of new features for flexible use. Unlike most software programs based on Linux and command lines, this application equipped with a Qt-based graphical user interface can be run on personal computers with multiple operating systems, making it much easier to use for experimental scientists. The software and related materials are freely available at GitHub (https://github.com/taoshen99/AutoMolDesigner) and Zenodo (https://zenodo.org/record/10097899).
Collapse
Affiliation(s)
- Tao Shen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Jiale Guo
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Zunsheng Han
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Gao Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Qingxin Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- School of Pharmacy, Jiangsu Ocean University, Lianyungang, Jiangsu 222005, China
| | - Xinxin Si
- School of Pharmacy, Jiangsu Ocean University, Lianyungang, Jiangsu 222005, China
| | - Dongmei Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Song Wu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Jie Xia
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
23
|
Ramírez-Rendón D, Guzmán-Chávez F, García-Ausencio C, Rodríguez-Sanoja R, Sánchez S. The untapped potential of actinobacterial lanthipeptides as therapeutic agents. Mol Biol Rep 2023; 50:10605-10616. [PMID: 37934370 PMCID: PMC10676316 DOI: 10.1007/s11033-023-08880-w] [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: 08/26/2023] [Accepted: 10/03/2023] [Indexed: 11/08/2023]
Abstract
The increase in bacterial resistance generated by the indiscriminate use of antibiotics in medical practice set new challenges for discovering bioactive natural products as alternatives for therapeutics. Lanthipeptides are an attractive natural product group that has been only partially explored and shows engaging biological activities. These molecules are small peptides with potential application as therapeutic agents. Some members show antibiotic activity against problematic drug-resistant pathogens and against a wide variety of viruses. Nevertheless, their biological activities are not restricted to antimicrobials, as their contribution to the treatment of cystic fibrosis, cancer, pain symptoms, control of inflammation, and blood pressure has been demonstrated. The study of biosynthetic gene clusters through genome mining has contributed to accelerating the discovery, enlargement, and diversification of this group of natural products. In this review, we provide insight into the recent advances in the development and research of actinobacterial lanthipeptides that hold great potential as therapeutics.
Collapse
Affiliation(s)
- Dulce Ramírez-Rendón
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, CDMX, 04510, Mexico City, México
| | - Fernando Guzmán-Chávez
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, CDMX, 04510, Mexico City, México
| | - Carlos García-Ausencio
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, CDMX, 04510, Mexico City, México
| | - Romina Rodríguez-Sanoja
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, CDMX, 04510, Mexico City, México
| | - Sergio Sánchez
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, CDMX, 04510, Mexico City, México.
| |
Collapse
|
24
|
Cesaro A, de la Fuente-Nunez C. Antibiotic identified by AI. Nat Chem Biol 2023; 19:1296-1298. [PMID: 37821719 PMCID: PMC11537272 DOI: 10.1038/s41589-023-01448-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2023]
Abstract
Computational approaches are emerging as powerful tools for the discovery of antibiotics. A study now uses machine learning to discover abaucin, a potent antibiotic that targets the bacterial pathogen Acinetobacter baumannii .
Collapse
Affiliation(s)
- Angela Cesaro
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA
- Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Cesar de la Fuente-Nunez
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA.
- Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
25
|
Zhang QW, Ren J, Lu JX, Chen XY, He XJ, Wang Q, Zhou ZD, Jin Z, Zeng ZL, Tang YZ. Design, synthesis, and biological evaluation of novel pleuromutilin derivatives containing benzimidazoles as effective anti-MRSA agents. Drug Dev Res 2023; 84:1437-1452. [PMID: 37534779 DOI: 10.1002/ddr.22095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 07/16/2023] [Indexed: 08/04/2023]
Abstract
A series of pleuromutilin derivatives containing benzimidazole were designed, synthesized, and evaluated for their antibacterial activities against Methicillin-resistant Staphylococcus aureus (MRSA) in this study. The in vitro antibacterial activities of the synthesized derivatives against four strains of S. aureus (MRSA ATCC 43300, S. aureus ATCC 29213, S. aureus 144, and S. aureus AD3) were determined by the broth dilution method. Among these derivatives, compound 58 exhibited superior in vitro antibacterial effect against MRSA (minimal inhibitory concentration [MIC] = 0.0625 μg/mL) than tiamulin (MIC = 0.5 μg/mL). Compound 58 possessed a faster bactericidal kinetic and a longer post-antibiotic effect time against MRSA than tiamulin. Meanwhile, at 8 μg/mL concentration, compound 58 did not display obviously cytotoxic effect on the RAW 264.7 cells. In addition, compound 58 (-2.04 log10 CFU/mL) displayed superior in vivo antibacterial efficacy than tiamulin (-1.02 log10 CFU/mL) in reducing MRSA load in mice thigh infection model. In molecular docking study, compound 58 can successfully attach to the 50S ribosomal active site (the binding free energy is -8.11 kcal/mol). Therefore, compound 58 was a potential antibacterial candidate for combating MRSA infections.
Collapse
Affiliation(s)
- Qi-Wen Zhang
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Jie Ren
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Jia-Xun Lu
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Xiao-Ying Chen
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Xian-Jin He
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Qi Wang
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Zi-Dan Zhou
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Zhen Jin
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - Zhen-Ling Zeng
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - You-Zhi Tang
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| |
Collapse
|
26
|
Kim D, Lee J, Shyaka C, Kwak JH, Pai H, Rho M, Ciufolini MA, Han M, Park JH, Kim YR, Jung S, Jang AR, Kim E, Lee JY, Lee H, Son YJ, Hwang HJ. Identification of Micrococcin P2-Derivatives as Antibiotic Candidates against Two Gram-Positive Pathogens. J Med Chem 2023; 66:14263-14277. [PMID: 37796116 DOI: 10.1021/acs.jmedchem.3c01309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/06/2023]
Abstract
Thiopeptides exhibit potent antimicrobial activity against Gram-positive pathogens by inhibiting bacterial protein synthesis. Micrococcins are among the structurally simpler thiopeptides, but they have not been exploited in detail. This research involved a computational simulation of micrococcin P2 (MP2) docking in parallel with the structure-activity relationship (SAR) studied. The incorporation of particular nitrogen heterocycles in the side chain of MP2 enhances the antimicrobial activity. Micrococcin analogues 6c and 6d thus proved to be more effective against impetigo and C. difficile infection (CDI), respectively, as compared to current first-line treatments. Compound 6c also showed a shorter treatment period than that of a first-line treatment for impetigo. This may be attributed to its ability to downregulate pro-inflammatory cytokines. Compound 6d had no observed recurrence for C. difficile and exerted a minimal impact on the beneficial gut microbiome. Their pharmacokinetic properties and low toxicity profile make these compounds ideal candidates for the treatment of impetigo and CDI and validate their involvement in preclinical development.
Collapse
Affiliation(s)
- Dahyun Kim
- A&J Science Co., Ltd., 80 Chumbok Ro, Dong Gu, Daegu 41061, Republic of Korea
| | - Jusuk Lee
- A&J Science Co., Ltd., 80 Chumbok Ro, Dong Gu, Daegu 41061, Republic of Korea
| | - Clovis Shyaka
- A&J Science Co., Ltd., 80 Chumbok Ro, Dong Gu, Daegu 41061, Republic of Korea
| | - Jin-Hwan Kwak
- School of Life Science, Handong Global University, 558 Handong Ro, Heunghae-Eup, Buk-Gu, Pohang 37554, Republic of Korea
- Office of the President, Sunlin University, 30, 36 Chogok-gil, Heunghae-Eup, Buk-Gu, Pohang 37560, Republic of Korea
| | - Hyunjoo Pai
- Department of Internal Medicine, Hanyang University College of Medicine, 232 Wangsimri Ro, Seongdong-Gu, Seoul 04763, Republic of Korea
| | - Mina Rho
- Department of Computer Science, Hanyang University, 222 Wangsimri Ro, Seongdong-Gu, Seoul 04763, Republic of Korea
| | - Marco A Ciufolini
- Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, BC V6K 1Z1, Canada
| | - Minwoo Han
- New Drug Development Center, Daegu Gyeongbuk Medical Innovation Foundation, 80 Chumbok Ro, Dong Gu, Daegu 41061, Republic of Korea
| | - Jong-Hwan Park
- Laboratory Animal Medicine, College of Veterinary Medicine and Animal Medical Institute, Chonnam National University, 77 Yongbong-ro, Buk-Gu, Gwangju 61186, Republic of Korea
- Nodcure Inc., 77 Yongbong-ro, Buk-Gu, Gwangju 61186, Republic of Korea
| | - Young-Rok Kim
- School of Life Science, Handong Global University, 558 Handong Ro, Heunghae-Eup, Buk-Gu, Pohang 37554, Republic of Korea
| | - Sungji Jung
- School of Life Science, Handong Global University, 558 Handong Ro, Heunghae-Eup, Buk-Gu, Pohang 37554, Republic of Korea
| | - Ah-Ra Jang
- Nodcure Inc., 77 Yongbong-ro, Buk-Gu, Gwangju 61186, Republic of Korea
| | - Eunjung Kim
- School of Life Science, Handong Global University, 558 Handong Ro, Heunghae-Eup, Buk-Gu, Pohang 37554, Republic of Korea
| | - Jee-Young Lee
- New Drug Development Center, Daegu Gyeongbuk Medical Innovation Foundation, 80 Chumbok Ro, Dong Gu, Daegu 41061, Republic of Korea
| | - Hakyeong Lee
- A&J Science Co., Ltd., 80 Chumbok Ro, Dong Gu, Daegu 41061, Republic of Korea
| | - Young-Jin Son
- A&J Science Co., Ltd., 80 Chumbok Ro, Dong Gu, Daegu 41061, Republic of Korea
| | - Hee-Jong Hwang
- A&J Science Co., Ltd., 80 Chumbok Ro, Dong Gu, Daegu 41061, Republic of Korea
| |
Collapse
|
27
|
Zheng M, Lupoli TJ. Counteracting antibiotic resistance enzymes and efflux pumps. Curr Opin Microbiol 2023; 75:102334. [PMID: 37329679 DOI: 10.1016/j.mib.2023.102334] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/02/2023] [Accepted: 05/17/2023] [Indexed: 06/19/2023]
Abstract
Bacterial pathogens are constantly evolving new resistance mechanisms against antibiotics; hence, strategies to potentiate existing antibiotics or combat mechanisms of resistance using adjuvants are always in demand. Recently, inhibitors have been identified that counteract enzymatic modification of the drugs isoniazid and rifampin, which have implications in the study of multi-drug-resistant mycobacteria. A wealth of structural studies on efflux pumps from diverse bacteria has also fueled the design of new small-molecule and peptide-based agents to prevent the active transport of antibiotics. We envision that these findings will inspire microbiologists to apply existing adjuvants to clinically relevant resistant strains, or to use described platforms to discover novel antibiotic adjuvant scaffolds.
Collapse
Affiliation(s)
- Meng Zheng
- Department of Chemistry, New York University, 100 Washington Square East, New York, NY 10003, USA
| | - Tania J Lupoli
- Department of Chemistry, New York University, 100 Washington Square East, New York, NY 10003, USA.
| |
Collapse
|
28
|
Pathak RK, Kim JM. Identification of histidine kinase inhibitors through screening of natural compounds to combat mastitis caused by Streptococcus agalactiae in dairy cattle. J Biol Eng 2023; 17:59. [PMID: 37752501 PMCID: PMC10523694 DOI: 10.1186/s13036-023-00378-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 09/18/2023] [Indexed: 09/28/2023] Open
Abstract
BACKGROUND Mastitis poses a major threat to dairy farms globally; it results in reduced milk production, increased treatment costs, untimely compromised genetic potential, animal deaths, and economic losses. Streptococcus agalactiae is a highly virulent bacteria that cause mastitis. The administration of antibiotics for the treatment of this infection is not advised due to concerns about the emergence of antibiotic resistance and potential adverse effects on human health. Thus, there is a critical need to identify new therapeutic approaches to combat mastitis. One promising target for the development of antibacterial therapies is the transmembrane histidine kinase of bacteria, which plays a key role in signal transduction pathways, secretion systems, virulence, and antibiotic resistance. RESULTS In this study, we aimed to identify novel natural compounds that can inhibit transmembrane histidine kinase. To achieve this goal, we conducted a virtual screening of 224,205 natural compounds, selecting the top ten based on their lowest binding energy and favorable protein-ligand interactions. Furthermore, molecular docking of eight selected antibiotics and five histidine kinase inhibitors with transmembrane histidine kinase was performed to evaluate the binding energy with respect to top-screened natural compounds. We also analyzed the ADMET properties of these compounds to assess their drug-likeness. The top two compounds (ZINC000085569031 and ZINC000257435291) and top-screened antibiotics (Tetracycline) that demonstrated a strong binding affinity were subjected to molecular dynamics simulations (100 ns), free energy landscape, and binding free energy calculations using the MM-PBSA method. CONCLUSION Our results suggest that the selected natural compounds have the potential to serve as effective inhibitors of transmembrane histidine kinase and can be utilized for the development of novel antibacterial veterinary medicine for mastitis after further validation through clinical studies.
Collapse
Affiliation(s)
- Rajesh Kumar Pathak
- Department of Animal Science and Technology, Chung-Ang University, Anseong-si, Gyeonggi-do, 17546, Republic of Korea
| | - Jun-Mo Kim
- Department of Animal Science and Technology, Chung-Ang University, Anseong-si, Gyeonggi-do, 17546, Republic of Korea.
| |
Collapse
|
29
|
El-Kimary EI, Korany MA, Issa AE, Basuny MG. Green Multiplex Chromatographic Determination of Nine Penicillin Antibiotics Residues in Industrial Air Dust and Wastewater Environmental Samples. J Chromatogr Sci 2023; 61:644-655. [PMID: 35523719 DOI: 10.1093/chromsci/bmac036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Indexed: 08/22/2023]
Abstract
Determination of penicillin residues in different industrial effluents including wastewater and air samples is important to prevent exposure to residual amounts of penicillin and the development of antibiotic resistance. A green high performance liquid chromatography (HPLC) method coupled with diode array detection has been developed and validated for multiplex determination of nine penicillin antibiotics in the industrial air dust and wastewater environmental samples of penicillin facility in addition to the monitoring of facility surface cleaning. Separation was performed on C18 column with gradient elution of methanol and phosphate buffer (pH 4) at a flow rate of 1.5 mL min-1 and ultra violet (UV) detection at 220 nm. Low limits of detection were achieved (0.1-0.3 μg mL-1) indicating good sensitivity of the proposed. The method was applied for ensuring the efficiency of cleaning validation after worst-case selection. Recovery studies of the studied penicillins from fortified stainless steel and polycarbonate surfaces and swabs were between 91.91 and 100.22% with relative standard deviation 0.11-1.79%. The presence of any of the studied penicillins in wastewater samples from penicillin plant drainage was checked. Also, total air dust concentration (mg m-3) and % of penicillin active material residues in air dust were calculated from the area of the exposed group in suspension, tablet and vial production lines. The proposed method can be recommended for routine analysis of air and wastewater environmental samples for the detection of penicillin antibiotics at low levels as well as monitoring of facility surface cleaning with high accuracy and precision.
Collapse
Affiliation(s)
- Eman I El-Kimary
- Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Alexandria University, El-Messalah, Alexandria 21521, Egypt
| | - Mohammed A Korany
- Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Alexandria University, El-Messalah, Alexandria 21521, Egypt
| | - Ahmed E Issa
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, El-Messalah, Alexandria 21521, Egypt
| | - Mohamed G Basuny
- Pharco Pharmaceutical Industrial Company, Quality Control Department, Alexandria 23511, Egypt
| |
Collapse
|
30
|
Moreno Cardenas C, Çiçek SS. Structure-dependent activity of plant natural products against methicillin-resistant Staphylococcus aureus. Front Microbiol 2023; 14:1234115. [PMID: 37649631 PMCID: PMC10463185 DOI: 10.3389/fmicb.2023.1234115] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 07/14/2023] [Indexed: 09/01/2023] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is one of the major causes for nosocomial infections and has been classified as "high priority pathogen" by the World Health Organization. Its ability to develop resistances has been a challenge for the last decades and is still a threat to health care systems, as strains with resistances to the so-called drugs of last resort have been discovered. Therefore, new antibiotics are urgently needed. Natural products are an important source for the development of new drugs, thereby mostly serving as lead compounds for further modification. In this review, the data on plant natural products with reported anti-MRSA activity until the end of 2022 is discussed, highlighting the most effective drugs with respect to their inhibitory concentrations as well as with regard to eventual synergistic effects with existing antibiotics. In the latter sense, the class of alkaloids must be mentioned, exhibiting additive or synergistic effects by inhibiting bacterial efflux pumps. With regard to the antibiotic activity, phloroglucinol derivatives certainly belong to the most promising compounds, revealing several candidates with remarkable effects, e.g., lupulone, ivesinol, rhodomyrtone, aspidinol, or hyperforin. Also, the class of terpenoids yielded noteworthy compounds, such as the sesquiterpene lactones parthenolide and lactopicrin as well as acetophenone sesquiterpenes and sphaerodiene type diterpenoids, respectively. In addition, pronounced effects were observed for the macrolide neurymenolide A and three flavonol dicoumaroylrhamnosides.
Collapse
Affiliation(s)
| | - Serhat S. Çiçek
- Department of Pharmaceutical Biology, Institute of Pharmacy, Kiel University, Kiel, Germany
| |
Collapse
|
31
|
Jia Y, Chen W, Tang R, Zhang J, Liu X, Dong R, Hu F, Jiang X. Multi-armed antibiotics for Gram-positive bacteria. Cell Host Microbe 2023; 31:1101-1110.e5. [PMID: 37442098 DOI: 10.1016/j.chom.2023.06.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 04/03/2023] [Accepted: 06/14/2023] [Indexed: 07/15/2023]
Abstract
Antibiotic resistance is a serious threat to public health. Here, we propose a multi-armed chemical scaffold (MACS) for antibiotic screening, which refers to multi-armed molecules (MAMs) consisting of a core unit and three or four arms, neither of which is active for pathogens. Based on a structure-activity relationship study of MAMs, we discover a class of multi-armed antibiotics (MAAs) with a core similar to ethylene (E), carbon atom (C), benzene (B), nitrogen atom (N), and triazine (T) and three or four 4-phenylbenzoic acid (PBA) arms, or a B core and three 4-vinylbenzoic acid (VBA) or 4-ethynylbenzoic acid (EBA) arms. They can selectively interact with Gram-positive bacteria and inhibit cell wall assembly by targeting the lipid carriers of cell wall biosynthesis. MAAs have excellent antibacterial activities against Gram-positive bacteria, including clinical multi-drug-resistant (MDR) isolates. Our study provides a chemical scaffold and identifies eight antibacterial lead compounds for the development of antibiotics.
Collapse
Affiliation(s)
- Yuexiao Jia
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, No. 1088 Xueyuan Rd., Nanshan District, Shenzhen, Guangdong 518055, P.R. China; Institute of Biomedical Engineering and Health Sciences, School of Medical and Health Engineering, Changzhou University, Changzhou, Jiangsu 213164, P.R. China
| | - Wenwen Chen
- School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong 518055, P.R. China
| | - Rongbing Tang
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, No. 1088 Xueyuan Rd., Nanshan District, Shenzhen, Guangdong 518055, P.R. China
| | - Jiangjiang Zhang
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, No. 1088 Xueyuan Rd., Nanshan District, Shenzhen, Guangdong 518055, P.R. China
| | - Xiaoyan Liu
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, No. 1088 Xueyuan Rd., Nanshan District, Shenzhen, Guangdong 518055, P.R. China
| | - Ruihua Dong
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, No. 1088 Xueyuan Rd., Nanshan District, Shenzhen, Guangdong 518055, P.R. China
| | - Fupin Hu
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Xingyu Jiang
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, No. 1088 Xueyuan Rd., Nanshan District, Shenzhen, Guangdong 518055, P.R. China.
| |
Collapse
|
32
|
Wang X, Zhou N, Wang B. Bacterial synthetic biology: tools for novel drug discovery. Expert Opin Drug Discov 2023; 18:1087-1097. [PMID: 37482696 DOI: 10.1080/17460441.2023.2239704] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 07/19/2023] [Indexed: 07/25/2023]
Abstract
INTRODUCTION Bacterial synthetic biology has provided powerful tools to revolutionize the drug discovery process. These tools can be harnessed to generate bacterial novel pharmaceutical compounds with enhanced bioactivity and selectivity or to create genetically modified microorganisms as living drugs. AREAS COVERED This review provides a current overview of the state-of-the-art in bacterial synthetic biology tools for novel drug discovery. The authors discuss the application of these tools including bioinformatic tools, CRISPR tools, engineered bacterial transcriptional regulators, and synthetic biosensors for novel drug discovery. Additionally, the authors present the recent progress on reprogramming bacteriophages as living drugs to fight against antibiotic-resistant pathogens. EXPERT OPINION The field of using bacterial synthetic biology tools for drug discovery is rapidly advancing. However, challenges remain in developing reliable and robust methods to engineer bacteria. Further advancements in synthetic biology hold promise to speed up drug discovery, facilitating the development of novel therapeutics against various diseases.
Collapse
Affiliation(s)
- Xiyan Wang
- College of Chemical and Biological Engineering & ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, China
| | - Nan Zhou
- College of Chemical and Biological Engineering & ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, China
| | - Baojun Wang
- College of Chemical and Biological Engineering & ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, China
- Research Center of Biological Computation, Zhejiang Laboratory, Hangzhou, China
| |
Collapse
|
33
|
Guevara YAS, Santos MHC, Gomes FIR, Mesquita FP, Souza PFN. A historical, economic, and technical-scientific approach to the current crisis in the development of antibacterial drugs: Promising role of antibacterial peptides in this scenario. Microb Pathog 2023; 179:106108. [PMID: 37044203 DOI: 10.1016/j.micpath.2023.106108] [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: 03/10/2023] [Revised: 04/04/2023] [Accepted: 04/05/2023] [Indexed: 04/14/2023]
Abstract
The emergence of antibiotic resistance (AMR) is a global public health problem. According to estimates, drug-resistant bacteria infect 2 million patients and perish 23,000 annually. To overcome this problem, antimicrobial peptides became a potential solution based on a new mechanism of action against bacteria. This article addresses the phenomenon of antibacterial resistance in most of its nuances, responding to historical, technical-scientific, and economic aspects. Likewise, it explores new therapeutic approaches to combat multi-resistant pathogens, specifically concerning antibacterial peptides, as a potential therapeutic tool to mitigate the current crisis of antibacterial drugs. It is expected that, with technological advances, especially with the advent and adoption of artificial intelligence, there will be an increase in diversified synthetic peptide production, which can face the challenges that we have in terms of antibacterial drugs.
Collapse
Affiliation(s)
- Yeimer A S Guevara
- Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Maria H C Santos
- Department of Biochemistry and Molecular Biology, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Francisco I R Gomes
- Department of Biochemistry and Molecular Biology, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Felipe P Mesquita
- Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Ceará, Brazil; Department of Biochemistry and Molecular Biology, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Pedro F N Souza
- Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Ceará, Brazil; Department of Biochemistry and Molecular Biology, Federal University of Ceará, Fortaleza, Ceará, Brazil.
| |
Collapse
|
34
|
Recent advances in carboxymethyl chitosan-based materials for biomedical applications. Carbohydr Polym 2023; 305:120555. [PMID: 36737218 DOI: 10.1016/j.carbpol.2023.120555] [Citation(s) in RCA: 56] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 12/12/2022] [Accepted: 01/03/2023] [Indexed: 01/09/2023]
Abstract
Chitosan (CS) and its derivatives have been applied extensively in the biomedical field owing to advantageous characteristics including biodegradability, biocompatibility, antibacterial activity and adhesive properties. The low solubility of CS at physiological pH limits its use in systems requiring higher dissolving ability and a suitable drug release rate. Besides, CS can result in fast drug release because of its high swelling degree and rapid water absorption in aqueous media. As a water-soluble derivative of CS, carboxymethyl chitosan (CMC) has certain improved properties, rendering it a more suitable candidate for wound healing, drug delivery and tissue engineering applications. This review will focus on the antibacterial, anticancer and antitumor, antioxidant and antifungal bioactivities of CMC and the most recently described applications of CMC in wound healing, drug delivery, tissue engineering, bioimaging and cosmetics.
Collapse
|
35
|
Gao P, Nasution AK, Yang S, Chen Z, Ono N, Kanaya S, Altaf-Ul-Amin MD. On Finding Natural Antibiotics based on TCM Formulae. Methods 2023; 214:35-45. [PMID: 37019293 DOI: 10.1016/j.ymeth.2023.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 02/12/2023] [Accepted: 04/01/2023] [Indexed: 04/05/2023] Open
Abstract
CONTEXT Novel kinds of antibiotics are needed to combat the emergence of antibacterial resistance. Natural products (NPs) have shown potential as antibiotic candidates. Current experimental methods are not yet capable of exploring the massive, redundant, and noise-involved chemical space of NPs. In silico approaches are needed to select NPs as antibiotic candidates. OBJECTIVE This study screens out NPs with antibacterial efficacy guided by both TCM and modern medicine and constructed a dataset aiming to serve the new antibiotic design. METHOD A knowledge-based network is proposed in this study involving NPs, herbs, the concepts of TCM, and the treatment protocols (or etiologies) of infectious in modern medicine. Using this network, the NPs candidates are screened out and compose the dataset. Feature selection of machine learning approaches is conducted to evaluate the constructed dataset and statistically validate the im- portance of all NPs candidates for different antibiotics by a classification task. RESULTS The extensive experiments prove the constructed dataset reaches a convincing classification performance with a 0.9421 weighted accuracy, 0.9324 recall, and 0.9409 precision. The further visu- alizations of sample importance prove the comprehensive evaluation for model interpretation based on medical value considerations.
Collapse
Affiliation(s)
- Pei Gao
- Nara Institute of Science and Technology (NAIST), Ikoma, Nara 630-0101, Japan
| | | | - Shuo Yang
- Nara Institute of Science and Technology (NAIST), Ikoma, Nara 630-0101, Japan
| | - Zheng Chen
- Osaka University, Suita, Osaka 567-0047, Japan
| | - Naoaki Ono
- Nara Institute of Science and Technology (NAIST), Ikoma, Nara 630-0101, Japan
| | - Shigehiko Kanaya
- Nara Institute of Science and Technology (NAIST), Ikoma, Nara 630-0101, Japan
| | - M D Altaf-Ul-Amin
- Nara Institute of Science and Technology (NAIST), Ikoma, Nara 630-0101, Japan.
| |
Collapse
|
36
|
Michael, Waturangi DE. Antibiofilm activity from endophyte bacteria, Vibrio cholerae strains, and actinomycetes isolates in liquid and solid culture. BMC Microbiol 2023; 23:83. [PMID: 36991312 PMCID: PMC10053847 DOI: 10.1186/s12866-023-02829-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Abstract
Background
Biofilm-associated infections are a global threat to our economy and human health; as such, development of antibiofilm compounds is an urgent need. Our previous study identified eleven environmental isolates of endophyte bacteria, actinomycetes, and two strains of Vibrio cholerae as having strong antibiofilm activity, but only tested crude extracts from liquid culture. Here we grew the same bacteria in solid culture to induce the formation of colony biofilms and the expression of genes that may ultimately produce antibiofilm compounds. This research aimed to compare antibiofilm inhibition and destruction activities between liquid and solid cultures of these eleven environmental isolates against the biofilms of representative pathogenic bacteria.
Results
We measured antibiofilm activity using the static antibiofilm assay and crystal violet staining. The majority of our isolates exhibited higher inhibitory antibiofilm activity in liquid media, including all endophyte bacteria, V. cholerae V15a, and actinomycetes strains (CW01, SW03, CW17). However, for V. cholerae strain B32 and two actinomycetes bacteria (TB12 and SW12), the solid crude extracts showed higher inhibitory activity. Regarding destructive antibiofilm activity, many endophyte isolates and V. cholerae strains showed no significant difference between culture methods; the exceptions were endophyte bacteria isolate JerF4 and V. cholerae B32. The liquid extract of isolate JerF4 showed higher destructive activity relative to the corresponding solid culture extract, while for V. cholerae strain B32 the solid extract showed higher activity against some biofilms of pathogenic bacteria.
Conclusions
Culture conditions, namely solid or liquid culture, can influence the activity of culture extracts against biofilms of pathogenic bacteria. We compared the antibiofilm activity and presented the data that majority of isolates showed a higher antibiofilm activity in liquid culture. Interestingly, solid extracts from three isolates (B32, TB12, and SW12) have a better inhibition or/and destruction antibiofilm activity compared to their liquid culture. Further research is needed to characterize the activities of specific metabolites in solid and liquid culture extracts and to determine the mechanisms of their antibiofilm actions.
Collapse
|
37
|
Gutierrez-Gongora D, Raouf-Alkadhimi F, Prosser RS, Geddes-McAlister J. Differentiated extracts from freshwater and terrestrial mollusks inhibit virulence factor production in Cryptococcus neoformans. Sci Rep 2023; 13:4928. [PMID: 36967422 PMCID: PMC10040410 DOI: 10.1038/s41598-023-32140-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 03/23/2023] [Indexed: 03/29/2023] Open
Abstract
The human fungal pathogen, Cryptococcus neoformans, is responsible for deadly infections among immunocompromised individuals with the evolution of antifungal resistance driving the solution to discover new compounds that inhibit fungal virulence factors rather than kill the pathogen. Recently, exploration into natural sources (e.g., plants, invertebrates, microbes) of antifungal agents has garnered attention by integrating a One Health approach for new compound discovery. Here, we explore extracts from three mollusk species (freshwater and terrestrial) and evaluate effects against the growth and virulence factor production (i.e., thermotolerance, melanin, capsule, and biofilm) in C. neoformans. We demonstrate that clarified extracts of Planorbella pilsbryi have a fungicidal effect on cryptococcal cells comparable to fluconazole. Similarly, all extracts of Cipangopaludina chinensis affect cryptococcal thermotolerance and impair biofilm and capsule production, with clarified extracts of Cepaea nemoralis also conveying the latter effect. Next, inhibitory activity of extracts against peptidases related to specific virulence factors, combined with stress assays and quantitative proteomics, defined distinct proteome signatures and proposed proteins driving the observed anti-virulence properties. Overall, this work highlights the potential of compounds derived from natural sources to inhibit virulence factor production in a clinically important fungal pathogen.
Collapse
Affiliation(s)
| | | | - Ryan S Prosser
- Department of Environmental Toxicology, University of Guelph, Guelph, ON, Canada
| | | |
Collapse
|
38
|
Zhang Y, Lin M, Qin Y, Lu H, Xu X, Gao C, Liu Y, Luo W, Luo X. Anti-Vibrio potential of natural products from marine microorganisms. Eur J Med Chem 2023; 252:115330. [PMID: 37011553 DOI: 10.1016/j.ejmech.2023.115330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/28/2023] [Accepted: 03/28/2023] [Indexed: 03/31/2023]
Abstract
The emergence of drug-resistant Vibrio poses a serious threat to aquaculture and human health, thus there is an urgent need for the discovery of new related antibiotics. Given that marine microorganisms (MMs) are evidenced as important sources of antibacterial natural products (NPs), great attention has been gained to the exploration of potential anti-Vibrio agents from MMs. This review summarizes the occurrence, structural diversity, and biological activities of 214 anti-Vibrio NPs isolated from MMs (from 1999 to July 2022), including 108 new compounds. They were predominantly originated from marine fungi (63%) and bacteria (30%) with great structural diversity, including polyketides, nitrogenous compounds, terpenoids, and steroids, among which polyketides account for nearly half (51%) of them. This review will shed light on the development of MMs derived NPs as potential anti-Vibrio lead compounds with promising applications in agriculture and human health.
Collapse
|
39
|
Fakee J, Bolton JJ, Le Roes-Hill M, Durrell KA, Antunes E, Beukes DR. Antimicrobial Activity of the Secondary Metabolites Isolated from a South African Red Seaweed, Laurencia corymbosa. Molecules 2023; 28:molecules28052063. [PMID: 36903309 PMCID: PMC10003847 DOI: 10.3390/molecules28052063] [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: 01/23/2023] [Revised: 02/17/2023] [Accepted: 02/19/2023] [Indexed: 02/25/2023] Open
Abstract
South Africa's highly diverse marine biota includes several endemic marine red algae of the Laurencia genus. Cryptic species and morphological variability make the taxonomy of Laurencia plant challenging, and a record of the secondary metabolites isolated from South African Laurencia spp. can be used to assess their chemotaxonomic significance. In addition, the rapid development of resistance against antibiotics, coupled with the inherent ability of seaweeds to resist pathogenic infection, supported this first phycochemical investigation of Laurencia corymbosa J. Agardh. A new tricyclic keto-cuparane (7) and two new cuparanes (4, 5) were obtained alongside known acetogenins, halo-chamigranes, and additional cuparanes. These compounds were screened against Acinetobacter baumannii, Enterococcus faecalis, Escherichia coli, Staphylococcus aureus, and Candida albicans, with 4 exhibiting excellent activity against the Gram-negative A. baumanii (minimum inhibitory concentration (MIC) 1 μg/mL) strain.
Collapse
Affiliation(s)
- Jameel Fakee
- Faculty of Pharmacy, Rhodes University, Makhanda 6140, South Africa
| | - John J. Bolton
- Department of Biological Sciences, University of Cape Town, Rondebosch 7701, South Africa
| | - Marilize Le Roes-Hill
- Applied Microbial and Health Biotechnology Institute, Cape Peninsula University of Technology, Bellville 7535, South Africa
| | - Kim A. Durrell
- Applied Microbial and Health Biotechnology Institute, Cape Peninsula University of Technology, Bellville 7535, South Africa
| | - Edith Antunes
- Department of Chemistry, University of the Western Cape, Bellville 7535, South Africa
| | - Denzil R. Beukes
- School of Pharmacy, University of the Western Cape, Bellville 7535, South Africa
- Correspondence: ; Tel.: +27-21-959-2352
| |
Collapse
|
40
|
Antibacterial natural products from microbial and fungal sources: a decade of advances. Mol Divers 2023; 27:517-541. [PMID: 35301633 DOI: 10.1007/s11030-022-10417-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 02/22/2022] [Indexed: 02/08/2023]
Abstract
Throughout the ages the world has witnessed the outbreak of many infectious diseases. Emerging microbial diseases pose a serious threat to public health. Increasing resistance of microorganisms towards the existing drugs makes them ineffective. In fact, anti-microbial resistance is declared as one of the top public health threats by WHO. Hence, there is an urge for the discovery of novel antimicrobial drugs to combat with this challenge. Structural diversity and unique pharmacological effects make natural products a prime source of novel drugs. Staggeringly, in spite of its extensive biodiversity, a prominent portion of microorganism species remains unexplored for the identification of bioactives. Microorganisms are a predominant source of new chemical entities and there are remarkable number of antimicrobial drugs developed from it. In this review, we discuss the contributions of microorganism based natural products as effective antibacterial agents, studied during the period of 2010-2020. The review encompasses over 140 structures which are either natural products or semi-synthetic derivatives of microbial natural products. 65 of them are identified as newly discovered natural products. All the compounds discussed herein, have exhibited promising efficacy against various bacterial strains.
Collapse
|
41
|
Tseduliak VM, Dolia B, Ostash I, Lopatniuk M, Busche T, Ochi K, Kalinowski J, Luzhetskyy A, Fedorenko V, Ostash B. Mutations within gene XNR_2147 for TetR-like protein enhance lincomycin resistance and endogenous specialized metabolism of Streptomyces albus J1074. J Appl Genet 2023; 64:185-195. [PMID: 36417169 DOI: 10.1007/s13353-022-00738-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 11/16/2022] [Accepted: 11/16/2022] [Indexed: 11/24/2022]
Abstract
Streptomyces albus J1074 is one of the most popular heterologous expression platforms among streptomycetes. Identification of new genes and mutations that influence specialized metabolism in this species is therefore of great applied interest. Here, we describe S. albus KO-1304 that was isolated as a spontaneous lincomycin-resistant variant of double rpsLR94G rsmGR15SG40E mutant KO-1295. Besides altered antibiotic resistance profile, KO-1304 exhibited increased antibiotic activity as compared to its parental strains. KO-1304 genome sequencing revealed mutations within gene XNR_2147 encoding putative TetR-like protein. Gene XNR_2146 for efflux protein is the most likely target of repressing action of Xnr_2147. Our data agree with the scenario where lincomycin resistance phenotype of KO-1304 arose from inability of mutated Xnr_2147 protein to repress XNR_2146. Introduction of additional copy of XNR_2146 into wild type strain increased antibiotic activity of the latter, attesting to the practical value of transporter genes for strain improvement.
Collapse
Affiliation(s)
- Vasylyna-Marta Tseduliak
- Department of Genetics and Biotechnology, Ivan Franko National University of Lviv, Hrushevskoho St. 4, Lviv, 79005, Ukraine
| | - Borys Dolia
- Department of Genetics and Biotechnology, Ivan Franko National University of Lviv, Hrushevskoho St. 4, Lviv, 79005, Ukraine
| | - Iryna Ostash
- Department of Genetics and Biotechnology, Ivan Franko National University of Lviv, Hrushevskoho St. 4, Lviv, 79005, Ukraine
| | - Maria Lopatniuk
- Helmholtz Institute for Pharmaceutical Research, Saarland Campus, Building C2.3, 66123, Saarbrucken, Germany
| | - Tobias Busche
- Technology Platform Genomics, CeBiTec, Bielefeld University, Sequenz 1, 33615, Bielefeld, Germany
| | - Kozo Ochi
- Department of Life Sciences, Hiroshima Institute of Technology, Saeki-Ku, Hiroshima, 731-5193, Japan
| | - Jörn Kalinowski
- Technology Platform Genomics, CeBiTec, Bielefeld University, Sequenz 1, 33615, Bielefeld, Germany
| | - Andriy Luzhetskyy
- Helmholtz Institute for Pharmaceutical Research, Saarland Campus, Building C2.3, 66123, Saarbrucken, Germany
| | - Victor Fedorenko
- Department of Genetics and Biotechnology, Ivan Franko National University of Lviv, Hrushevskoho St. 4, Lviv, 79005, Ukraine
| | - Bohdan Ostash
- Department of Genetics and Biotechnology, Ivan Franko National University of Lviv, Hrushevskoho St. 4, Lviv, 79005, Ukraine.
| |
Collapse
|
42
|
Albini F, Bormann S, Gerschel P, Ludwig VA, Neumann W. Dithiolopyrrolones are Prochelators that are Activated by Glutathione. Chemistry 2023; 29:e202202567. [PMID: 36214647 PMCID: PMC10099403 DOI: 10.1002/chem.202202567] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Indexed: 11/06/2022]
Abstract
Dithiolopyrrolones (DTPs), such as holomycin, are natural products that hold promise as scaffolds for antibiotics as they exhibit inhibitory activity against antibiotic-resistant pathogens. They consist of a unique bicyclic core containing a disulfide that is crucial for their biological activity. Herein, we establish the DTPs as prochelators. We show that the disulfides are reduced at cellular gluathione levels. This activates the drugs and initiates interactions with targets, particularly metal coordination. In addition, we report an expedient synthesis for the DTPs thiolutin and aureothricin, providing facile access to important natural DTPs and derivatives thereof.
Collapse
Affiliation(s)
- Francesca Albini
- Inorganic Chemistry I - Bioinorganic Chemistry, Ruhr-University Bochum, 44780, Bochum, Germany
| | - Stefan Bormann
- Inorganic Chemistry I - Bioinorganic Chemistry, Ruhr-University Bochum, 44780, Bochum, Germany
| | - Philipp Gerschel
- Inorganic Chemistry I - Bioinorganic Chemistry, Ruhr-University Bochum, 44780, Bochum, Germany
| | - Veza A Ludwig
- Inorganic Chemistry I - Bioinorganic Chemistry, Ruhr-University Bochum, 44780, Bochum, Germany
| | - Wilma Neumann
- Inorganic Chemistry I - Bioinorganic Chemistry, Ruhr-University Bochum, 44780, Bochum, Germany
| |
Collapse
|
43
|
Deng Y, Zhang Y, Chen XH, Li CH. Antibacterial activity evaluation of pleuromutilin derivatives with 4(3H)-quinazolinone scaffold against methicillin-resistant Staphylococcusaureus. Eur J Med Chem 2023; 246:114960. [PMID: 36462445 DOI: 10.1016/j.ejmech.2022.114960] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 11/11/2022] [Accepted: 11/22/2022] [Indexed: 11/24/2022]
Abstract
Growing antibiotic resistance is causing a health care crisis, leading to an urgent need for new antibiotics to tackle serious hospital and community infections. Pleuromutilin, a naturally occurring product with moderate antibacterial activity, has a unique structure that has attracted great efforts to modify its scaffold to obtain lead compounds. Herein, we report the synthesis of a series of novel pleuromutilin derivatives with a scaffold of 4(3H)-quinazolinone or its analogues at the C-14 side chain and investigated their in vitro activity against Staphylococcus aureus and Staphylococcus epidermidis as well as Gram-negative bacteria (Escherichia coli and Salmonella enterica subsp. enterica serovar pullorum). Structure-activity relationship (SAR) studies showed that the substituents on the benzene ring of 4(3H)-quinazolinone was not as important as the substituted position to improve antibacterial activity while the substituted groups on the N-3 position of 4(3H)-quinazolinone had strong impact on the efficacy. The replacement of the benzene moiety of 4(3H)-quinazolinone with other rings (pyridine, pyrrole, thiophene, or cyclopentyl) also showed high antibacterial efficacy, meaning the benzene ring was dispensable for exerting powerful antibacterial properties. In vitro pharmacokinetics investigations and cytotoxicity assays indicated that 2-mercapto-4(3H)-quinazolinone scaffold was superior to 2-(piperazin-1-yl)quinazolin-4(3H)-one. Among this series of pleuromutilin analogues, compound 23 with a structure of 2-mercapto-3H-pyrrolo[2,3-d]pyrimidin-4(7H)-one displayed the best in vitro antibacterial activity against MRSA (MIC = 0.063 μg/mL) and low cytotoxicity to RAW 264.7 cells (IC50>100 μM) and was demonstrated to inhibit MRSA effectively in a mouse thigh infection model, outperforming the comparator, tiamulin.
Collapse
Affiliation(s)
- Yu Deng
- Institute of Veterinary Sciences & Medicines, Chongqing Academy of Animal Sciences, Rongchang, 402460, China; National Pig Technology Innovation Center, Rongchang, 402460, China
| | - Yang Zhang
- National Pig Technology Innovation Center, Rongchang, 402460, China
| | - Xiao-Hu Chen
- Department of Clinical Laboratory, Rongchang District People's Hospital, Rongchang, 402460, China
| | - Cheng-Hong Li
- Institute of Veterinary Sciences & Medicines, Chongqing Academy of Animal Sciences, Rongchang, 402460, China; National Pig Technology Innovation Center, Rongchang, 402460, China.
| |
Collapse
|
44
|
Du RL, Chow HY, Chen YW, Chan PH, Daniel RA, Wong KY. Gossypol acetate: A natural polyphenol derivative with antimicrobial activities against the essential cell division protein FtsZ. Front Microbiol 2023; 13:1080308. [PMID: 36713210 PMCID: PMC9878342 DOI: 10.3389/fmicb.2022.1080308] [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: 10/26/2022] [Accepted: 12/28/2022] [Indexed: 01/14/2023] Open
Abstract
Antimicrobial resistance has attracted worldwide attention and remains an urgent issue to resolve. Discovery of novel compounds is regarded as one way to circumvent the development of resistance and increase the available treatment options. Gossypol is a natural polyphenolic aldehyde, and it has attracted increasing attention as a possible antibacterial drug. In this paper, we studied the antimicrobial properties (minimum inhibitory concentrations) of gossypol acetate against both Gram-positive and Gram-negative bacteria strains and dig up targets of gossypol acetate using in vitro assays, including studying its effects on functions (GTPase activity and polymerization) of Filamenting temperature sensitive mutant Z (FtsZ) and its interactions with FtsZ using isothermal titration calorimetry (ITC), and in vivo assays, including visualization of cell morphologies and proteins localizations using a microscope. Lastly, Bacterial membrane permeability changes were studied, and the cytotoxicity of gossypol acetate was determined. We also estimated the interactions of gossypol acetate with the promising target. We found that gossypol acetate can inhibit the growth of Gram-positive bacteria such as the model organism Bacillus subtilis and the pathogen Staphylococcus aureus [both methicillin-sensitive (MSSA) and methicillin-resistant (MRSA)]. In addition, gossypol acetate can also inhibit the growth of Gram-negative bacteria when the outer membrane is permeabilized by Polymyxin B nonapeptide (PMBN). Using a cell biological approach, we show that gossypol acetate affects cell division in bacteria by interfering with the assembly of the cell division FtsZ ring. Biochemical analysis shows that the GTPase activity of FtsZ was inhibited and polymerization of FtsZ was enhanced in vitro, consistent with the block to cell division in the bacteria tested. The binding mode of gossypol acetate in FtsZ was modeled using molecular docking and provides an understanding of the compound mode of action. The results point to gossypol (S2303) as a promising antimicrobial compound that inhibits cell division by affecting FtsZ polymerization and has potential to be developed into an effective antimicrobial drug by chemical modification to minimize its cytotoxic effects in eukaryotic cells that were identified in this work.
Collapse
Affiliation(s)
- Ruo-Lan Du
- Department of Applied Biology and Chemical Technology and the State Key Laboratory of Chemical Biology and Drug Discovery, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Ho-Yin Chow
- Department of Applied Biology and Chemical Technology and the State Key Laboratory of Chemical Biology and Drug Discovery, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Yu Wei Chen
- Department of Applied Biology and Chemical Technology and the State Key Laboratory of Chemical Biology and Drug Discovery, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Pak-Ho Chan
- Department of Applied Biology and Chemical Technology and the State Key Laboratory of Chemical Biology and Drug Discovery, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Richard A. Daniel
- Centre for Bacterial Cell Biology, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom,Richard A. Daniel,
| | - Kwok-Yin Wong
- Department of Applied Biology and Chemical Technology and the State Key Laboratory of Chemical Biology and Drug Discovery, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China,*Correspondence: Kwok-Yin Wong,
| |
Collapse
|
45
|
Cheng W, Xu T, Cui L, Xue Z, Liu J, Yang R, Qin S, Guo Y. Discovery of Amphiphilic Xanthohumol Derivatives as Membrane-Targeting Antimicrobials against Methicillin-Resistant Staphylococcus aureus. J Med Chem 2023; 66:962-975. [PMID: 36584344 DOI: 10.1021/acs.jmedchem.2c01793] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Infections caused by multidrug-resistant (MDR) bacteria are increasing worldwide, and with limited clinically available antibiotics, it is urgent to develop new antimicrobials to combat these MDR bacteria. Here, a class of novel amphiphilic xanthohumol derivatives were prepared using a building-block approach. Bioactivity assays showed that the molecule IV15 not only exhibited a remarkable antibacterial effect against clinical methicillin-resistant Staphylococcus aureus (MRSA) isolates (MICs: 1-2 μg/mL) but also had the advantages of rapid bactericidal properties, low toxicity, good plasma stability, and not readily inducing bacterial resistance. Mechanistic studies indicated that IV15 has good membrane-targeting ability and can bind to phosphatidylglycerol and cardiolipin in bacterial membranes, thus disrupting the bacterial cell membranes and causing increased intracellular reactive oxygen species and leakage of proteins and DNA, eventually resulting in bacterial death. Notably, IV15 exhibited remarkable in vivo anti-MRSA efficacy, superior to vancomycin, making it a potential candidate to combat MRSA infections.
Collapse
Affiliation(s)
- Wanqing Cheng
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| | - Ting Xu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| | - Liping Cui
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| | - Zihan Xue
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| | - Jifeng Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| | - Ruige Yang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| | - Shangshang Qin
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| | - Yong Guo
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan Province, China.,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
| |
Collapse
|
46
|
Fernandez-Soto P, Celi D, Tejera E, Alvarez-Suarez JM, Machado A. Cinnamomum sp. and Pelargonium odoratissimum as the Main Contributors to the Antibacterial Activity of the Medicinal Drink Horchata: A Study Based on the Antibacterial and Chemical Analysis of 21 Plants. Molecules 2023; 28:693. [PMID: 36677749 PMCID: PMC9862262 DOI: 10.3390/molecules28020693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/28/2022] [Accepted: 01/04/2023] [Indexed: 01/13/2023] Open
Abstract
Horchata, a herbal infusion drink from Ecuador containing a mixture of medicinal plants, has been reported to exhibit anti-inflammatory, analgesic, diuretic, and antioxidant activity. The antibacterial activity of each of the plants contained in the horchata mixture has not been fully evaluated. Thus, in this study, we analysed the antibacterial activity of 21 plants used in horchata, collected from the Ecuadorian Andes region, against bacterial strains of clinical importance. The methanolic extract of Cinnamomum sp. showed minimal inhibitory concentration (MIC) values of 250 µg/mL against Staphylococcus aureus ATCC25923 and Methicillin-resistant S. aureus (MRSA), while Pelargonium odoratissimum exhibited a MIC value of 500 µg/mL towards S. aureus ATCC25923. The high-performance liquid chromatography-diode array detector-tandem mass spectrometry (HPLC-DAD-MS/MS) analyses identified in Cinnamomum sp. epicatechin tannins, cinnamaldehyde, and prehelminthosporol molecules, whereas in P. odoratissimum, gallocatechin and epigallocatechin tannins, some flavonoids, and gallic acid and derivatives were identified. Finally, Cinnamomum sp. and P. odoratissimum showed partial inhibition of biofilm formation of S. aureus ATCC25923 and MRSA. Overall, our findings revealed which of the plants used in horchata are responsible for the antibacterial activity attributed to this herbal drink and exhibit the potential for Cinnamomum sp. and P. odoratissimum secondary metabolites to be explored as scaffolds in drug development.
Collapse
Affiliation(s)
- Paulina Fernandez-Soto
- Facultad de Ciencias de la Salud, Carrera de Enfermería, Grupo de Bio-Quimioinformática, Universidad de Las Américas (UDLA), Quito 170125, Ecuador
| | - Diana Celi
- Facultad de Ingeniería y Ciencias Aplicadas, Carrera de Ingeniería Agroindustrial, Universidad de Las Américas (UDLA), Quito 170125, Ecuador
| | - Eduardo Tejera
- Facultad de Ingeniería y Ciencias Agropecuarias Aplicadas, Grupo de Bio-Quimioinformática, Universidad de Las Américas (UDLA), Quito 170125, Ecuador
| | - José Miguel Alvarez-Suarez
- Colegio de Ciencias e Ingenierías, Departamento de Ingeniería en Alimentos, Universidad San Francisco de Quito (USFQ), Quito 170901, Ecuador
| | - António Machado
- Colegio de Ciencias Biológicas y Ambientales (COCIBA), Instituto de Microbiología, Laboratorio de Bacteriología, Universidad San Francisco de Quito (USFQ), Quito 170901, Ecuador
| |
Collapse
|
47
|
Seyfert CE, Porten C, Yuan B, Deckarm S, Panter F, Bader CD, Coetzee J, Deschner F, Tehrani KHME, Higgins PG, Seifert H, Marlovits TC, Herrmann J, Müller R. Darobactins Exhibiting Superior Antibiotic Activity by Cryo-EM Structure Guided Biosynthetic Engineering. Angew Chem Int Ed Engl 2023; 62:e202214094. [PMID: 36308277 PMCID: PMC10107326 DOI: 10.1002/anie.202214094] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Indexed: 11/06/2022]
Abstract
Over recent decades, the pipeline of antibiotics acting against Gram-negative bacteria is running dry, as most discovered candidate antibiotics suffer from insufficient potency, pharmacokinetic properties, or toxicity. The darobactins, a promising new small peptide class of drug candidates, bind to novel antibiotic target BamA, an outer membrane protein. Previously, we reported that biosynthetic engineering in a heterologous host generated novel darobactins with enhanced antibacterial activity. Here we utilize an optimized purification method and present cryo-EM structures of the Bam complex with darobactin 9 (D9), which served as a blueprint for the biotechnological generation of twenty new darobactins including halogenated analogs. The newly engineered darobactin 22 binds more tightly to BamA and outperforms the favorable activity profile of D9 against clinically relevant pathogens such as carbapenem-resistant Acinetobacter baumannii up to 32-fold, without observing toxic effects.
Collapse
Affiliation(s)
- Carsten E Seyfert
- Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Department of Pharmacy at, Saarland University Campus Building E8.1, 66123 Saarbrücken (Germany).,German Centre for Infection Research (DZIF), partnersite Hannover-Braunschweig, Germany
| | - Christoph Porten
- Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Department of Pharmacy at, Saarland University Campus Building E8.1, 66123 Saarbrücken (Germany).,German Centre for Infection Research (DZIF), partnersite Hannover-Braunschweig, Germany
| | - Biao Yuan
- University Medical Center Hamburg-Eppendorf (UKE), Institute of Structural and Systems Biology, Notkestraße 85, Building 15, 22607, Hamburg, Germany.,Centre for Structural Systems Biology (CSSB), Hamburg, Germany
| | - Selina Deckarm
- Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Department of Pharmacy at, Saarland University Campus Building E8.1, 66123 Saarbrücken (Germany).,German Centre for Infection Research (DZIF), partnersite Hannover-Braunschweig, Germany
| | - Fabian Panter
- Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Department of Pharmacy at, Saarland University Campus Building E8.1, 66123 Saarbrücken (Germany).,German Centre for Infection Research (DZIF), partnersite Hannover-Braunschweig, Germany
| | - Chantal D Bader
- Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Department of Pharmacy at, Saarland University Campus Building E8.1, 66123 Saarbrücken (Germany).,German Centre for Infection Research (DZIF), partnersite Hannover-Braunschweig, Germany
| | - Janetta Coetzee
- Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Department of Pharmacy at, Saarland University Campus Building E8.1, 66123 Saarbrücken (Germany).,German Centre for Infection Research (DZIF), partnersite Hannover-Braunschweig, Germany
| | - Felix Deschner
- Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Department of Pharmacy at, Saarland University Campus Building E8.1, 66123 Saarbrücken (Germany).,German Centre for Infection Research (DZIF), partnersite Hannover-Braunschweig, Germany
| | - Kamaleddin H M E Tehrani
- Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Department of Pharmacy at, Saarland University Campus Building E8.1, 66123 Saarbrücken (Germany).,German Centre for Infection Research (DZIF), partnersite Hannover-Braunschweig, Germany
| | - Paul G Higgins
- Microbiology, Immunology and Hygiene, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.,German Center for Infection Research (DZIF), partner site Bonn-Cologne, Germany
| | - Harald Seifert
- Microbiology, Immunology and Hygiene, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.,German Center for Infection Research (DZIF), partner site Bonn-Cologne, Germany
| | - Thomas C Marlovits
- University Medical Center Hamburg-Eppendorf (UKE), Institute of Structural and Systems Biology, Notkestraße 85, Building 15, 22607, Hamburg, Germany.,Centre for Structural Systems Biology (CSSB), Hamburg, Germany.,Deutsches Elektronen-Synchrotron Zentrum (DESY), Hamburg, Germany
| | - Jennifer Herrmann
- Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Department of Pharmacy at, Saarland University Campus Building E8.1, 66123 Saarbrücken (Germany).,German Centre for Infection Research (DZIF), partnersite Hannover-Braunschweig, Germany
| | - Rolf Müller
- Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Department of Pharmacy at, Saarland University Campus Building E8.1, 66123 Saarbrücken (Germany).,German Centre for Infection Research (DZIF), partnersite Hannover-Braunschweig, Germany
| |
Collapse
|
48
|
da Silva L, Donato IA, Gonçalves CAC, Scherf JR, dos Santos HS, Mori E, Coutinho HDM, da Cunha FAB. Antibacterial potential of chalcones and its derivatives against Staphylococcus aureus. 3 Biotech 2023; 13:1. [PMID: 36466769 PMCID: PMC9712905 DOI: 10.1007/s13205-022-03398-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 10/12/2022] [Indexed: 12/02/2022] Open
Abstract
Chalcones are natural substances found in the metabolism of several botanical families. Their structure consists of 1,3-diphenyl-2-propen-1-one and they are characterized by having in their chains an α, β-unsaturated carbonyl system, two phenol rings and a three-carbon chain that unites them. In plants, Chalcones are mainly involved in the biosynthesis of flavonoids and isoflavonoids through the phenylalanine derivation. This group of substances has been shown to be a viable alternative for the investigation of its antibacterial potential, considering the numerous biological activities reported and the increase of the microbial resistance that concern global health agencies. Staphylococcus aureus is a bacterium that has stood out for its ability to adapt and develop resistance to a wide variety of drugs. This literature review aimed to highlight recent advances in the use of Chalcones and derivatives as antibacterial agents against S. aureus, focusing on research articles available on the Science Direct, Pub Med and Scopus data platforms in the period 2015-2021. It was constructed informative tables that provided an overview of which types of Chalcones are being studied more (Natural or Synthetic); its chemical name and main Synthesis Methodology. From the analysis of the data, it was observed that the compounds based on Chalcones have great potential in medicinal chemistry as antibacterial agents and that the molecular skeletons of these compounds as well as their derivatives can be easily obtained through substitutions in the A and B rings of Chalcones, in order to obtain the desired bioactivity. It was verified that Chalcones and derivatives are promising agents for combating the multidrug resistance of S. aureus to drugs. Supplementary Information The online version contains supplementary material available at 10.1007/s13205-022-03398-7.
Collapse
Affiliation(s)
- Larissa da Silva
- Laboratory of Semi-Arid Bioprospecting (LABSEMA), Department of Biological Chemistry, URCA, Crato, CE Brazil
| | - Isydorio Alves Donato
- Laboratory of Semi-Arid Bioprospecting (LABSEMA), Department of Biological Chemistry, URCA, Crato, CE Brazil
| | | | - Jackelyne Roberta Scherf
- Graduate Program in Pharmaceutical Sciences, Federal University of Pernambuco, UFPE, Recife, PE Brazil
| | - Hélcio Silva dos Santos
- Laboratory of Chemistry of Natural and Synthetic Product, State university of Ceará, UECE, Fortaleza, CE Brazil
| | - Edna Mori
- CECAPE, College of Dentistry, Juazeiro do Norte, CE 63024-015 Brazil
| | | | | |
Collapse
|
49
|
Li J, Yi W, Luo Y, Yang K, He L, Xu C, Deng L, He D. GSH-depleting and H 2O 2-self-supplying hybrid nanozymes for intensive catalytic antibacterial therapy by photothermal-augmented co-catalysis. Acta Biomater 2023; 155:588-600. [PMID: 36328125 DOI: 10.1016/j.actbio.2022.10.050] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 09/22/2022] [Accepted: 10/24/2022] [Indexed: 11/09/2022]
Abstract
Nanozyme-based chemodynamic therapy (CDT) has shown tremendous potential in the treatment of bacterial infections. However, the CDT antibacterial efficacy is severely limited by the catalytic activity of nanozymes or the infection microenvironments such as insufficient hydrogen peroxide (H2O2) and overexpressed glutathione (GSH). Herein, a versatile hybrid nanozyme (MoS2/CuO2) is rationally constructed by simply decorating ultrasmall CuO2 nanodots onto lamellar MoS2 platelets of hydrangea-like MoS2 nanocarrier via a covalent Cu-S bond. The MoS2/CuO2 nanozyme exhibits the peroxidase-mimic activity for catalytically converting H2O2 produced by acid-triggered decomposition of the decorated CuO2 into hydroxyl radical (•OH). Meanwhile, the MoS2/CuO2 can consume GSH overexpressed in the infection sites via redox reaction mediated by polyvalent transition metal ions (Cu2+ and Mo6+) for enhanced CDT. More importantly, MoS2 support can promote the conversion of Cu2+ to Cu+ by a co-catalytic reaction based on the Mo4+/Mo6+ redox couples, and provide photonic hyperthermia (PTT) to augment the peroxidase-mimic activity. The developed MoS2/CuO2 nanozymes possesses a desirable catalytic property, as well as a remarkably improved antibacterial efficiency both in vitro and in vivo. Taken together, this study proposes a synergetic multiple enhancement strategy to successfully construct the versatile hybrid nanozymes for intensive in vivo PTT/CDT dual-mode anti-infective therapy. STATEMENT OF SIGNIFICANCE: Chemodynamic therapy (CDT) has shown great potentialities in the treatment of bacterial infections, while its therapeutic efficiency is severely limited by the infection microenvironments such as insufficient hydrogen peroxide (H2O2) and overexpressed glutathione (GSH). Here, we rationally construct a hybrid nanozyme (MoS2/CuO2) with peroxidase-like activity that can enhance CDT by regulating local microenvironments, that is, simultaneously self-supplying H2O2 and consuming GSH. Importantly, MoS2 support can promote the conversion of Cu2+ to Cu+ by the Mo4+/Mo6+ redox couples, and provide photonic hyperthermia (PTT) to augment the peroxidase-mimic activity. The developed MoS2/CuO2 shows desirable PTT/CDT dual-mode antibacterial efficacy both in vitro and in vivo. This study proposes a versatile hybrid nanozyme with multiple enhancement effects for intensive in vivo anti-infective therapy.
Collapse
Affiliation(s)
- Junqin Li
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, China
| | - Wenhua Yi
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, China
| | - Yuze Luo
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, China
| | - Ke Yang
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, China
| | - Lidan He
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, China
| | - Caiyun Xu
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, China
| | - Le Deng
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, China
| | - Dinggeng He
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, China.
| |
Collapse
|
50
|
Walesch S, Birkelbach J, Jézéquel G, Haeckl FPJ, Hegemann JD, Hesterkamp T, Hirsch AKH, Hammann P, Müller R. Fighting antibiotic resistance-strategies and (pre)clinical developments to find new antibacterials. EMBO Rep 2022; 24:e56033. [PMID: 36533629 PMCID: PMC9827564 DOI: 10.15252/embr.202256033] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 11/23/2022] [Accepted: 11/25/2022] [Indexed: 12/23/2022] Open
Abstract
Antibacterial resistance is one of the greatest threats to human health. The development of new therapeutics against bacterial pathogens has slowed drastically since the approvals of the first antibiotics in the early and mid-20th century. Most of the currently investigated drug leads are modifications of approved antibacterials, many of which are derived from natural products. In this review, we highlight the challenges, advancements and current standing of the clinical and preclinical antibacterial research pipeline. Additionally, we present novel strategies for rejuvenating the discovery process and advocate for renewed and enthusiastic investment in the antibacterial discovery pipeline.
Collapse
Affiliation(s)
- Sebastian Walesch
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS)SaarbrückenGermany,Department of PharmacySaarland UniversitySaarbrückenGermany,Helmholtz Centre for Infection research (HZI)BraunschweigGermany,German Center for infection research (DZIF)BraunschweigGermany
| | - Joy Birkelbach
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS)SaarbrückenGermany,Department of PharmacySaarland UniversitySaarbrückenGermany,Helmholtz Centre for Infection research (HZI)BraunschweigGermany,German Center for infection research (DZIF)BraunschweigGermany
| | - Gwenaëlle Jézéquel
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS)SaarbrückenGermany,Helmholtz Centre for Infection research (HZI)BraunschweigGermany
| | - F P Jake Haeckl
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS)SaarbrückenGermany,Department of PharmacySaarland UniversitySaarbrückenGermany,Helmholtz Centre for Infection research (HZI)BraunschweigGermany,German Center for infection research (DZIF)BraunschweigGermany
| | - Julian D Hegemann
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS)SaarbrückenGermany,Department of PharmacySaarland UniversitySaarbrückenGermany,Helmholtz Centre for Infection research (HZI)BraunschweigGermany,German Center for infection research (DZIF)BraunschweigGermany
| | - Thomas Hesterkamp
- Helmholtz Centre for Infection research (HZI)BraunschweigGermany,German Center for infection research (DZIF)BraunschweigGermany
| | - Anna K H Hirsch
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS)SaarbrückenGermany,Department of PharmacySaarland UniversitySaarbrückenGermany,Helmholtz Centre for Infection research (HZI)BraunschweigGermany,German Center for infection research (DZIF)BraunschweigGermany,Helmholtz International Lab for Anti‐InfectivesSaarbrückenGermany
| | - Peter Hammann
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS)SaarbrückenGermany,Department of PharmacySaarland UniversitySaarbrückenGermany,Helmholtz Centre for Infection research (HZI)BraunschweigGermany,German Center for infection research (DZIF)BraunschweigGermany
| | - Rolf Müller
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS)SaarbrückenGermany,Department of PharmacySaarland UniversitySaarbrückenGermany,Helmholtz Centre for Infection research (HZI)BraunschweigGermany,German Center for infection research (DZIF)BraunschweigGermany,Helmholtz International Lab for Anti‐InfectivesSaarbrückenGermany
| |
Collapse
|