1
|
Ortiz-López FJ, Oves-Costales D, Guerrero Garzón JF, Gren T, Baggesgaard Sterndorff E, Jiang X, Sparholt Jo Rgensen T, Blin K, Fernández-Pastor I, Tormo JR, Martín J, Sánchez P, Moreno MDLC, Reyes F, Genilloud O, Weber T. Genome-Led Discovery of the Antibacterial Cyclic Lipopeptide Kutzneridine A and Its Silent Biosynthetic Gene Cluster from Kutzneria Species. JOURNAL OF NATURAL PRODUCTS 2024; 87:2515-2522. [PMID: 39332023 DOI: 10.1021/acs.jnatprod.4c00633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/29/2024]
Abstract
Genome analysis of Kutzneria sp. CA-103260 revealed a putative lipopeptide-encoding biosynthetic gene cluster (BGC) that was cloned into a bacterial artificial chromosome (BAC) and heterologously expressed in Streptomyces coelicolor M1152. As a result, a novel cyclic lipo-tetrapeptide containing two diaminopropionic acid residues and an exotic N,N-acetonide ring, kutzneridine A (1), was isolated and structurally characterized. Evaluation of the extraction conditions and isotope-labeling chemical modifications showed that the acetonide ring originated from acetone during isolation. The BGC was analyzed in silico and a biosynthetic pathway to 1 was proposed. Kutzneridine A displayed remarkable antibacterial activity against methicillin-resistant Staphylococcus aureus and vancomycin-resistant Enterococci.
Collapse
Affiliation(s)
- Francisco Javier Ortiz-López
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía, Avda. del Conocimiento 34, 18016 Armilla Granada, Spain
| | - Daniel Oves-Costales
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía, Avda. del Conocimiento 34, 18016 Armilla Granada, Spain
| | - Jaime Felipe Guerrero Garzón
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, So̷ltoft Plads, building 220, 2800 Kgs. Lyngby, Denmark
| | - Tetiana Gren
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, So̷ltoft Plads, building 220, 2800 Kgs. Lyngby, Denmark
| | - Eva Baggesgaard Sterndorff
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, So̷ltoft Plads, building 220, 2800 Kgs. Lyngby, Denmark
| | - Xinglin Jiang
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, So̷ltoft Plads, building 220, 2800 Kgs. Lyngby, Denmark
| | - Tue Sparholt Jo Rgensen
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, So̷ltoft Plads, building 220, 2800 Kgs. Lyngby, Denmark
| | - Kai Blin
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, So̷ltoft Plads, building 220, 2800 Kgs. Lyngby, Denmark
| | - Ignacio Fernández-Pastor
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía, Avda. del Conocimiento 34, 18016 Armilla Granada, Spain
| | - José R Tormo
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía, Avda. del Conocimiento 34, 18016 Armilla Granada, Spain
| | - Jesús Martín
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía, Avda. del Conocimiento 34, 18016 Armilla Granada, Spain
| | - Pilar Sánchez
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía, Avda. del Conocimiento 34, 18016 Armilla Granada, Spain
| | - Mercedes de la Cruz Moreno
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía, Avda. del Conocimiento 34, 18016 Armilla Granada, Spain
| | - Fernando Reyes
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía, Avda. del Conocimiento 34, 18016 Armilla Granada, Spain
| | - Olga Genilloud
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía, Avda. del Conocimiento 34, 18016 Armilla Granada, Spain
| | - Tilmann Weber
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, So̷ltoft Plads, building 220, 2800 Kgs. Lyngby, Denmark
| |
Collapse
|
2
|
Thombare VJ, Swarbrick JD, Azad MAK, Zhu Y, Lu J, Yu HY, Wickremasinghe H, He X, Bandiatmakur M, Li R, Bergen PJ, Velkov T, Wang J, Roberts KD, Li J, Patil NA. Exploring Structure-Activity Relationships and Modes of Action of Laterocidine. ACS CENTRAL SCIENCE 2024; 10:1703-1717. [PMID: 39345814 PMCID: PMC11428279 DOI: 10.1021/acscentsci.4c00776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 07/05/2024] [Accepted: 07/22/2024] [Indexed: 10/01/2024]
Abstract
A significant increase in life-threatening infections caused by Gram-negative "superbugs" is a serious threat to global health. With a dearth of new antibiotics in the developmental pipeline, antibiotics with novel mechanisms of action are urgently required to prevent a return to the preantibiotic era. A key strategy to develop novel anti-infective treatments is to discover new natural scaffolds with distinct mechanisms of action. Laterocidine is a unique cyclic lipodepsipeptide with activity against multiple problematic multidrug-resistant Gram-negative pathogens, including Pseudomonas aeruginosa, Acinetobacter baumannii, and Enterobacterales. Here, we developed a total chemical synthesis methodology for laterocidine and undertook systematic structure-activity relationship studies with chemical biology and NMR. We discovered important structural features that drive the antimicrobial activity of laterocidine, leading to the discovery of an engineered peptide surpassing the efficacy of the original peptide. This engineered peptide demonstrated complete inhibition of the growth of a polymyxin-resistant strain of Pseudomonas aeruginosa in static time-kill experiments.
Collapse
Affiliation(s)
- Varsha J Thombare
- Biomedicine Discovery Institute, Infection Program and Department of Pharmacology and Infection Program and Department of Microbiology, Monash University, Melbourne, VIC 3800, Australia
| | - James D Swarbrick
- Biomedicine Discovery Institute, Infection Program and Department of Pharmacology and Infection Program and Department of Microbiology, Monash University, Melbourne, VIC 3800, Australia
| | - Mohammad A K Azad
- Biomedicine Discovery Institute, Infection Program and Department of Pharmacology and Infection Program and Department of Microbiology, Monash University, Melbourne, VIC 3800, Australia
| | - Yan Zhu
- Biomedicine Discovery Institute, Infection Program and Department of Pharmacology and Infection Program and Department of Microbiology, Monash University, Melbourne, VIC 3800, Australia
| | - Jing Lu
- Biomedicine Discovery Institute, Infection Program and Department of Pharmacology and Infection Program and Department of Microbiology, Monash University, Melbourne, VIC 3800, Australia
| | - Heidi Y Yu
- Biomedicine Discovery Institute, Infection Program and Department of Pharmacology and Infection Program and Department of Microbiology, Monash University, Melbourne, VIC 3800, Australia
| | - Hasini Wickremasinghe
- Biomedicine Discovery Institute, Infection Program and Department of Pharmacology and Infection Program and Department of Microbiology, Monash University, Melbourne, VIC 3800, Australia
| | - Xiaoji He
- Biomedicine Discovery Institute, Infection Program and Department of Pharmacology and Infection Program and Department of Microbiology, Monash University, Melbourne, VIC 3800, Australia
| | - Mahimna Bandiatmakur
- Biomedicine Discovery Institute, Infection Program and Department of Pharmacology and Infection Program and Department of Microbiology, Monash University, Melbourne, VIC 3800, Australia
| | - Rong Li
- Biomedicine Discovery Institute, Infection Program and Department of Pharmacology and Infection Program and Department of Microbiology, Monash University, Melbourne, VIC 3800, Australia
| | - Phillip J Bergen
- Biomedicine Discovery Institute, Infection Program and Department of Pharmacology and Infection Program and Department of Microbiology, Monash University, Melbourne, VIC 3800, Australia
| | - Tony Velkov
- Biomedicine Discovery Institute, Infection Program and Department of Pharmacology and Infection Program and Department of Microbiology, Monash University, Melbourne, VIC 3800, Australia
| | - Jiping Wang
- Biomedicine Discovery Institute, Infection Program and Department of Pharmacology and Infection Program and Department of Microbiology, Monash University, Melbourne, VIC 3800, Australia
| | - Kade D Roberts
- Biomedicine Discovery Institute, Infection Program and Department of Pharmacology and Infection Program and Department of Microbiology, Monash University, Melbourne, VIC 3800, Australia
| | - Jian Li
- Biomedicine Discovery Institute, Infection Program and Department of Pharmacology and Infection Program and Department of Microbiology, Monash University, Melbourne, VIC 3800, Australia
| | - Nitin A Patil
- Biomedicine Discovery Institute, Infection Program and Department of Pharmacology and Infection Program and Department of Microbiology, Monash University, Melbourne, VIC 3800, Australia
| |
Collapse
|
3
|
Iqbal S, Begum F, Alfaifi MY, Elbehairi SEI, Siddique A, Shaw P. Exploring Antimicrobial Potency, ADMET, and Optimal Drug Target of a Non-ribosomal Peptide Sevadicin from Bacillus pumilus, through In Vitro Assay and Molecular Dynamics Simulation. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10355-8. [PMID: 39316258 DOI: 10.1007/s12602-024-10355-8] [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] [Accepted: 08/27/2024] [Indexed: 09/25/2024]
Abstract
The current study was designed to explore the biosynthetic potential of sevadicin in Bacillus pumilus species and its interaction with bacterial drug target molecules. The non-ribosomal peptide (NRP) cluster in B. pumilus SF-4 was preliminarily confirmed using PCR-based screening, and the bioactivity of strain SF-4 culture extract was assessed against a set of human pathogenic strains. The susceptibility assay showed that strain SF-4 extract had higher inhibitory concentrations (312-375 µg/mL) than ciprofloxacin. Genome mining of B. pumilus strains (n = 22) using AntiSMASH and BAGEL identified sevadicin coding biosynthetic gene cluster only in strain SF-4, constitutes of two core biosynthetic genes, three additional biosynthetic genes, two transport-related genes, and one regulatory gene. The molecular docking of sevadicin with various putative bacterial drug targets such as dihydropteroate, muramyl ligase E, topoisomerase, penicillin-binding protein, and in vitro safety analyses were conducted with detailed ADMET screening. The results showed that sevadicin makes hydrophobic interaction with MurE (PDB ID: 1E8C and 4C13) via hydrogen bonding, suggesting bacterial growth inhibition by disrupting the cell wall synthesis pathway and exhibiting a secure biosafety profile. The stability and compactness of sevadicin/MurE complexes were assessed via molecular dynamic simulation using RMSD, RMSF, and Rg. The simulation results revealed the binding stability of sevadicin/MurE complexes and indicated that the complexes can't be easily deformed. In conclusion, the current study explored the biosynthesis of sevadicin in B. pumilus for the first time and found that sevadicin inhibits bacterial growth by inhibiting cell wall synthesis via targeting the MurE enzyme and exhibits no toxicity.
Collapse
Affiliation(s)
- Sajid Iqbal
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang, P.R. China
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China
| | - Farida Begum
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang, P.R. China
| | - Mohammad Y Alfaifi
- Biology Department, Faculty of Science, King Khalid University, Abha, 9004, Saudi Arabia
- Tissue Culture and Cancer Biology Research Laboratory, King Khalid University, Abha, 9004, Saudi Arabia
| | - Serag Eldin I Elbehairi
- Biology Department, Faculty of Science, King Khalid University, Abha, 9004, Saudi Arabia
- Tissue Culture and Cancer Biology Research Laboratory, King Khalid University, Abha, 9004, Saudi Arabia
| | - Abubakar Siddique
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, P.R. China
| | - Peter Shaw
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang, P.R. China.
| |
Collapse
|
4
|
Chen N, Cai P, Zhang D, Zhang J, Zhong Z, Li YX. Metabolic engineering of "last-line antibiotic" colistin in Paenibacillus polymyxa. Metab Eng 2024; 85:35-45. [PMID: 39019251 DOI: 10.1016/j.ymben.2024.07.005] [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: 04/10/2024] [Revised: 07/12/2024] [Accepted: 07/14/2024] [Indexed: 07/19/2024]
Abstract
Colistin, also known as polymyxin E, is a lipopeptide antibiotic used to treat infections caused by multidrug-resistant gram-negative bacteria. It is considered a "last-line antibiotic", but its clinical development is hindered by low titer and impurities resulting from the presence of diverse homologs in microbial fermentation. To ensure consistent pharmaceutical activity and kinetics, it is crucial to have high-purity colistin active pharmaceutical ingredient (API) in the pharmaceutical industry. This study focused on the metabolic engineering of a natural colistin producer strain to produce colistin with a high titer and purity. Guided by genome mining, we identified Paenibacillus polymyxa ATCC 842 as a natural colistin producer capable of generating a high proportion of colistin A. By systematically inactivating seven non-essential biosynthetic gene clusters (BGCs) of peptide metabolites that might compete precursors with colistin or inhibit colistin production, we created an engineered strain, P14, which exhibited an 82% increase in colistin titer and effectively eliminated metabolite impurities such as tridecaptin, paenibacillin, and paenilan. Additionally, we engineered the L-2,4-diaminobutyric acid (L-2,4-DABA) pathway to further enhance colistin production, resulting in the engineered strain P19, which boosted a remarkable colistin titer of 649.3 mg/L - a 269% improvement compared to the original strain. By concurrently feeding L-isoleucine and L-leucine, we successfully produced high-purity colistin A, constituting 88% of the total colistin products. This study highlights the potential of metabolic engineering in improving the titer and purity of lipopeptide antibiotics in the non-model strain, making them more suitable for clinical use. These findings indicate that efficiently producing colistin API in high purity directly from fermentation can now be achieved in a straightforward manner.
Collapse
Affiliation(s)
- Nanzhu Chen
- Department of Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Peiyan Cai
- Department of Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Dengwei Zhang
- Department of Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Junliang Zhang
- Department of Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Zheng Zhong
- Department of Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Yong-Xin Li
- Department of Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong, China.
| |
Collapse
|
5
|
Chen J, Wang W, Hu X, Yue Y, Lu X, Wang C, Wei B, Zhang H, Wang H. Medium-sized peptides from microbial sources with potential for antibacterial drug development. Nat Prod Rep 2024; 41:1235-1263. [PMID: 38651516 DOI: 10.1039/d4np00002a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Abstract
Covering: 1993 to the end of 2022As the rapid development of antibiotic resistance shrinks the number of clinically available antibiotics, there is an urgent need for novel options to fill the existing antibiotic pipeline. In recent years, antimicrobial peptides have attracted increased interest due to their impressive broad-spectrum antimicrobial activity and low probability of antibiotic resistance. However, macromolecular antimicrobial peptides of plant and animal origin face obstacles in antibiotic development because of their extremely short elimination half-life and poor chemical stability. Herein, we focus on medium-sized antibacterial peptides (MAPs) of microbial origin with molecular weights below 2000 Da. The low molecular weight is not sufficient to form complex protein conformations and is also associated to a better chemical stability and easier modifications. Microbially-produced peptides are often composed of a variety of non-protein amino acids and terminal modifications, which contribute to improving the elimination half-life of compounds. Therefore, MAPs have great potential for drug discovery and are likely to become key players in the development of next-generation antibiotics. In this review, we provide a detailed exploration of the modes of action demonstrated by 45 MAPs and offer a concise summary of the structure-activity relationships observed in these MAPs.
Collapse
Affiliation(s)
- Jianwei Chen
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China
| | - Wei Wang
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China
| | - Xubin Hu
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yujie Yue
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China
| | - Xingyue Lu
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China
| | - Chenjie Wang
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China
| | - Bin Wei
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China
| | - Huawei Zhang
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China
| | - Hong Wang
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China
| |
Collapse
|
6
|
Chunduru J, LaRoe N, Garza J, Hamood AN, Paré PW. Nosocomial Bacteria Inhibition with Polymyxin B: In Silico Gene Mining and In Vitro Analysis. Antibiotics (Basel) 2024; 13:745. [PMID: 39200045 PMCID: PMC11350920 DOI: 10.3390/antibiotics13080745] [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: 07/11/2024] [Revised: 08/03/2024] [Accepted: 08/06/2024] [Indexed: 09/01/2024] Open
Abstract
Multidrug-resistant bacteria present a significant public health challenge; such pathogens exhibit reduced susceptibility to conventional antibiotics, limiting current treatment options. Cationic non-ribosomal peptides (CNRPs) such as brevicidine and polymyxins have emerged as promising candidates to block Gram-negative bacteria. To investigate the capability of bacteria to biosynthesize CNRPs, and specifically polymyxins, over 11,000 bacterial genomes were mined in silico. Paenibacillus polymyxa was identified as having a robust biosynthetic capacity, based on multiple polymyxin gene clusters. P. polymyxa biosynthetic competence was confirmed by metabolite characterization via HPLC purification and MALDI TOF/TOF analysis. When grown in a selected medium, the metabolite yield was 4 mg/L with a 20-fold specific activity increase. Polymyxin B (PMB) was assayed with select nosocomial pathogens, including Pseudomonas aeruginosa, Klebsiella pneumonia, and Acinetobacter baumaii, which exhibited minimum inhibitory concentrations of 4, 1, and 1 µg/mL, respectively.
Collapse
Affiliation(s)
- Jayendra Chunduru
- Chemistry & Biochemistry Department, Texas Tech University, Lubbock, TX 79409, USA
| | - Nicholas LaRoe
- Chemistry & Biochemistry Department, Texas Tech University, Lubbock, TX 79409, USA
| | - Jeremy Garza
- Department of Immunology & Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA (A.N.H.)
| | - Abdul N. Hamood
- Department of Immunology & Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA (A.N.H.)
| | - Paul W. Paré
- Chemistry & Biochemistry Department, Texas Tech University, Lubbock, TX 79409, USA
| |
Collapse
|
7
|
Zhang MY, Li S, Wang CY, Zhang ZX, Han YL, Liu XQ, Cheng J, Zhou XY, Zhang YX. A novel antimicrobial peptide S24 combats serious wound infections caused by Pseudomonas aeruginosa and Acinetobacter baumannii. J Antimicrob Chemother 2024; 79:1951-1961. [PMID: 38863365 DOI: 10.1093/jac/dkae191] [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: 01/08/2024] [Accepted: 05/23/2024] [Indexed: 06/13/2024] Open
Abstract
OBJECTIVES Pseudomonas aeruginosa and Acinetobacter baumannii are ranked as top-priority organisms by WHO. Antimicrobial peptides (AMPs) are promising antimicrobial agents that are highly effective against serious bacterial infections. METHODS In our previous study, a series of α-helical AMPs were screened using a novel multiple-descriptor strategy. The current research suggested that S24 exhibited strong antimicrobial activity against major pathogenic bacteria, and displayed minimal haemolysis, good serum stability and maintained salt resistance. RESULTS We found that S24 exerted an antimicrobial effect by destroying outer membrane permeability and producing a strong binding effect on bacterial genomic DNA that inhibits genomic DNA migration. Furthermore, S24 exerted a strong ability to promote healing in wound infected by P. aeruginosa, A. baumannii and mixed strains in a mouse model. CONCLUSIONS Overall, S24 showed good stability under physiological conditions and excellent antimicrobial activity, suggesting it may be a potential candidate for the development of serious bacterial infection treatment.
Collapse
Affiliation(s)
- Meng-Yue Zhang
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Shuang Li
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Cai-Yun Wang
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Zi-Xuan Zhang
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yu-Ling Han
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xin-Qi Liu
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Juan Cheng
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xun-Yong Zhou
- Weihuakang (Shenzhen) Biotech. Co., Ltd., Shenzhen 518001, China
| | - Yi-Xuan Zhang
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
8
|
Kifle BA, Sime AM, Gemeda MT, Woldesemayat AA. Shotgun metagenomic insights into secondary metabolite biosynthetic gene clusters reveal taxonomic and functional profiles of microbiomes in natural farmland soil. Sci Rep 2024; 14:15096. [PMID: 38956049 PMCID: PMC11220033 DOI: 10.1038/s41598-024-63254-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Accepted: 05/27/2024] [Indexed: 07/04/2024] Open
Abstract
Antibiotic resistance is a worldwide problem that imposes a devastating effect on developing countries and requires immediate interventions. Initially, most of the antibiotic drugs were identified by culturing soil microbes. However, this method is prone to discovering the same antibiotics repeatedly. The present study employed a shotgun metagenomics approach to investigate the taxonomic diversity, functional potential, and biosynthetic capacity of microbiomes from two natural agricultural farmlands located in Bekeka and Welmera Choke Kebelle in Ethiopia for the first time. Analysis of the small subunit rRNA revealed bacterial domain accounting for 83.33% and 87.24% in the two selected natural farmlands. Additionally, the analysis showed the dominance of Proteobacteria representing 27.27% and 28.79% followed by Actinobacteria making up 12.73% and 13.64% of the phyla composition. Furthermore, the analysis revealed the presence of unassigned bacteria in the studied samples. The metagenome functional analysis showed 176,961 and 104, 636 number of protein-coding sequences (pCDS) from the two samples found a match with 172,655 and 102, 275 numbers of InterPro entries, respectively. The Genome ontology annotation suggests the presence of 5517 and 3293 pCDS assigned to the "biosynthesis process". Numerous Kyoto Encyclopedia of Genes and Genomes modules (KEGG modules) involved in the biosynthesis of terpenoids and polyketides were identified. Furthermore, both known and novel Biosynthetic gene clusters, responsible for the production of secondary metabolites, such as polyketide synthases, non-ribosomal peptide synthetase, ribosomally synthesized and post-translationally modified peptides (Ripp), and Terpene, were discovered. Generally, from the results it can be concluded that the microbiomes in the selected sampling sites have a hidden functional potential for the biosynthesis of secondary metabolites. Overall, this study can serve as a strong preliminary step in the long journey of bringing new antibiotics to the market.
Collapse
Affiliation(s)
- Bezayit Amare Kifle
- Department of Biotechnology, College of Biological and Chemical Engineering, Addis Ababa Science and Technology University, Addis Ababa, Ethiopia
| | - Amsale Melkamu Sime
- Department of Biotechnology, College of Biological and Chemical Engineering, Addis Ababa Science and Technology University, Addis Ababa, Ethiopia
| | - Mesfin Tafesse Gemeda
- Department of Biotechnology, College of Biological and Chemical Engineering, Addis Ababa Science and Technology University, Addis Ababa, Ethiopia
| | - Adugna Abdi Woldesemayat
- Department of Biotechnology, College of Biological and Chemical Engineering, Addis Ababa Science and Technology University, Addis Ababa, Ethiopia.
| |
Collapse
|
9
|
Zeng P, Wang H, Zhang P, Leung SSY. Unearthing naturally-occurring cyclic antibacterial peptides and their structural optimization strategies. Biotechnol Adv 2024; 73:108371. [PMID: 38704105 DOI: 10.1016/j.biotechadv.2024.108371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 03/08/2024] [Accepted: 04/29/2024] [Indexed: 05/06/2024]
Abstract
Natural products with antibacterial activity are highly desired globally to combat against multidrug-resistant (MDR) bacteria. Antibacterial peptide (ABP), especially cyclic ABP (CABP), is one of the abundant classes. Most of them were isolated from microbes, demonstrating excellent bactericidal effects. With the improved proteolytic stability, CABPs are normally considered to have better druggability than linear peptides. However, most clinically-used CABP-based antibiotics, such as colistin, also face the challenges of drug resistance soon after they reached the market, urgently requiring the development of next-generation succedaneums. We present here a detail review on the novel naturally-occurring CABPs discovered in the past decade and some of them are under clinical trials, exhibiting anticipated application potential. According to their chemical structures, they were broadly classified into five groups, including (i) lactam/lactone-based CABPs, (ii) cyclic lipopeptides, (iii) glycopeptides, (iv) cyclic sulfur-rich peptides and (v) multiple-modified CABPs. Their chemical structures, antibacterial spectrums and proposed mechanisms are discussed. Moreover, engineered analogs of these novel CABPs are also summarized to preliminarily analyze their structure-activity relationship. This review aims to provide a global perspective on research and development of novel CABPs to highlight the effectiveness of derivatives design in identifying promising antibacterial agents. Further research efforts in this area are believed to play important roles in fighting against the multidrug-resistance crisis.
Collapse
Affiliation(s)
- Ping Zeng
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Honglan Wang
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Pengfei Zhang
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Sharon Shui Yee Leung
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong.
| |
Collapse
|
10
|
Zhao X, Zhong X, Yang S, Deng J, Deng K, Huang Z, Li Y, Yin Z, Liu Y, Viel JH, Wan H. Guiding antibiotics towards their target using bacteriophage proteins. Nat Commun 2024; 15:5287. [PMID: 38902231 PMCID: PMC11190222 DOI: 10.1038/s41467-024-49603-4] [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: 12/18/2023] [Accepted: 06/11/2024] [Indexed: 06/22/2024] Open
Abstract
Novel therapeutic strategies against difficult-to-treat bacterial infections are desperately needed, and the faster and cheaper way to get them might be by repurposing existing antibiotics. Nanodelivery systems enhance the efficacy of antibiotics by guiding them to their targets, increasing the local concentration at the site of infection. While recently described nanodelivery systems are promising, they are generally not easy to adapt to different targets, and lack biocompatibility or specificity. Here, nanodelivery systems are created that source their targeting proteins from bacteriophages. Bacteriophage receptor-binding proteins and cell-wall binding domains are conjugated to nanoparticles, for the targeted delivery of rifampicin, imipenem, and ampicillin against bacterial pathogens. They show excellent specificity against their targets, and accumulate at the site of infection to deliver their antibiotic payload. Moreover, the nanodelivery systems suppress pathogen infections more effectively than 16 to 32-fold higher doses of free antibiotics. This study demonstrates that bacteriophage sourced targeting proteins are promising candidates to guide nanodelivery systems. Their specificity, availability, and biocompatibility make them great options to guide the antibiotic nanodelivery systems that are desperately needed to combat difficult-to-treat infections.
Collapse
Affiliation(s)
- Xinghong Zhao
- Center for Sustainable Antimicrobials, Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China.
- Center for Infectious Diseases Control (CIDC), College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China.
| | - Xinyi Zhong
- Center for Sustainable Antimicrobials, Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
- Center for Infectious Diseases Control (CIDC), College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Shinong Yang
- Center for Sustainable Antimicrobials, Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
- Center for Infectious Diseases Control (CIDC), College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Jiarong Deng
- Center for Sustainable Antimicrobials, Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
- Center for Infectious Diseases Control (CIDC), College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Kai Deng
- Center for Sustainable Antimicrobials, Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
- Center for Infectious Diseases Control (CIDC), College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Zhengqun Huang
- Center for Sustainable Antimicrobials, Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
- Center for Infectious Diseases Control (CIDC), College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Yuanfeng Li
- Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Zhongqiong Yin
- Center for Sustainable Antimicrobials, Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China.
| | - Yong Liu
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325001, China.
| | - Jakob H Viel
- Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9747AG, Groningen, Netherlands
| | - Hongping Wan
- Center for Sustainable Antimicrobials, Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China.
- Center for Infectious Diseases Control (CIDC), College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China.
| |
Collapse
|
11
|
Cheng Z, He BB, Lei K, Gao Y, Shi Y, Zhong Z, Liu H, Liu R, Zhang H, Wu S, Zhang W, Tang X, Li YX. Rule-based omics mining reveals antimicrobial macrocyclic peptides against drug-resistant clinical isolates. Nat Commun 2024; 15:4901. [PMID: 38851779 PMCID: PMC11162475 DOI: 10.1038/s41467-024-49215-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 05/28/2024] [Indexed: 06/10/2024] Open
Abstract
Antimicrobial resistance remains a significant global threat, driving up mortality rates worldwide. Ribosomally synthesized and post-translationally modified peptides have emerged as a promising source of novel peptide antibiotics due to their diverse chemical structures. Here, we report the discovery of new aminovinyl-(methyl)cysteine (Avi(Me)Cys)-containing peptide antibiotics through a synergistic approach combining biosynthetic rule-based omics mining and heterologous expression. We first bioinformatically identify 1172 RiPP biosynthetic gene clusters (BGCs) responsible for Avi(Me)Cys-containing peptides formation from a vast pool of over 50,000 bacterial genomes. Subsequently, we successfully establish the connection between three identified BGCs and the biosynthesis of five peptide antibiotics via biosynthetic rule-guided metabolic analysis. Notably, we discover a class V lanthipeptide, massatide A, which displays excellent activity against gram-positive pathogens, including drug-resistant clinical isolates like linezolid-resistant S. aureus and methicillin-resistant S. aureus, with a minimum inhibitory concentration of 0.25 μg/mL. The remarkable performance of massatide A in an animal infection model, coupled with a relatively low risk of resistance and favorable safety profile, positions it as a promising candidate for antibiotic development. Our study highlights the potential of Avi(Me)Cys-containing peptides in expanding the arsenal of antibiotics against multi-drug-resistant bacteria, offering promising drug leads in the ongoing battle against infectious diseases.
Collapse
Affiliation(s)
- Zhuo Cheng
- Department of Chemistry and The Swire Institute of Marine Science, The University of Hong Kong, Pokfulam Road, Hong Kong, China
- Institute of Chemical Biology, Shenzhen Bay Laboratory, Shenzhen, 515832, China
| | - Bei-Bei He
- Department of Chemistry and The Swire Institute of Marine Science, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Kangfan Lei
- 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
| | - Ying Gao
- Department of Chemistry and The Swire Institute of Marine Science, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Yuqi Shi
- Department of Chemistry and The Swire Institute of Marine Science, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Zheng Zhong
- Department of Chemistry and The Swire Institute of Marine Science, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Hongyan Liu
- Department of Chemistry and The Swire Institute of Marine Science, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Runze Liu
- Department of Chemistry and The Swire Institute of Marine Science, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Haili Zhang
- Institute of Chemical Biology, Shenzhen Bay Laboratory, Shenzhen, 515832, 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
| | - Wenxuan 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.
| | - Xiaoyu Tang
- Institute of Chemical Biology, Shenzhen Bay Laboratory, Shenzhen, 515832, China.
| | - Yong-Xin Li
- Department of Chemistry and The Swire Institute of Marine Science, The University of Hong Kong, Pokfulam Road, Hong Kong, China.
| |
Collapse
|
12
|
Buchholz KR, Reichelt M, Johnson MC, Robinson SJ, Smith PA, Rutherford ST, Quinn JG. Potent activity of polymyxin B is associated with long-lived super-stoichiometric accumulation mediated by weak-affinity binding to lipid A. Nat Commun 2024; 15:4733. [PMID: 38830951 PMCID: PMC11148078 DOI: 10.1038/s41467-024-49200-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 05/23/2024] [Indexed: 06/05/2024] Open
Abstract
Polymyxins are gram-negative antibiotics that target lipid A, the conserved membrane anchor of lipopolysaccharide in the outer membrane. Despite their clinical importance, the molecular mechanisms underpinning polymyxin activity remain unresolved. Here, we use surface plasmon resonance to kinetically interrogate interactions between polymyxins and lipid A and derive a phenomenological model. Our analyses suggest a lipid A-catalyzed, three-state mechanism for polymyxins: transient binding, membrane insertion, and super-stoichiometric cluster accumulation with a long residence time. Accumulation also occurs for brevicidine, another lipid A-targeting antibacterial molecule. Lipid A modifications that impart polymyxin resistance and a non-bactericidal polymyxin derivative exhibit binding that does not evolve into long-lived species. We propose that transient binding to lipid A permeabilizes the outer membrane and cluster accumulation enables the bactericidal activity of polymyxins. These findings could establish a blueprint for discovery of lipid A-targeting antibiotics and provide a generalizable approach to study interactions with the gram-negative outer membrane.
Collapse
Affiliation(s)
- Kerry R Buchholz
- Department of Infectious Diseases, Genentech, Inc., South San Francisco, CA, USA.
| | - Mike Reichelt
- Department of Pathology, Genentech, Inc., South San Francisco, CA, USA
| | - Matthew C Johnson
- Department of Structural Biology, Genentech, Inc., South San Francisco, CA, USA
| | - Sarah J Robinson
- Department of Discovery Chemistry, Genentech, Inc., South San Francisco, CA, USA
| | - Peter A Smith
- Department of Infectious Diseases, Genentech, Inc., South San Francisco, CA, USA
- Revagenix, Inc., San Mateo, CA, USA
| | - Steven T Rutherford
- Department of Infectious Diseases, Genentech, Inc., South San Francisco, CA, USA.
| | - John G Quinn
- Department of Biochemical and Cellular Pharmacology, Genentech, Inc., South San Francisco, CA, USA.
| |
Collapse
|
13
|
Palpal-Latoc D, Horsfall AJ, Cameron AJ, Campbell G, Ferguson SA, Cook GM, Sander V, Davidson AJ, Harris PWR, Brimble MA. Synthesis, Structure-Activity Relationship Study, Bioactivity, and Nephrotoxicity Evaluation of the Proposed Structure of the Cyclic Lipodepsipeptide Brevicidine B. JOURNAL OF NATURAL PRODUCTS 2024; 87:764-773. [PMID: 38423998 DOI: 10.1021/acs.jnatprod.3c00876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
The brevicidines represent a novel class of nonribosomal antimicrobial peptides that possess remarkable potency and selectivity toward highly problematic and resistant Gram-negative pathogenic bacteria. A recently discovered member of the brevicidine family, coined brevicidine B (2), comprises a single amino acid substitution (from d-Tyr2 to d-Phe2) in the amino acid sequence of the linear moiety of brevicidine (1) and was reported to exhibit broader antimicrobial activity against both Gram-negative (MIC = 2-4 μgmL-1) and Gram-positive (MIC = 2-8 μgmL-1) pathogens. Encouraged by this, we herein report the first total synthesis of the proposed structure of brevicidine B (2), building on our previously reported synthetic strategy to access brevicidine (1). In agreement with the original isolation paper, pleasingly, synthetic 2 demonstrated antimicrobial activity toward Escherichia coli, Pseudomonas aeruginosa, and Klebsiella pneumoniae (MIC = 4-8 μgmL-1). Interestingly, however, synthetic 2 was inactive toward all of the tested Gram-positive pathogens, including methicillin-resistant Staphylococcus aureus strains. Substitution of d-Phe2 with its enantiomer, and other hydrophobic residues, yields analogues that were either inactive or only exhibited activity toward Gram-negative strains. The striking difference in the biological activity of our synthetic 2 compared to the reported natural compound warrants the re-evaluation of the original natural product for purity or possible differences in relative configuration. Finally, the evaluation of synthetic 1 and 2 in a human kidney organoid model of nephrotoxicity revealed substantial toxicity of both compounds, although 1 was less toxic than 2 and polymyxin B. These results indicate that modification to position 2 may afford a strategy to mitigate the nephrotoxicity of brevicidine.
Collapse
Affiliation(s)
- Dennise Palpal-Latoc
- School of Chemical Sciences, The University of Auckland, 23 Symonds Street, Auckland 1010, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, 3 Symonds Street, Auckland 1010, New Zealand
- School of Biological Sciences, The University of Auckland, 3A Symonds Street, Auckland 1010, New Zealand
| | - Aimee J Horsfall
- School of Chemical Sciences, The University of Auckland, 23 Symonds Street, Auckland 1010, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, 3 Symonds Street, Auckland 1010, New Zealand
- School of Biological Sciences, The University of Auckland, 3A Symonds Street, Auckland 1010, New Zealand
| | - Alan J Cameron
- School of Chemical Sciences, The University of Auckland, 23 Symonds Street, Auckland 1010, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, 3 Symonds Street, Auckland 1010, New Zealand
- School of Biological Sciences, The University of Auckland, 3A Symonds Street, Auckland 1010, New Zealand
| | - Georgia Campbell
- Department of Microbiology and Immunology, School of Medical Sciences, The University of Otago, 720 Cumberland Street, Dunedin 9054, New Zealand
| | - Scott A Ferguson
- Department of Microbiology and Immunology, School of Medical Sciences, The University of Otago, 720 Cumberland Street, Dunedin 9054, New Zealand
| | - Gregory M Cook
- Department of Microbiology and Immunology, School of Medical Sciences, The University of Otago, 720 Cumberland Street, Dunedin 9054, New Zealand
| | - Veronika Sander
- Faculty of Medical and Health Sciences, The University of Auckland 85 Park Road, Grafton, Auckland 1023, New Zealand
| | - Alan J Davidson
- Faculty of Medical and Health Sciences, The University of Auckland 85 Park Road, Grafton, Auckland 1023, New Zealand
| | - Paul W R Harris
- School of Chemical Sciences, The University of Auckland, 23 Symonds Street, Auckland 1010, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, 3 Symonds Street, Auckland 1010, New Zealand
- School of Biological Sciences, The University of Auckland, 3A Symonds Street, Auckland 1010, New Zealand
| | - Margaret A Brimble
- School of Chemical Sciences, The University of Auckland, 23 Symonds Street, Auckland 1010, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, 3 Symonds Street, Auckland 1010, New Zealand
- School of Biological Sciences, The University of Auckland, 3A Symonds Street, Auckland 1010, New Zealand
| |
Collapse
|
14
|
Yang P, Mao W, Zhang J, Yang Y, Zhang F, Ouyang X, Li B, Wu X, Ba Z, Ran K, Tian Y, Liu H, Zhang Y, Gou S, Zhong C, Ni J. A novel antimicrobial peptide with broad-spectrum and exceptional stability derived from the natural peptide Brevicidine. Eur J Med Chem 2024; 269:116337. [PMID: 38537511 DOI: 10.1016/j.ejmech.2024.116337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 03/01/2024] [Accepted: 03/14/2024] [Indexed: 04/07/2024]
Abstract
The global issue of antibiotic resistance is increasingly severe, highlighting the urgent necessity for the development of new antibiotics. Brevicidine, a natural cyclic lipopeptide, exhibits remarkable antimicrobial activity against Gram-negative bacteria. In this study, a comprehensive structure-activity relationship of Brevicidine was investigated through 20 newly synthesized cyclic lipopeptide analogs, resulting in the identification of an optimal linear analog 22. The sequence of analog 22 consisted of five d-amino acids and four non-natural amino acid 2,5-diaminovaleric acid (Orn) and conjugated with decanoic acid at N-terminal. Compared to Brevicidine, analog 22 was easier to synthesize, and exerted broad spectrum antimicrobial activity and excellent stability (t1/2 = 40.98 h). Additionally, analog 22 demonstrated a rapid bactericidal effect by permeating non-specifically through the bacterial membranes, thereby minimizing the likelihood of inducing resistance. Moreover, it exhibited remarkable efficacy in combating bacterial biofilms and reversing bacterial resistance to conventional antibiotics. Furthermore, it effectively suppressed the growth of bacteria in vital organs of mice infected with S. aureus ATCC 25923. In conclusion, analog 22 may represent a potential antimicrobial peptide for further optimization.
Collapse
Affiliation(s)
- Ping Yang
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China
| | - Wenbo Mao
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China
| | - Jingying Zhang
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China
| | - Yinyin Yang
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China
| | - Fangyan Zhang
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China
| | - Xu Ouyang
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China
| | - Beibei Li
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China
| | - Xiaoyan Wu
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China
| | - Zufang Ba
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China
| | - Kaixin Ran
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China
| | - Yali Tian
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China
| | - Hui Liu
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China; 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, PR China
| | - Yun Zhang
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China; 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, PR China
| | - Sanhu Gou
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China; 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, PR China
| | - Chao Zhong
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China; 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, PR China.
| | - Jingman Ni
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China; 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, PR China.
| |
Collapse
|
15
|
González-León Y, De la Vega-Camarillo E, Ramírez-Vargas R, Anducho-Reyes MA, Mercado-Flores Y. Whole genome analysis of Bacillus velezensis 160, biological control agent of corn head smut. Microbiol Spectr 2024; 12:e0326423. [PMID: 38363138 PMCID: PMC10986511 DOI: 10.1128/spectrum.03264-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 01/18/2024] [Indexed: 02/17/2024] Open
Abstract
Corn head smut is a disease caused by the fungus Sporisorium reilianum. This phytosanitary problem has existed for several decades in the Mezquital Valley, an important corn-producing area in central Mexico. To combat the problem, a strain identified as Bacillus subtilis 160 was applied in the field, where it decreased disease incidence and increased crop productivity. In this study, the sequencing and analysis of the whole genome sequence of this strain were carried out to identify its genetic determinants for the production of antimicrobials. The B. subtilis 160 strain was found to be Bacillus velezensis. Its genome has a size of 4,297,348 bp, a GC content of 45.8%, and 4,174 coding sequences. Comparative analysis with the genomes of four other B. velezensis strains showed that they share 2,804 genes and clusters for the production of difficidin, bacillibactin, bacilysin, macrolantin, bacillaene, fengycin, butirosin A, locillomycin, and surfactin. For the latter metabolite, unlike the other strains that have only one cluster, B. velezensis 160 has three. A cluster for synthesizing laterocidine, an antimicrobial reported only in Brevibacillus laterosporus, was also identified. IMPORTANCE In this study, we performed sequencing and analysis of the complete genome of the strain initially identified as Bacillus subtilis 160 as part of its characterization. This bacterium has shown its ability to control corn head smut in the field, a disease caused by the basidiomycete fungus Sporisorium reilianum. Analyzing the complete genome sequence not only provides a more precise taxonomic identification but also sheds light on the genetic potential of this bacterium, especially regarding mechanisms that allow it to exert biological control. Employing molecular and bioinformatics tools in studying the genomes of agriculturally significant microorganisms offers insights into the development of biofungicides and bioinoculants. These innovations aim to enhance plant growth and pave the way for strategies that boost crop productivity.
Collapse
|
16
|
Sun R, You R, Yu X, Zhao D, Li L. Discovery and Synthesis of a Gram-Negative-Active Cationic Lipopeptide Antibiotic Inspired by Primary Sequences from Underexplored Gram-Negative Bacteria. Org Lett 2024; 26:1348-1352. [PMID: 38341869 DOI: 10.1021/acs.orglett.3c04232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2024]
Abstract
The emergence of multidrug-resistant Gram-negative pathogens poses a serious threat to global health. Gram-negative bacteria have become increasingly recognized as underexplored sources of Gram-negative-active cationic lipopeptide (CLP) antibiotics. We systematically screened 8982 sequenced genomes from 42 underexplored Gram-negative bacterial genera and identified eight potential CLP biosynthetic gene clusters. Their predicted products were rapidly accessed by solid-phase total synthesis, which led to the novel antibiotic chospeptin with good activities against clinically isolated colistin-resistant Gram-negative pathogens.
Collapse
Affiliation(s)
- Runze Sun
- State Key Laboratory of Microbial Metabolism and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Ruixiang You
- State Key Laboratory of Microbial Metabolism and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
- College of Life Sciences, Shanghai Normal University, 100 Guilin Road, Shanghai 200234, China
| | - Xuchang Yu
- State Key Laboratory of Microbial Metabolism and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
- State Key Laboratory of Bioreactor Engineering and School of Biotechnology, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Di Zhao
- State Key Laboratory of Microbial Metabolism and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Lei Li
- State Key Laboratory of Microbial Metabolism and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| |
Collapse
|
17
|
Munawar S, Zahoor AF, Hussain SM, Ahmad S, Mansha A, Parveen B, Ali KG, Irfan A. Steglich esterification: A versatile synthetic approach toward the synthesis of natural products, their analogues/derivatives. Heliyon 2024; 10:e23416. [PMID: 38170008 PMCID: PMC10758822 DOI: 10.1016/j.heliyon.2023.e23416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 12/03/2023] [Accepted: 12/04/2023] [Indexed: 01/05/2024] Open
Abstract
The exploitation of natural products and their analogues in the field of pharmacology has been regarded as of great importance. It can be attributed to the fact that these scaffolds exhibit diverse chemical properties, distinct biological activities and zenith specificity in their biochemical processes, enabling them to act as favorable structures for lead compounds. The synthesis of natural products has been a crafty and hard-to-achieve task. Steglich esterification reaction has played a significant role in that area. It is a mild and efficient technique for constructing ester linkages. This technique involves the establishment of ester moiety via a carbodiimide-based condensation of a carboxylic acid with an alcohol, thiol or an amine catalyzed by dimethyl aminopyridine (DMAP). Specifically, labile reagents with multiple reactive sites are esterified efficiently with the classical and modified Steglich esterification conditions, which accounts for their synthetic utility. This review encloses the performance of the Steglich esterification reaction in forging the ester linkage for executing the total synthesis of natural products and their derivatives since 2018.
Collapse
Affiliation(s)
- Saba Munawar
- Department of Chemistry, Government College University Faisalabad, 38000, Faisalabad, Pakistan
| | - Ameer Fawad Zahoor
- Department of Chemistry, Government College University Faisalabad, 38000, Faisalabad, Pakistan
| | - Syed Makhdoom Hussain
- Department of Zoology, Government College University Faisalabad, 38000, Faisalabad, Pakistan
| | - Sajjad Ahmad
- Department of Chemistry, University of Engineering and Technology Lahore, Faisalabad Campus, 38000, Faisalabad, Pakistan
| | - Asim Mansha
- Department of Chemistry, Government College University Faisalabad, 38000, Faisalabad, Pakistan
| | - Bushra Parveen
- Department of Chemistry, Government College University Faisalabad, 38000, Faisalabad, Pakistan
| | - Kulsoom Ghulam Ali
- Department of Chemistry, Government College University Faisalabad, 38000, Faisalabad, Pakistan
| | - Ahmad Irfan
- Department of Chemistry, King Khalid University, Abha, 61413, P.O. Box 9004, Saudi Arabia
| |
Collapse
|
18
|
Zhong X, Deng K, Yang X, Song X, Zou Y, Zhou X, Tang H, Li L, Fu Y, Yin Z, Wan H, Zhao X. Brevicidine acts as an effective sensitizer of outer membrane-impermeable conventional antibiotics for Acinetobacter baumannii treatment. Front Microbiol 2023; 14:1304198. [PMID: 38173680 PMCID: PMC10762313 DOI: 10.3389/fmicb.2023.1304198] [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: 09/29/2023] [Accepted: 11/27/2023] [Indexed: 01/05/2024] Open
Abstract
The antibiotic resistance of Acinetobacter baumannii poses a significant threat to global public health, especially those strains that are resistant to carbapenems. Therefore, novel strategies are desperately needed for the treatment of infections caused by antibiotic-resistant A. baumannii. In this study, we report that brevicidine, a bacterial non-ribosomally produced cyclic lipopeptide, shows synergistic effects with multiple outer membrane-impermeable conventional antibiotics against A. baumannii. In particular, brevicidine, at a concentration of 1 μM, lowered the minimum inhibitory concentration of erythromycin, azithromycin, and rifampicin against A. baumannii strains by 32-128-fold. Furthermore, mechanistic studies were performed by employing erythromycin as an example of an outer membrane-impermeable conventional antibiotic, which showed the best synergistic effects with brevicidine against the tested A. baumannii strains in the present study. The results demonstrate that brevicidine disrupted the outer membrane of A. baumannii at a concentration range of 0.125-4 μM in a dose-dependent manner. This capacity of brevicidine could help the tested outer membrane-impermeable antibiotics enter A. baumannii cells and thereafter exert their antimicrobial activity. In addition, the results show that brevicidine-erythromycin combination exerted strong A. baumannii killing capacity by the enhanced inhibition of adenosine triphosphate biosynthesis and accumulation of reactive oxygen species, which are the main mechanisms causing the death of bacteria. Interestingly, brevicidine and erythromycin combination showed good therapeutic effects on A. baumannii-induced mouse peritonitis-sepsis models. These findings demonstrate that brevicidine is a promising sensitizer candidate of outer membrane-impermeable conventional antibiotics for treating A. baumannii infections in the post-antibiotic age.
Collapse
Affiliation(s)
- Xinyi Zhong
- Center for Sustainable Antimicrobials, Department of Pharmacy, Sichuan Agricultural University, Chengdu, China
- Center for Infectious Diseases Control (CIDC), Sichuan Agricultural University, Chengdu, China
| | - Kai Deng
- Center for Infectious Diseases Control (CIDC), Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Xiuhan Yang
- Center for Sustainable Antimicrobials, Department of Pharmacy, Sichuan Agricultural University, Chengdu, China
- Center for Infectious Diseases Control (CIDC), Sichuan Agricultural University, Chengdu, China
| | - Xu Song
- Center for Sustainable Antimicrobials, Department of Pharmacy, Sichuan Agricultural University, Chengdu, China
- Center for Infectious Diseases Control (CIDC), Sichuan Agricultural University, Chengdu, China
| | - Yuanfeng Zou
- Center for Sustainable Antimicrobials, Department of Pharmacy, Sichuan Agricultural University, Chengdu, China
- Center for Infectious Diseases Control (CIDC), Sichuan Agricultural University, Chengdu, China
| | - Xun Zhou
- Center for Sustainable Antimicrobials, Department of Pharmacy, Sichuan Agricultural University, Chengdu, China
- Center for Infectious Diseases Control (CIDC), Sichuan Agricultural University, Chengdu, China
| | - Huaqiao Tang
- Center for Sustainable Antimicrobials, Department of Pharmacy, Sichuan Agricultural University, Chengdu, China
- Center for Infectious Diseases Control (CIDC), Sichuan Agricultural University, Chengdu, China
| | - Lixia Li
- Center for Sustainable Antimicrobials, Department of Pharmacy, Sichuan Agricultural University, Chengdu, China
- Center for Infectious Diseases Control (CIDC), Sichuan Agricultural University, Chengdu, China
| | - Yuping Fu
- Center for Sustainable Antimicrobials, Department of Pharmacy, Sichuan Agricultural University, Chengdu, China
- Center for Infectious Diseases Control (CIDC), Sichuan Agricultural University, Chengdu, China
| | - Zhongqiong Yin
- Center for Sustainable Antimicrobials, Department of Pharmacy, Sichuan Agricultural University, Chengdu, China
- Center for Infectious Diseases Control (CIDC), Sichuan Agricultural University, Chengdu, China
| | - Hongping Wan
- Center for Infectious Diseases Control (CIDC), Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Xinghong Zhao
- Center for Sustainable Antimicrobials, Department of Pharmacy, Sichuan Agricultural University, Chengdu, China
- Center for Infectious Diseases Control (CIDC), Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
19
|
Guo L, Wambui J, Wang C, Muchaamba F, Fernandez-Cantos MV, Broos J, Tasara T, Kuipers OP, Stephan R. Cesin, a short natural variant of nisin, displays potent antimicrobial activity against major pathogens despite lacking two C-terminal macrocycles. Microbiol Spectr 2023; 11:e0531922. [PMID: 37754751 PMCID: PMC10581189 DOI: 10.1128/spectrum.05319-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 08/06/2023] [Indexed: 09/28/2023] Open
Abstract
Nisin is a widely used lantibiotic owing to its potent antimicrobial activity and its food-grade status. Its mode of action includes cell wall synthesis inhibition and pore formation, which are attributed to the lipid II binding and pore-forming domains, respectively. We discovered cesin, a short natural variant of nisin, produced by the psychrophilic anaerobe Clostridium estertheticum. Unlike other natural nisin variants, cesin lacks the two terminal macrocycles constituting the pore-forming domain. The current study aimed at heterologous expression and characterization of the antimicrobial activity and physicochemical properties of cesin. Following the successful heterologous expression of cesin in Lactococcus lactis, the lantibiotic demonstrated a broad and potent antimicrobial profile comparable to that of nisin. Determination of its mode of action using lipid II and lipoteichoic acid binding assays linked the potent antimicrobial activity to lipid II binding and electrostatic interactions with teichoic acids. Fluorescence microscopy showed that cesin lacks pore-forming ability in its natural form. Stability tests have shown the lantibiotic is highly stable at different pH values and temperature conditions, but that it can be degraded by trypsin. However, a bioengineered analog, cesin R15G, overcame the trypsin degradation, while keeping full antimicrobial activity. This study shows that cesin is a novel (small) nisin variant that efficiently kills target bacteria by inhibiting cell wall synthesis without pore formation. IMPORTANCE The current increase in antibiotic-resistant pathogens necessitates the discovery and application of novel antimicrobials. In this regard, we recently discovered cesin, which is a short natural variant of nisin produced by the psychrophilic Clostridium estertheticum. However, its suitability as an antimicrobial compound was in doubt due to its structural resemblance to nisin(1-22), a bioengineered short variant of nisin with low antimicrobial activity. Here, we show by heterologous expression, purification, and characterization that the potency of cesin is not only much higher than that of nisin(1-22), but that it is even comparable to the full-length nisin, despite lacking two C-terminal rings that are essential for nisin's activity. We show that cesin is a suitable scaffold for bioengineering to improve its applicability, such as resistance to trypsin. This study demonstrates the suitability of cesin for future application in food and/or for health as a potent and stable antimicrobial compound.
Collapse
Affiliation(s)
- Longcheng Guo
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, the Netherlands
| | - Joseph Wambui
- Institute for Food Safety and Hygiene, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Chenhui Wang
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, the Netherlands
| | - Francis Muchaamba
- Institute for Food Safety and Hygiene, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Maria Victoria Fernandez-Cantos
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, the Netherlands
| | - Jaap Broos
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, the Netherlands
| | - Taurai Tasara
- Institute for Food Safety and Hygiene, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Oscar P. Kuipers
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, the Netherlands
| | - Roger Stephan
- Institute for Food Safety and Hygiene, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| |
Collapse
|
20
|
Abd El-Aal AAA, Jayakumar FA, Lahiri C, Tan KO, Reginald K. Novel cationic cryptides in Penaeus vannamei demonstrate antimicrobial and anti-cancer activities. Sci Rep 2023; 13:14673. [PMID: 37673929 PMCID: PMC10482825 DOI: 10.1038/s41598-023-41581-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 08/29/2023] [Indexed: 09/08/2023] Open
Abstract
Cryptides are a subfamily of bioactive peptides that exist in all living organisms. They are latently encrypted in their parent sequences and exhibit a wide range of biological activities when decrypted via in vivo or in vitro proteases. Cationic cryptides tend to be drawn to the negatively charged membranes of microbial and cancer cells, causing cell death through various mechanisms. This makes them promising candidates for alternative antimicrobial and anti-cancer therapies, as their mechanism of action is independent of gene mutations. In the current study, we employed an in silico approach to identify novel cationic cryptides with potential antimicrobial and anti-cancer activities in atypical and systematic strategy by reanalysis of a publicly available RNA-seq dataset of Pacific white shrimp (Penaus vannamei) in response to bacterial infection. Out of 12 cryptides identified, five were selected based on their net charges and potential for cell penetration. Following chemical synthesis, the cryptides were assayed in vitro to test for their biological activities. All five cryptides demonstrated a wide range of selective activity against the tested microbial and cancer cells, their anti-biofilm activities against mature biofilms, and their ability to interact with Gram-positive and negative bacterial membranes. Our research provides a framework for a comprehensive analysis of transcriptomes in various organisms to uncover novel bioactive cationic cryptides. This represents a significant step forward in combating the crisis of multi-drug-resistant microbial and cancer cells, as these cryptides neither induce mutations nor are influenced by mutations in the cells they target.
Collapse
Affiliation(s)
- Amr Adel Ahmed Abd El-Aal
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, 47500, Bandar Sunway, Selangor, Malaysia
- Marine Microbiology Lab., National Institute of Oceanography and Fisheries (NIOF), Alexandria, 84511, Egypt
| | - Fairen Angelin Jayakumar
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, 47500, Bandar Sunway, Selangor, Malaysia
| | - Chandrajit Lahiri
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, 47500, Bandar Sunway, Selangor, Malaysia
- Department of Biotechnology, Atmiya University, Rajkot, Gujarat, 360005, India
| | - Kuan Onn Tan
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, 47500, Bandar Sunway, Selangor, Malaysia
| | - Kavita Reginald
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, 47500, Bandar Sunway, Selangor, Malaysia.
| |
Collapse
|
21
|
Kim H, Lee Y, Hwang YJ, Lee MH, Balaraju K, Jeon Y. Identification and characterization of Brevibacillus halotolerans B-4359: a potential antagonistic bacterium against red pepper anthracnose in Korea. Front Microbiol 2023; 14:1200023. [PMID: 37405162 PMCID: PMC10315534 DOI: 10.3389/fmicb.2023.1200023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 05/31/2023] [Indexed: 07/06/2023] Open
Abstract
Our study aimed to identify potential biocontrol agents (BCAs) against major phytopathogens under in vitro conditions by screening the Freshwater Bioresources Culture Collection (FBCC), Korea. Of the identified 856 strains, only 65 exhibited antagonistic activity, among which only one representative isolation, Brevibacillus halotolerans B-4359 was selected based on its in vitro antagonistic activity and enzyme production. Cell-free culture filtrate (CF) and volatile organic compounds (VOCs) of B-4359 were shown to be effective against the mycelial growth of Colletotrichum acutatum. Notably, B-4359 was found to promote spore germination in C. acutatum instead of exhibiting a suppressive effect when the bacterial suspension was mixed with the spore suspension of C. acutatum. However, B-4359 showed an excellent biological control effect on the anthracnose of red pepper fruits. Compared to other treatments and untreated control, B-4359 played a more effective role in controlling anthracnose disease under field conditions. The strain was identified as B. halotolerans using BIOLOG and 16S rDNA sequencing analyses. The genetic mechanism underlying the biocontrol traits of B-4359 was characterized using the whole-genome sequence of B-4359, which was closely compared with related strains. The whole-genome sequence of B-4359 consisted of 5,761,776 bp with a GC content of 41.0%, including 5,118 coding sequences, 117 tRNA, and 36 rRNA genes. The genomic analysis identified 23 putative secondary metabolite biosynthetic gene clusters. Our results provide a deep understanding of B-4359 as an effective biocontrol agent against red pepper anthracnose for sustainable agriculture.
Collapse
Affiliation(s)
- Heejin Kim
- Department of Plant Medicals, Andong National University, Andong, Republic of Korea
| | - Younmi Lee
- Department of Plant Medicals, Andong National University, Andong, Republic of Korea
| | - Ye-Ji Hwang
- Microbiology Research Department, Nakdonggang National Institute of Biological Resources, Sangju, Republic of Korea
| | - Mi-Hwa Lee
- Microbiology Research Department, Nakdonggang National Institute of Biological Resources, Sangju, Republic of Korea
| | - Kotnala Balaraju
- Agricultural Science and Technology Research Institute, Andong National University, Andong, Republic of Korea
| | - Yongho Jeon
- Department of Plant Medicals, Andong National University, Andong, Republic of Korea
| |
Collapse
|
22
|
Xu D, Zhang Z, Yao L, Wu L, Zhu Y, Zhao M, Xu H. Advances in the adenylation domain: discovery of diverse non-ribosomal peptides. Appl Microbiol Biotechnol 2023:10.1007/s00253-023-12585-2. [PMID: 37233756 DOI: 10.1007/s00253-023-12585-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/05/2023] [Accepted: 05/10/2023] [Indexed: 05/27/2023]
Abstract
Non-ribosomal peptide synthetases are mega-enzyme assembly lines that synthesize many clinically useful compounds. As a gatekeeper, they have an adenylation (A)-domain that controls substrate specificity and plays an important role in product structural diversity. This review summarizes the natural distribution, catalytic mechanism, substrate prediction methods, and in vitro biochemical analysis of the A-domain. Taking genome mining of polyamino acid synthetases as an example, we introduce research on mining non-ribosomal peptides based on A-domains. We discuss how non-ribosomal peptide synthetases can be engineered based on the A-domain to obtain novel non-ribosomal peptides. This work provides guidance for screening non-ribosomal peptide-producing strains, offers a method to discover and identify A-domain functions, and will accelerate the engineering and genome mining of non-ribosomal peptide synthetases. KEY POINTS: • Introducing adenylation domain structure, substrate prediction, and biochemical analysis methods • Advances in mining homo polyamino acids based on adenylation domain analysis • Creating new non-ribosomal peptides by engineering adenylation domains.
Collapse
Affiliation(s)
- Delei Xu
- College of Biological and Food Engineering, Changshu Institute of Technology, Changshu, 215500, China.
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing, 211816, China.
- Nanjing Xuankai Biotechnology Co., Ltd, Nanjing, 210000, China.
| | - Zihan Zhang
- College of Biological and Food Engineering, Changshu Institute of Technology, Changshu, 215500, China
| | - Luye Yao
- College of Biological and Food Engineering, Changshu Institute of Technology, Changshu, 215500, China
| | - LingTian Wu
- College of Biological and Food Engineering, Changshu Institute of Technology, Changshu, 215500, China
| | - Yibo Zhu
- College of Biological and Food Engineering, Changshu Institute of Technology, Changshu, 215500, China
| | - Meilin Zhao
- College of Biological and Food Engineering, Changshu Institute of Technology, Changshu, 215500, China
| | - Hong Xu
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing, 211816, China
| |
Collapse
|
23
|
Li L. Accessing hidden microbial biosynthetic potential from underexplored sources for novel drug discovery. Biotechnol Adv 2023:108176. [PMID: 37211187 DOI: 10.1016/j.biotechadv.2023.108176] [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: 11/14/2022] [Revised: 05/15/2023] [Accepted: 05/16/2023] [Indexed: 05/23/2023]
Abstract
Microbial natural products and their structural analogues have widely used as pharmaceutical agents, especially for infectious diseases and cancer. Despite this success, new structural classes with innovative chemistry and modes of action are urgently needed to be developed to combat the growing antimicrobial resistance and other public health problems. The advances in next-generation sequencing technologies and powerful computational tools open up new opportunities to explore microbial biosynthetic potential from underexplored sources, with millions of secondary metabolites awaiting discovery. The review highlights challenges associated with discovery of new chemical entities, rich reservoirs provided by untapped taxa, ecological niches or host microbiomes, emerging synthetic biotechnologies to unearth the hidden microbial biosynthetic potential for novel drug discovery at scale and speed.
Collapse
Affiliation(s)
- Lei Li
- State Key Laboratory of Microbial Metabolism and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Rd., Shanghai 200240, China.
| |
Collapse
|
24
|
Zhao X, Zhong X, Yang S, Deng K, Liu L, Song X, Zou Y, Li L, Zhou X, Jia R, Lin J, Tang H, Ye G, Yang J, Zhao S, Lang Y, Wan H, Yin Z, Kuipers OP. Elucidating the Mechanism of Action of the Gram-Negative-Pathogen-Selective Cyclic Antimicrobial Lipopeptide Brevicidine. Antimicrob Agents Chemother 2023; 67:e0001023. [PMID: 36912655 PMCID: PMC10190627 DOI: 10.1128/aac.00010-23] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 02/13/2023] [Indexed: 03/14/2023] Open
Abstract
Due to the accelerated appearance of antimicrobial-resistant (AMR) pathogens in clinical infections, new first-in-class antibiotics, operating via novel modes of action, are desperately needed. Brevicidine, a bacterial nonribosomally produced cyclic lipopeptide, has shown potent and selective antimicrobial activity against Gram-negative pathogens. However, before our investigations, little was known about how brevicidine exerts its potent bactericidal effect against Gram-negative pathogens. In this study, we find that brevicidine has potent antimicrobial activity against AMR Enterobacteriaceae pathogens, with MIC values ranging between 0.5 μM (0.8 mg/L) and 2 μM (3.0 mg/L). In addition, brevicidine showed potent antibiofilm activity against the Enterobacteriaceae pathogens, with the same 100% inhibition and 100% eradication concentration of 4 μM (6.1 mg/L). Further mechanistic studies showed that brevicidine exerts its potent bactericidal activity by interacting with lipopolysaccharide in the outer membrane, targeting phosphatidylglycerol and cardiolipin in the inner membrane, and dissipating the proton motive force of bacteria. This results in metabolic perturbation, including the inhibition of ATP synthesis; the inhibition of the dehydrogenation of NADH; the accumulation of reactive oxygen species in bacteria; and the inhibition of protein synthesis. Finally, brevicidine showed a good therapeutic effect in a mouse peritonitis-sepsis model. Our findings pave the way for further research on the clinical applications of brevicidine to combat prevalent infections caused by AMR Gram-negative pathogens worldwide.
Collapse
Affiliation(s)
- Xinghong Zhao
- Lab for Sustainable Antimicrobials, Department of Pharmacy, Sichuan Agricultural University, Chengdu, China
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, The Netherlands
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Xinyi Zhong
- Lab for Sustainable Antimicrobials, Department of Pharmacy, Sichuan Agricultural University, Chengdu, China
| | - Shinong Yang
- Lab for Sustainable Antimicrobials, Department of Pharmacy, Sichuan Agricultural University, Chengdu, China
| | - Kai Deng
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Lu Liu
- Lab for Sustainable Antimicrobials, Department of Pharmacy, Sichuan Agricultural University, Chengdu, China
| | - Xu Song
- Lab for Sustainable Antimicrobials, Department of Pharmacy, Sichuan Agricultural University, Chengdu, China
| | - Yuanfeng Zou
- Lab for Sustainable Antimicrobials, Department of Pharmacy, Sichuan Agricultural University, Chengdu, China
| | - Lixia Li
- Lab for Sustainable Antimicrobials, Department of Pharmacy, Sichuan Agricultural University, Chengdu, China
| | - Xun Zhou
- Lab for Sustainable Antimicrobials, Department of Pharmacy, Sichuan Agricultural University, Chengdu, China
| | - Renyong Jia
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Juchun Lin
- Lab for Sustainable Antimicrobials, Department of Pharmacy, Sichuan Agricultural University, Chengdu, China
| | - Huaqiao Tang
- Lab for Sustainable Antimicrobials, Department of Pharmacy, Sichuan Agricultural University, Chengdu, China
| | - Gang Ye
- Lab for Sustainable Antimicrobials, Department of Pharmacy, Sichuan Agricultural University, Chengdu, China
| | - Jianqing Yang
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Sichuan Agricultural University at Wenjiang, Chengdu, Sichuan, China
| | - Shan Zhao
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Yifei Lang
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Hongping Wan
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Zhongqiong Yin
- Lab for Sustainable Antimicrobials, Department of Pharmacy, Sichuan Agricultural University, Chengdu, China
| | - Oscar P. Kuipers
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
25
|
Ballantine RD, Al Ayed K, Bann SJ, Hoekstra M, Martin NI, Cochrane SA. Linearization of the Brevicidine and Laterocidine Lipopeptides Yields Analogues That Retain Full Antibacterial Activity. J Med Chem 2023; 66:6002-6009. [PMID: 37071814 PMCID: PMC10150354 DOI: 10.1021/acs.jmedchem.3c00308] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2023]
Abstract
Brevicidine and laterocidine are macrocyclic lipodepsipeptides with selective activity against Gram-negative bacteria, including colistin-resistant strains. Previously, the macrocyclic core of these peptides was thought essential for antibacterial activity. In this study, we show that C-terminal amidation of linear brevicidine and laterocidine scaffolds, and substitution of the native Thr9, yields linear analogues that retain the potent antibacterial activity and low hemolysis of the parent compounds. Furthermore, an alanine scan of both peptides revealed that the aromatic and basic amino acids within the common central scaffold are essential for antibacterial activity. This linearization strategy for modification of cyclic peptides is a highly effective way to reduce the time and cost of peptide synthesis and may be applicable to other non-ribosomal antibacterial peptides.
Collapse
Affiliation(s)
- Ross D Ballantine
- School of Chemistry and Chemical Engineering, Queen's University Belfast, Stranmillis Road, Belfast BT9 5AG, U.K
| | - Karol Al Ayed
- Biological Chemistry Group, Institute of Biology, Leiden University, Sylviusweg 72, Leiden 2333 BE, The Netherlands
| | - Samantha J Bann
- School of Chemistry and Chemical Engineering, Queen's University Belfast, Stranmillis Road, Belfast BT9 5AG, U.K
| | - Michael Hoekstra
- Biological Chemistry Group, Institute of Biology, Leiden University, Sylviusweg 72, Leiden 2333 BE, The Netherlands
| | - Nathaniel I Martin
- Biological Chemistry Group, Institute of Biology, Leiden University, Sylviusweg 72, Leiden 2333 BE, The Netherlands
| | - Stephen A Cochrane
- School of Chemistry and Chemical Engineering, Queen's University Belfast, Stranmillis Road, Belfast BT9 5AG, U.K
| |
Collapse
|
26
|
Jonkergouw C, Beyeh NK, Osmekhina E, Leskinen K, Taimoory SM, Fedorov D, Anaya-Plaza E, Kostiainen MA, Trant JF, Ras RHA, Saavalainen P, Linder MB. Repurposing host-guest chemistry to sequester virulence and eradicate biofilms in multidrug resistant Pseudomonas aeruginosa and Acinetobacter baumannii. Nat Commun 2023; 14:2141. [PMID: 37059703 PMCID: PMC10104825 DOI: 10.1038/s41467-023-37749-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 03/29/2023] [Indexed: 04/16/2023] Open
Abstract
The limited diversity in targets of available antibiotic therapies has put tremendous pressure on the treatment of bacterial pathogens, where numerous resistance mechanisms that counteract their function are becoming increasingly prevalent. Here, we utilize an unconventional anti-virulence screen of host-guest interacting macrocycles, and identify a water-soluble synthetic macrocycle, Pillar[5]arene, that is non-bactericidal/bacteriostatic and has a mechanism of action that involves binding to both homoserine lactones and lipopolysaccharides, key virulence factors in Gram-negative pathogens. Pillar[5]arene is active against Top Priority carbapenem- and third/fourth-generation cephalosporin-resistant Pseudomonas aeruginosa and Acinetobacter baumannii, suppressing toxins and biofilms and increasing the penetration and efficacy of standard-of-care antibiotics in combined administrations. The binding of homoserine lactones and lipopolysaccharides also sequesters their direct effects as toxins on eukaryotic membranes, neutralizing key tools that promote bacterial colonization and impede immune defenses, both in vitro and in vivo. Pillar[5]arene evades both existing antibiotic resistance mechanisms, as well as the build-up of rapid tolerance/resistance. The versatility of macrocyclic host-guest chemistry provides ample strategies for tailored targeting of virulence in a wide range of Gram-negative infectious diseases.
Collapse
Affiliation(s)
- Christopher Jonkergouw
- Aalto University, School of Chemical Engineering, Department of Bioproducts and Biosystems, Kemistintie 1, 02150, Espoo, Finland.
| | - Ngong Kodiah Beyeh
- Oakland University, Department of Chemistry, 146 Library Drive, Rochester, MI, 48309-4479, USA
- Aalto University, School of Science, Department of Applied Physics, Puumiehenkuja 2, Espoo, Finland
| | - Ekaterina Osmekhina
- Aalto University, School of Chemical Engineering, Department of Bioproducts and Biosystems, Kemistintie 1, 02150, Espoo, Finland
| | - Katarzyna Leskinen
- University of Helsinki, Translational Immunology Research Program, Haartmaninkatu 8, 0014, Helsinki, Finland
| | - S Maryamdokht Taimoory
- University of Windsor, Department of Chemistry and Biochemistry, Windsor, ON, N9B 3P4, Canada
- University of Michigan, Department of Chemistry, Ann Arbor, MI, USA
| | - Dmitrii Fedorov
- Aalto University, School of Chemical Engineering, Department of Bioproducts and Biosystems, Kemistintie 1, 02150, Espoo, Finland
| | - Eduardo Anaya-Plaza
- Aalto University, School of Chemical Engineering, Department of Bioproducts and Biosystems, Kemistintie 1, 02150, Espoo, Finland
| | - Mauri A Kostiainen
- Aalto University, School of Chemical Engineering, Department of Bioproducts and Biosystems, Kemistintie 1, 02150, Espoo, Finland
| | - John F Trant
- University of Windsor, Department of Chemistry and Biochemistry, Windsor, ON, N9B 3P4, Canada
| | - Robin H A Ras
- Aalto University, School of Chemical Engineering, Department of Bioproducts and Biosystems, Kemistintie 1, 02150, Espoo, Finland
- Aalto University, School of Science, Department of Applied Physics, Puumiehenkuja 2, Espoo, Finland
| | - Päivi Saavalainen
- University of Helsinki, Translational Immunology Research Program, Haartmaninkatu 8, 0014, Helsinki, Finland.
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland.
| | - Markus B Linder
- Aalto University, School of Chemical Engineering, Department of Bioproducts and Biosystems, Kemistintie 1, 02150, Espoo, Finland.
| |
Collapse
|
27
|
Han P, Ma A, Ning Y, Chen Z, Liu Y, Liu Z, Li S, Jia Y. Global gene-mining strategy for searching nonribosomal peptides as antimicrobial agents from microbial sources. Lebensm Wiss Technol 2023. [DOI: 10.1016/j.lwt.2023.114708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2023]
|
28
|
Al Ayed K, Zamarbide Losada D, Machushynets NV, Terlouw B, Elsayed SS, Schill J, Trebosc V, Pieren M, Medema MH, van Wezel GP, Martin NI. Total Synthesis and Structure Assignment of the Relacidine Lipopeptide Antibiotics and Preparation of Analogues with Enhanced Stability. ACS Infect Dis 2023; 9:739-748. [PMID: 37000899 PMCID: PMC10111413 DOI: 10.1021/acsinfecdis.3c00043] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
Abstract
The unabated rise of antibiotic resistance has raised the specter of a post-antibiotic era and underscored the importance of developing new classes of antibiotics. The relacidines are a recently discovered group of nonribosomal lipopeptide antibiotics that show promising activity against Gram-negative pathogens and share structural similarities with brevicidine and laterocidine. While the first reports of the relacidines indicated that they possess a C-terminal five-amino acid macrolactone, an N-terminal lipid tail, and an overall positive charge, no stereochemical configuration was assigned, thereby precluding a full structure determination. To address this issue, we here report a bioinformatics guided total synthesis of relacidine A and B and show that the authentic natural products match our predicted and synthesized structures. Following on this, we also synthesized an analogue of relacidine A wherein the ester linkage of the macrolactone was replaced by the corresponding amide. This analogue was found to possess enhanced hydrolytic stability while maintaining the antibacterial activity of the natural product in both in vitro and in vivo efficacy studies.
Collapse
Affiliation(s)
- Karol Al Ayed
- Biological Chemistry Group, Institute of Biology, Leiden University, Sylviusweg 72, 2333 BE Leiden, Netherlands
| | - Denise Zamarbide Losada
- Biological Chemistry Group, Institute of Biology, Leiden University, Sylviusweg 72, 2333 BE Leiden, Netherlands
| | - Nataliia V. Machushynets
- Molecular Biotechnology Group, Institute of Biology, Leiden University, Sylviusweg 72, 2333 BE Leiden, Netherlands
| | - Barbara Terlouw
- Bioinformatics Group, Wageningen University, Droevendaalsesteeg 1, 6708 PB Wageningen, Netherlands
| | - Somayah S. Elsayed
- Molecular Biotechnology Group, Institute of Biology, Leiden University, Sylviusweg 72, 2333 BE Leiden, Netherlands
| | - Julian Schill
- BioVersys AG, c/o Technologiepark, Hochbergerstrasse 60c, CH-4057 Basel, Switzerland
| | - Vincent Trebosc
- BioVersys AG, c/o Technologiepark, Hochbergerstrasse 60c, CH-4057 Basel, Switzerland
| | - Michel Pieren
- BioVersys AG, c/o Technologiepark, Hochbergerstrasse 60c, CH-4057 Basel, Switzerland
| | - Marnix H. Medema
- Bioinformatics Group, Wageningen University, Droevendaalsesteeg 1, 6708 PB Wageningen, Netherlands
| | - Gilles P. van Wezel
- Molecular Biotechnology Group, Institute of Biology, Leiden University, Sylviusweg 72, 2333 BE Leiden, Netherlands
| | - Nathaniel I. Martin
- Biological Chemistry Group, Institute of Biology, Leiden University, Sylviusweg 72, 2333 BE Leiden, Netherlands
| |
Collapse
|
29
|
Ballantine RD, Al Ayed K, Bann SJ, Hoekstra M, Martin NI, Cochrane SA. Synthesis and structure-activity relationship studies of N-terminal analogues of the lipopeptide antibiotics brevicidine and laterocidine. RSC Med Chem 2022; 13:1640-1643. [PMID: 36545437 PMCID: PMC9749936 DOI: 10.1039/d2md00281g] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 10/26/2022] [Indexed: 11/06/2022] Open
Abstract
The brevicidine and laterocidine family of lipopeptide antibiotics exhibit strong activity against multidrug-resistant Gram-negative bacteria, while showing low propensity to induce resistance. Both peptides feature a branched lipid tail on the N-terminal residue, which for brevicidine is chiral. Here, we report the synthesis and biological evaluation of a library of brevicidine and laterocidine analogues wherein the N-terminal lipid is replaced with linear achiral fatty acids. Optimal lipid chain lengths were determined and new analogues with strong activity against colistin-resistant E. coli produced.
Collapse
Affiliation(s)
- Ross D. Ballantine
- School of Chemistry and Chemical Engineering, David Keir Building, Queen's University BelfastStranmillis RoadBelfastBT9 5AGUK
| | - Karol Al Ayed
- Biological Chemistry Group, Institute of Biology, Leiden UniversitySylviusweg 722333 BELeidenThe Netherlands
| | - Samantha J. Bann
- School of Chemistry and Chemical Engineering, David Keir Building, Queen's University BelfastStranmillis RoadBelfastBT9 5AGUK
| | - Michael Hoekstra
- Biological Chemistry Group, Institute of Biology, Leiden UniversitySylviusweg 722333 BELeidenThe Netherlands
| | - Nathaniel I. Martin
- Biological Chemistry Group, Institute of Biology, Leiden UniversitySylviusweg 722333 BELeidenThe Netherlands
| | - Stephen A. Cochrane
- School of Chemistry and Chemical Engineering, David Keir Building, Queen's University BelfastStranmillis RoadBelfastBT9 5AGUK
| |
Collapse
|
30
|
Tian P, Ye W, Zhang X, Tong Y, Qian PY, Tong R. Ten-step asymmetric total syntheses of potent antibiotics anthracimycin and anthracimycin B. Chem Sci 2022; 13:12776-12781. [PMID: 36519065 PMCID: PMC9645392 DOI: 10.1039/d2sc05049h] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 10/14/2022] [Indexed: 10/06/2023] Open
Abstract
The increase in antibiotic resistance calls for the development of novel antibiotics with new molecular structures and new modes of action. However, in the past few decades only a few novel antibiotics have been discovered and progressed into clinically used drugs. The discovery of a potent anthracimycin antibiotic represents a major advance in the field of antibiotics. Anthracimycin is a structurally novel macrolide natural product with an excellent biological activity profile: (i) potent in vitro antibacterial activity (MIC 0.03-1.0 μg mL-1) against many methicillin-resistant Staphylococcus aureus (MRSA) strains, Bacillus anthracis (anthrax), and Mycobacterium tuberculosis; (ii) low toxicity to human cells (IC50 > 30 μM); (iii) a novel mechanism of action (inhibiting DNA/RNA synthesis). While the first total synthesis of anthracimycin was elegantly accomplished by Brimble et al. with 20 steps, we report a 10-step asymmetric total synthesis of anthracimycin and anthracimycin B (first total synthesis). Our convergent strategy features (i) one-pot sequential Mukaiyama vinylogous aldol/intramolecular Diels-Alder reaction to construct trans-decalin with high yield and excellent endo/exo selectivity and (ii) Z-selective ring-closing metathesis to forge the 14-membered ring. In vitro antibacterial evaluation suggested that our synthetic samples exhibited similar antibacterial potency to the naturally occurring anthracimycins against Gram-positive strains. Our short and reliable synthetic route provides a supply of anthracimycins for further in-depth studies and allows medicinal chemists to prepare a library of analogues for establishing structure-activity relationships.
Collapse
Affiliation(s)
- Peilin Tian
- Department of Chemistry, The Hong Kong University of Science and Technology Clearwater Bay Kowloon Hong Kong China +86 23581594 +86 23587357
| | - Wenkang Ye
- Department of Ocean Science, The Hong Kong University of Science and Technology Clear Water Bay Kowloon Hong Kong China
| | - Xiayan Zhang
- Department of Chemistry, The Hong Kong University of Science and Technology Clearwater Bay Kowloon Hong Kong China +86 23581594 +86 23587357
| | - Yi Tong
- Department of Chemistry, The Hong Kong University of Science and Technology Clearwater Bay Kowloon Hong Kong China +86 23581594 +86 23587357
| | - Pei-Yuan Qian
- Department of Ocean Science, The Hong Kong University of Science and Technology Clear Water Bay Kowloon Hong Kong China
- The Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou) Nansha Guangzhou 511458 China
| | - Rongbiao Tong
- Department of Chemistry, The Hong Kong University of Science and Technology Clearwater Bay Kowloon Hong Kong China +86 23581594 +86 23587357
- The Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou) Nansha Guangzhou 511458 China
| |
Collapse
|
31
|
Xing R, Song X, Liu L, Wang Y, Zhang Y, Peng S, Jia R, Zhao X, Zou Y, Li L, Wan H, Zhou X, Shi F, Ye G, Yin Z. Quaternized chitosan-coated liposomes enhance immune responses by co-delivery of antigens and resveratrol. Int J Pharm 2022; 628:122277. [DOI: 10.1016/j.ijpharm.2022.122277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 10/04/2022] [Accepted: 10/06/2022] [Indexed: 10/31/2022]
|
32
|
C-terminal modification of a de novo designed antimicrobial peptide via capping of macrolactam rings. Bioorg Chem 2022; 130:106251. [DOI: 10.1016/j.bioorg.2022.106251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 10/20/2022] [Accepted: 11/01/2022] [Indexed: 11/06/2022]
|
33
|
Le Han H, Jiang L, Thu Tran TN, Muhammad N, Kim SG, Tran Pham VP, Ng YJ, Khoo KS, Chew KW, Phuong Nguyen TD. Whole-genome analysis and secondary metabolites production of a new strain Brevibacillus halotolerans 7WMA2: A potential biocontrol agent against fungal pathogens. CHEMOSPHERE 2022; 307:136004. [PMID: 35970213 DOI: 10.1016/j.chemosphere.2022.136004] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 07/05/2022] [Accepted: 08/07/2022] [Indexed: 06/15/2023]
Abstract
The extensive usage of synthetic fungicides against fungal diseases has caused adverse impacts on both human and agricultural crops. Therefore, the current study aims to establish a new bacterium 7WMA2, as a biocontrol agent to achieve better antifungal results. The strain 7WMA2 was isolated from marine sediment, displayed a broad spectrum of several fungi that includes Alternaria alternata, Cladosporium sp., Candida albicans, Fusarium oxysporum, Trichosporon pullulans, and Trichophyton rubrum. The 16S rRNA phylogeny inferred that strain 7WMA2 was a member of Brevibacillus. The phylogenetic and biochemical analyses revealed that the strain 7WMA2 belongs to the species of Brevibacillus halotolerans. The complete genome sequence of Brevibacillus halotolerans 7WMA2 consists of a circular chromosome of 5,351,077 bp length with a GC content of 41.39 mol %, including 4433 CDS, 111 tRNA genes, and 36 rRNA genes. The genomic analysis showed 23 putative biosynthetic secondary metabolite gene clusters responsible for non-ribosomal peptides, polyketides and siderophores. The antifungal compounds concentrated from cell-free fermentation broth demonstrated strong inhibition of fungi, and the compounds are considerably thermal stable and adaptable to pH range 2-12. This complete genome sequence has provided insight for further exploration of antagonistic ability and its secondary metabolite compounds indicated feasibility as biological control agents against fungal infections.
Collapse
Affiliation(s)
- Ho Le Han
- Biological Resource Center/Korean Collection for Type Cultures (KCTC), Korea Research Institute of Bioscience and Biotechnology, Jeongeup, 56212, Republic of Korea; Scientific Management Department, Dong A University, Da Nang City, Viet Nam
| | - Lingmin Jiang
- Biological Resource Center/Korean Collection for Type Cultures (KCTC), Korea Research Institute of Bioscience and Biotechnology, Jeongeup, 56212, Republic of Korea
| | - Thi Ngoc Thu Tran
- The University of Danang, University of Technology and Education, Danang City 550000, Viet Nam
| | - Neak Muhammad
- Biological Resource Center/Korean Collection for Type Cultures (KCTC), Korea Research Institute of Bioscience and Biotechnology, Jeongeup, 56212, Republic of Korea; University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
| | - Song-Gun Kim
- Biological Resource Center/Korean Collection for Type Cultures (KCTC), Korea Research Institute of Bioscience and Biotechnology, Jeongeup, 56212, Republic of Korea; University of Science and Technology (UST), Daejeon, 34113, Republic of Korea.
| | | | - Yan Jer Ng
- Department of Chemical and Environmental Engineering, Faculty of Science and Engineering, University of Nottingham Malaysia, Jalan Broga, 43500, Semenyih, Selangor Darul Ehsan, Malaysia
| | - Kuan Shiong Khoo
- Faculty of Applied Sciences, UCSI University, UCSI Heights, 56000 Cheras, Kuala Lumpur, Malaysia
| | - Kit Wayne Chew
- School of Energy and Chemical Engineering, Xiamen University Malaysia, Jalan Sunsuria, Bandar Sunsuria, 43900, Sepang, Selangor Darul Ehsan, Malaysia.
| | - Thi Dong Phuong Nguyen
- The University of Danang, University of Technology and Education, Danang City 550000, Viet Nam.
| |
Collapse
|
34
|
Jiang M, Chen R, Zhang J, Chen F, Wang KJ. A Novel Antimicrobial Peptide Spampcin 56-86 from Scylla paramamosain Exerting Rapid Bactericidal and Anti-Biofilm Activity In Vitro and Anti-Infection In Vivo. Int J Mol Sci 2022; 23:ijms232113316. [PMID: 36362111 PMCID: PMC9653689 DOI: 10.3390/ijms232113316] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/27/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022] Open
Abstract
The abuse of antibiotics leads to the increase of bacterial resistance, which seriously threatens human health. Therefore, there is an urgent need to find effective alternatives to antibiotics, and antimicrobial peptides (AMPs) are the most promising antibacterial agents and have received extensive attention. In this study, a novel potential AMP was identified from the marine invertebrate Scylla paramamosain and named Spampcin. After bioinformatics analysis and AMP database prediction, four truncated peptides (Spa31, Spa22, Spa20 and Spa14) derived from Spampcin were screened, all of which showed potent antimicrobial activity with different antibacterial spectrum. Among them, Spampcin56-86 (Spa31 for short) exhibited strong bactericidal activity against a variety of clinical pathogens and could rapidly kill the tested bacteria within minutes. Further analysis of the antibacterial mechanism revealed that Spa31 disrupted the integrity of the bacterial membrane (as confirmed by scanning electron microscopy observation, NPN, and PI staining assays), leading to bacterial rupture, leakage of cellular contents (such as elevated extracellular ATP), increased ROS production, and ultimately cell death. Furthermore, Spa31 was found to interact with LPS and effectively inhibit bacterial biofilms. The antibacterial activity of Spa31 had good thermal stability, certain ion tolerance, and no obvious cytotoxicity. It is worth noting that Spa31 could significantly improve the survival rate of zebrafish Danio rerio infected with Pseudomonas aeruginosa, indicating that Spa31 played an important role in anti-infection in vivo. This study will enrich the database of marine animal AMPs and provide theoretical reference and scientific basis for the application of marine AMPs in medical fields.
Collapse
Affiliation(s)
- Manyu Jiang
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Sciences, Xiamen University, Xiamen 361102, China
| | - Roushi Chen
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Sciences, Xiamen University, Xiamen 361102, China
| | - Jingrong Zhang
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Sciences, Xiamen University, Xiamen 361102, China
| | - Fangyi Chen
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Sciences, Xiamen University, Xiamen 361102, China
- State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, College of Ocean & Earth Sciences, Xiamen University, Xiamen 361102, China
- Fujian Innovation Research Institute for Marine Biological Antimicrobial Peptide Industrial Technology, College of Ocean & Earth Sciences, Xiamen University, Xiamen 361102, China
- Correspondence: (F.C.); (K.-J.W.)
| | - Ke-Jian Wang
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Sciences, Xiamen University, Xiamen 361102, China
- State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, College of Ocean & Earth Sciences, Xiamen University, Xiamen 361102, China
- Fujian Innovation Research Institute for Marine Biological Antimicrobial Peptide Industrial Technology, College of Ocean & Earth Sciences, Xiamen University, Xiamen 361102, China
- Correspondence: (F.C.); (K.-J.W.)
| |
Collapse
|
35
|
Clements-Decker T, Kode M, Khan S, Khan W. Underexplored bacteria as reservoirs of novel antimicrobial lipopeptides. Front Chem 2022; 10:1025979. [PMID: 36277345 PMCID: PMC9581180 DOI: 10.3389/fchem.2022.1025979] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 09/13/2022] [Indexed: 11/13/2022] Open
Abstract
Natural products derived from microorganisms play a prominent role in drug discovery as potential anti-infective agents. Over the past few decades, lipopeptides produced by particularly Bacillus, Pseudomonas, Streptomyces, Paenibacillus, and cyanobacteria species, have been extensively studied for their antimicrobial potential. Subsequently, daptomycin and polymyxin B were approved by the Food and Drug Administration as lipopeptide antibiotics. Recent studies have however, indicated that Serratia, Brevibacillus, and Burkholderia, as well as predatory bacteria such as Myxococcus, Lysobacter, and Cystobacter, hold promise as relatively underexplored sources of novel classes of lipopeptides. This review will thus highlight the structures and the newly discovered scaffolds of lipopeptide families produced by these bacterial genera, with potential antimicrobial activities. Additionally, insight into the mode of action and biosynthesis of these lipopeptides will be provided and the application of a genome mining approach, to ascertain the biosynthetic gene cluster potential of these bacterial genera (genomes available on the National Center for Biotechnology Information) for their future pharmaceutical exploitation, will be discussed.
Collapse
Affiliation(s)
| | - Megan Kode
- Department of Microbiology, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
| | - Sehaam Khan
- Faculty of Health Sciences, University of Johannesburg, Doornfontein, South Africa
| | - Wesaal Khan
- Department of Microbiology, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
- *Correspondence: Wesaal Khan,
| |
Collapse
|
36
|
Liu Y, Han P, Jia Y, Chen Z, Li S, Ma A. Antibacterial Regularity Mining Beneath the Systematic Activity Database of Lipopeptides Brevilaterins: An Instructive Activity Handbook for Its Food Application. Foods 2022; 11:foods11192991. [PMID: 36230066 PMCID: PMC9563055 DOI: 10.3390/foods11192991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/22/2022] [Accepted: 09/23/2022] [Indexed: 12/01/2022] Open
Abstract
Bacterial contamination is a primary threat to food safety. Therefore, the persistent development of natural antibacterial agents has become essential work. The present essay attempts to establish a systematic antibacterial activity database to instruct the food application of brevilaterins, promising antibacterial lipopeptides from Brevibacillus laterosporus S62-9. Minimum inhibitory concentrations (MIC) and minimum bactericidal concentrations (MBC) were systematically collected from 43 species of standard bacteria and 140 strains of isolated bacteria (food spoilage bacteria and antibiotic-resistant bacteria) using a broth dilution method. The results showed that brevilaterins performed a broad-spectrum inhibitory (0.5~128 μg/mL) and bactericidal activity (1~256 μg/mL), especially efficient against Gram-positive bacteria and spoilage bacteria from grain products. Moreover, brevilaterins not only inhibit and kill multiple antibiotic-resistant bacteria but do not readily develop resistance, with a small specific value of MBC/MIC (1~8). Furthermore, brevilaterins would interact with negatively charged sodium dodecyl sulfate and bind amphipathic soybean phospholipid with an affinity constant of KD = 4.70 × 10−4 M. No significant activity difference was found between brevilaterin B and brevilaterin C. Collectively, this work contributed rich antibacterial data of brevilaterins and revealed the antibacterial regularity beneath these data, which can be used as an activity handbook to instruct their application in food safety.
Collapse
|
37
|
Han P, Chen Z, Liu Y, Ma A, Li S, Jia Y. An accurate strategy for pointing the key biocatalytic sites of bre2691A protein for modification of the brevilaterin from Brevibacillus laterosporus. Microb Cell Fact 2022; 21:196. [PMID: 36123650 PMCID: PMC9484153 DOI: 10.1186/s12934-022-01918-x] [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: 06/08/2022] [Accepted: 09/05/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Brevilaterin A-E, a novel class of multi-component cationic antimicrobial lipopeptides, were biosynthesized by a non-ribosomal peptides synthetase (NRPS) in Brevibacillus laterosporus. However, the antimicrobial abilities of different brevilaterin components varied greatly, and this multi-component form was impeding the scale production of the excellent component, and a little information about the brevilaterin biosynthesis mechanism was available to apply in brevilaterin design modification. In this study, we used an accurate strategy that revealed the reason for producing multi-component was the substrate selectivity of bre2691A protein being not enough specific and pinpointed the key design sites to make the specificity of bre2691A enhanced. RESULTS Bioinformatic analysis revealed that the biocatalytic site of bre2691A, which was an adenylation domain catalyzed and recognized methionine, leucine, valine and isoleucine and thus introduced them into brevilaterins and caused different components (brevilaterin A-E), was consisted of A1 ~ A10 residues named specificity-conferring code. Coupling molecular docking simulations with mutation studies identified A2 and A7 as critical residues, where determined substrate-specificity and impacted activity. The in virto activity assay showed that the A2 mutant (G193A) would lose activity against methionine and have no effect on the other three amino acids, the A7 mutant (G285C) would enhance the catalytic activity against four substrates, especially against leucine at almost a double activity. When the A2 and A7 residues were synchronously mutated, this mutant would be more focused on recognizing leucine. CONCLUSIONS An accurate strategy that combined with bioinformatics and site-directed mutation techniques revealed the pivotal site A2 and A7 positions of bre2691A protein that could be used to design and modify brevilaterins, thus further providing a reasonable direction of genetic engineering for Brevibacillus laterosporus. A deeper understanding of the function of crucial residues in the adenylation domain would make it get more accurate and highly efficient design and more fully utilized. Furthermore, it would contribute to biotechnological applications, namely for the large centralized synthesis of antimicrobial peptides, or for the optimization of their production.
Collapse
Affiliation(s)
- Panpan Han
- School of Food and Health, Beijing Technology and Business University, No.33 Fucheng Road, Haidian District, Beijing, 100048, China
| | - Zhou Chen
- School of Food and Health, Beijing Technology and Business University, No.33 Fucheng Road, Haidian District, Beijing, 100048, China
| | - Yangliu Liu
- School of Food and Health, Beijing Technology and Business University, No.33 Fucheng Road, Haidian District, Beijing, 100048, China
| | - Aijin Ma
- School of Food and Health, Beijing Technology and Business University, No.33 Fucheng Road, Haidian District, Beijing, 100048, China
| | - Siting Li
- School of Food and Health, Beijing Technology and Business University, No.33 Fucheng Road, Haidian District, Beijing, 100048, China
| | - Yingmin Jia
- School of Food and Health, Beijing Technology and Business University, No.33 Fucheng Road, Haidian District, Beijing, 100048, China.
| |
Collapse
|
38
|
Mendivil-Perez M, Jimenez-Del-Rio M, Velez-Pardo C. Polycationic peptide R7-G-Aβ25-35 selectively induces cell death in leukemia Jurkat T cells through speedy mitochondrial depolarization, and CASPASE-3 -independent mechanism. Biochem Biophys Rep 2022; 31:101300. [PMID: 35755270 PMCID: PMC9214795 DOI: 10.1016/j.bbrep.2022.101300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/26/2022] [Accepted: 06/13/2022] [Indexed: 11/02/2022] Open
Abstract
Background Methods Results Conclusion Polycationic arginine (R) residue bound Aβ25-35 peptide is cytotoxic to Jurkat cells. R7-G-Aβ25-35 is more effective killing leukemia cells than Aβ25-35-G-R7. R7-G-Aβ25-35 induces alteration of cell metabolism, and reduces cell proliferation. R7-G-Aβ25-35 provokes loss of ΔΨm and produces high amount of ROS. R7-G-Aβ25-35 is harmless to normal proliferative mesenchymal stromal cells.
Collapse
|
39
|
Long L, Sulaiman JE, Xiao Y, Cheng A, Wang R, Malit JJ, Wong WC, Liu W, Li YX, Chen F, Lam H, Qian PY. Mode of action of elasnin as biofilm formation eradicator of methicillin-resistant Staphylococcus aureus. Front Microbiol 2022; 13:967845. [PMID: 36003935 PMCID: PMC9393526 DOI: 10.3389/fmicb.2022.967845] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 07/20/2022] [Indexed: 11/13/2022] Open
Abstract
Biofilm is made up of microbes and their extracellular matrix, making microorganisms highly tolerant, resistant, and resilient to a wide range of antimicrobials. Biofilm treatment with conventional antimicrobial agents can accelerate the evolution and spread of resistance due to the reduced efficacy and increased gene transfer and differentiation within biofilms. Therefore, effective biofilm-targeting compounds are currently highly sought after. In the present study, we identified elasnin as a potent biofilm-targeting compound against methicillin-resistant Staphylococcus aureus (MRSA). Elasnin effectively inhibited biofilm formation and especially eradicated the pre-formed biofilms of MRSA with low cytotoxicity and low risk of resistance development and retains its activity in a chronic wound biofilms model. A comprehensive mechanistic study using multi-omics and confocal and scanning electron microscopy revealed that elasnin induced the biofilm matrix destruction in a time-dependent manner and interfered with the cell division during the exponential phase, primarily by repressing the expression of virulence factors. Cells released from the elasnin-treated biofilms exhibited a defective appearance and became more sensitive to beta-lactam antibiotic penicillin G. Through gene overexpression and deletion assay, we discovered the key role of sarZ during elasnin-induced biofilm eradication. Overall, the present study identified elasnin as a potent biofilm eradicator against MRSA that harbors potential to be developed for biofilm removal and chronic wound treatment, and provided new insights into the molecular targets for biofilm eradication in MRSA.
Collapse
Affiliation(s)
- Lexin Long
- Department of Ocean Science, The Hong Kong University of Science and Technology, Kowloon, Hong Kong, China
- SZU-HKUST Joint PhD Program in Marine Environmental Science, Shenzhen University, Shenzhen, China
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, China
| | - Jordy Evan Sulaiman
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Kowloon, Hong Kong SAR, China
| | - Yao Xiao
- Department of Ocean Science, The Hong Kong University of Science and Technology, Kowloon, Hong Kong, China
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, China
| | - Aifang Cheng
- Department of Ocean Science, The Hong Kong University of Science and Technology, Kowloon, Hong Kong, China
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, China
| | - Ruojun Wang
- Department of Ocean Science, The Hong Kong University of Science and Technology, Kowloon, Hong Kong, China
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, China
| | - Jessie James Malit
- Department of Ocean Science, The Hong Kong University of Science and Technology, Kowloon, Hong Kong, China
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, China
| | - Wai Chuen Wong
- Department of Ocean Science, The Hong Kong University of Science and Technology, Kowloon, Hong Kong, China
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, China
| | - Wenchao Liu
- Department of Ocean Science, The Hong Kong University of Science and Technology, Kowloon, Hong Kong, China
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, China
| | - Yong-Xin Li
- Department of Chemistry, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- The Swire Institute of Marine Science and Hong Kong Branch of Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Feng Chen
- Institute for Advanced Study, Shenzhen University, Shenzhen, China
| | - Henry Lam
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Kowloon, Hong Kong SAR, China
- Henry Lam,
| | - Pei-Yuan Qian
- Department of Ocean Science, The Hong Kong University of Science and Technology, Kowloon, Hong Kong, China
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, China
- *Correspondence: Pei-Yuan Qian,
| |
Collapse
|
40
|
Zhao Y, Xie X, Li J, Shi Y, Chai A, Fan T, Li B, Li L. Comparative Genomics Insights into a Novel Biocontrol Agent Paenibacillus peoriae Strain ZF390 against Bacterial Soft Rot. BIOLOGY 2022; 11:1172. [PMID: 36009799 PMCID: PMC9404902 DOI: 10.3390/biology11081172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/27/2022] [Accepted: 08/03/2022] [Indexed: 11/16/2022]
Abstract
Bacterial soft rot, caused by Pectobacterium brasiliense, can infect several economically important horticultural crops. However, the management strategies available to control this disease are limited. Plant-growth-promoting rhizobacteria (PGPR) have been considered to be promising biocontrol agents. With the aim of obtaining a strain suitable for agricultural applications, 161 strains were isolated from the rhizosphere soil of healthy cucumber plants and screened through plate bioassays and greenhouse tests. Paenibacillus peoriae ZF390 exhibited an eminent control effect against soft rot disease and a broad antagonistic activity spectrum in vitro. Moreover, ZF390 showed good activities of cellulase, protease, and phosphatase and a tolerance of heavy metal. Whole-genome sequencing was performed and annotated to explore the underlying biocontrol mechanisms. Strain ZF390 consists of one 6,193,667 bp circular chromosome and three plasmids. Comparative genome analysis revealed that ZF390 involves ten gene clusters responsible for secondary metabolite antibiotic synthesis, matching its excellent biocontrol activity. Plenty of genes related to plant growth promotion, biofilm formation, and induced systemic resistance were mined to reveal the biocontrol mechanisms that might consist in strain ZF390. Overall, these findings suggest that strain ZF390 could be a potential biocontrol agent in bacterial-soft-rot management, as well as a source of antimicrobial mechanisms for further exploitation.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Baoju Li
- Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Lei Li
- Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| |
Collapse
|
41
|
Wu C, Tang J, Limlingan Malit JJ, Wang R, Sung HHY, Williams ID, Qian PY. Bathiapeptides: Polythiazole-Containing Peptides from a Marine Biofilm-Derived Bacillus sp. JOURNAL OF NATURAL PRODUCTS 2022; 85:1751-1762. [PMID: 35703501 DOI: 10.1021/acs.jnatprod.2c00290] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Bacteria in marine biofilms are a rich reservoir of natural products. To facilitate novel secondary metabolite discovery, we investigated the metabolic profile of a marine biofilm-derived Bacillus sp. B19-2 by combining bioinformatics and LC-UV-MS analyses. After dereplication and purification of putatively unknown compounds, a new family of compounds 1-8 was uncovered and named bathiapeptides. Structural elucidation using NMR, HRESIMS, ozonolysis, advanced Marfey's analysis, and X-ray diffraction revealed that bathiapeptides are polypeptides that contain a rare polythiazole moiety. These compounds exhibited strong cytotoxicity against Hep G2, HeLa, MCF-7, and MGC-803 cell lines, and the lowest IC50 value was 0.5 μM. An iterative biosynthesis logic in bathiapeptides' biosynthesis was proposed based on the identified chemical structures and putative gene cluster analysis.
Collapse
Affiliation(s)
- Chuanhai Wu
- Southern Marine Science and Engineering Guangdong Laboratory, Guangzhou, Guangdong, 511458 People's Republic of China
- Department of Ocean Science, The Hong Kong University of Science and Technology, Hong Kong, People's Republic of China
| | - Jianwei Tang
- Southern Marine Science and Engineering Guangdong Laboratory, Guangzhou, Guangdong, 511458 People's Republic of China
- Department of Ocean Science, The Hong Kong University of Science and Technology, Hong Kong, People's Republic of China
| | - Jessie James Limlingan Malit
- Southern Marine Science and Engineering Guangdong Laboratory, Guangzhou, Guangdong, 511458 People's Republic of China
- Department of Ocean Science, The Hong Kong University of Science and Technology, Hong Kong, People's Republic of China
| | - Ruojun Wang
- Southern Marine Science and Engineering Guangdong Laboratory, Guangzhou, Guangdong, 511458 People's Republic of China
- Department of Ocean Science, The Hong Kong University of Science and Technology, Hong Kong, People's Republic of China
| | - Herman H-Y Sung
- Southern Marine Science and Engineering Guangdong Laboratory, Guangzhou, Guangdong, 511458 People's Republic of China
- Department of Chemistry, The Hong Kong University of Science and Technology, Hong Kong, People's Republic of China
| | - Ian D Williams
- Southern Marine Science and Engineering Guangdong Laboratory, Guangzhou, Guangdong, 511458 People's Republic of China
- Department of Chemistry, The Hong Kong University of Science and Technology, Hong Kong, People's Republic of China
| | - Pei-Yuan Qian
- Southern Marine Science and Engineering Guangdong Laboratory, Guangzhou, Guangdong, 511458 People's Republic of China
- Department of Ocean Science, The Hong Kong University of Science and Technology, Hong Kong, People's Republic of China
| |
Collapse
|
42
|
Wang Z, Koirala B, Hernandez Y, Brady SF. Discovery of Paenibacillaceae Family Gram-Negative-Active Cationic Lipopeptide Antibiotics Using Evolution-Guided Chemical Synthesis. Org Lett 2022; 24:4943-4948. [PMID: 35776528 DOI: 10.1021/acs.orglett.2c01879] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Cationic nonribosomal lipopeptides (CNRLPs) from Paenibacillus spp. have been a rewarding source of Gram-negative-active antibiotics. Here we systematically screened sequenced bacterial genomes for CNRLP biosynthetic gene clusters (BGCs) that we predicted might encode additional Gram-negative-active antibiotics. Total chemical synthesis of the bioinformatically predicted products of seven such BGCs led to our identification of new laterocidine, tridecaptin, and paenibacterin-like antibiotics with potent activity against both multiple-drug-resistant Gram-negative and Gram-positive pathogens.
Collapse
Affiliation(s)
- Zongqiang Wang
- Laboratory of Genetically Encoded Small Molecules, The Rockefeller University, 1230 York Avenue, New York, New York 10065, United States
| | - Bimal Koirala
- Laboratory of Genetically Encoded Small Molecules, The Rockefeller University, 1230 York Avenue, New York, New York 10065, United States
| | - Yozen Hernandez
- Laboratory of Genetically Encoded Small Molecules, The Rockefeller University, 1230 York Avenue, New York, New York 10065, United States
| | - Sean F Brady
- Laboratory of Genetically Encoded Small Molecules, The Rockefeller University, 1230 York Avenue, New York, New York 10065, United States
| |
Collapse
|
43
|
Han P, Chen Z, Liu Y, Ma A, Li S, Jia Y. Structural Organization of Brevilaterin Biosynthesis in Brevibacillus laterosporus S62-9: A Novel MbtH-Independent Cationic Antimicrobial Peptide Synthetase System. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:7471-7478. [PMID: 35675382 DOI: 10.1021/acs.jafc.2c01143] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Cationic antimicrobial peptides, produced by nonribosomal peptide synthetases (NRPSs), have received great attention in different applications, including as biocontrol and antimicrobial agents against foodborne pathogenic bacteria. Also, Brevibacillus spp. is a competent microorganism to produce cationic antimicrobial peptides yet has received little attention. Herein, Brevibacillus laterosporus S62-9 genome mining revealed an integrated cationic antimicrobial peptide synthetase system that synthesized brevilaterin. Combining biochemical analysis with bioinformatics elucidated that the A domain from this system was the MbtH-independent enzyme and showed activity against the same amino acid in the structure of brevilaterin. Moreover, the creations of the first three and position 12 residues in the sequence were targeted to bre261, bre270, bre2691A, and bre2662, respectively. Further analysis of the specificity-conferring code of the A domain suggested that a tiny difference would make the activity of the A domain very diverse and the range of substrate selection would be enlarged or narrowed by changing some residues in the code. The dissection of this biosynthesis mechanism would contribute to the successful realization of reasonable artificial design and the modification of bioactive peptides, and this capable organism also would be more fully utilized.
Collapse
Affiliation(s)
- Panpan Han
- School of Food and Health, Beijing Technology and Business University, No.33 Fucheng Road, Haidian District, Beijing 100048, China
| | - Zhou Chen
- School of Food and Health, Beijing Technology and Business University, No.33 Fucheng Road, Haidian District, Beijing 100048, China
| | - Yangliu Liu
- School of Food and Health, Beijing Technology and Business University, No.33 Fucheng Road, Haidian District, Beijing 100048, China
| | - Aijin Ma
- School of Food and Health, Beijing Technology and Business University, No.33 Fucheng Road, Haidian District, Beijing 100048, China
| | - Siting Li
- School of Food and Health, Beijing Technology and Business University, No.33 Fucheng Road, Haidian District, Beijing 100048, China
| | - Yingmin Jia
- School of Food and Health, Beijing Technology and Business University, No.33 Fucheng Road, Haidian District, Beijing 100048, China
| |
Collapse
|
44
|
Malit JJL, Leung HYC, Qian PY. Targeted Large-Scale Genome Mining and Candidate Prioritization for Natural Product Discovery. Mar Drugs 2022; 20:398. [PMID: 35736201 PMCID: PMC9231227 DOI: 10.3390/md20060398] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/08/2022] [Accepted: 06/14/2022] [Indexed: 12/20/2022] Open
Abstract
Large-scale genome-mining analyses have identified an enormous number of cryptic biosynthetic gene clusters (BGCs) as a great source of novel bioactive natural products. Given the sheer number of natural product (NP) candidates, effective strategies and computational methods are keys to choosing appropriate BGCs for further NP characterization and production. This review discusses genomics-based approaches for prioritizing candidate BGCs extracted from large-scale genomic data, by highlighting studies that have successfully produced compounds with high chemical novelty, novel biosynthesis pathway, and potent bioactivities. We group these studies based on their BGC-prioritization logics: detecting presence of resistance genes, use of phylogenomics analysis as a guide, and targeting for specific chemical structures. We also briefly comment on the different bioinformatics tools used in the field and examine practical considerations when employing a large-scale genome mining study.
Collapse
Affiliation(s)
- Jessie James Limlingan Malit
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou 511458, China; (J.J.L.M.); (H.Y.C.L.)
- Department of Ocean Science and Hong Kong Branch of the Southern Marine Science and Engineering Guangdong Laboratory, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Hiu Yu Cherie Leung
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou 511458, China; (J.J.L.M.); (H.Y.C.L.)
- Department of Ocean Science and Hong Kong Branch of the Southern Marine Science and Engineering Guangdong Laboratory, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Pei-Yuan Qian
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou 511458, China; (J.J.L.M.); (H.Y.C.L.)
- Department of Ocean Science and Hong Kong Branch of the Southern Marine Science and Engineering Guangdong Laboratory, The Hong Kong University of Science and Technology, Hong Kong, China
| |
Collapse
|
45
|
Maiti PK, Mandal S. Comprehensive genome analysis of Lentzea reveals repertoire of polymer-degrading enzymes and bioactive compounds with clinical relevance. Sci Rep 2022; 12:8409. [PMID: 35589875 PMCID: PMC9120177 DOI: 10.1038/s41598-022-12427-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 05/03/2022] [Indexed: 11/09/2022] Open
Abstract
The genus Lentzea is a rare group of actinobacteria having potential for the exploration of bioactive compounds. Despite its proven ability to produce compounds with medical relevance, Lentzea genome analysis remains unexplored. Here we show a detailed understanding of the genetic features, biosynthetic gene clusters (BGCs), and genetic clusters for carbohydrate-active enzymes present in the Lentzea genome. Our analysis determines the genes for core proteins, non-ribosomal peptide synthetase condensation domain, and polyketide synthases-ketide synthase domain. The antiSMASH-based sequence analysis identifies 692 BGCs among which 8% are identical to the BGCs that produce geosmin, citrulassin, achromosin (lassopeptide), vancosamine, anabaenopeptin NZ857/nostamide A, alkylresorcinol, BE-54017, and bezastatin. The remaining BGCs code for advanced category antimicrobials like calcium-dependent, glycosylated, terpenoids, lipopeptides, thiopeptide, lanthipeptide, lassopeptide, lingual antimicrobial peptide and lantibiotics together with antiviral, antibacterial, antifungal, antiparasitic, anticancer agents. About 28% of the BGCs, that codes for bioactive secondary metabolites, are exclusive in Lentzea and could lead to new compound discoveries. We also find 7121 genes that code for carbohydrate-degrading enzymes which could essentially convert a wide range of polymeric carbohydrates. Genome mining of such genus is very much useful to give scientific leads for experimental validation in the discovery of new-generation bioactive molecules of biotechnological importance.
Collapse
Affiliation(s)
- Pulak Kumar Maiti
- Laboratory of Molecular Bacteriology, Department of Microbiology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata, 700019, India.
| | - Sukhendu Mandal
- Laboratory of Molecular Bacteriology, Department of Microbiology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata, 700019, India.
| |
Collapse
|
46
|
Hemmerling F, Piel J. Strategies to access biosynthetic novelty in bacterial genomes for drug discovery. Nat Rev Drug Discov 2022; 21:359-378. [PMID: 35296832 DOI: 10.1038/s41573-022-00414-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/24/2022] [Indexed: 12/17/2022]
Abstract
Bacteria provide a rich source of natural products with potential therapeutic applications, such as novel antibiotic classes or anticancer drugs. Bioactivity-guided screening of bacterial extracts and characterization of biosynthetic pathways for drug discovery is now complemented by the availability of large (meta)genomic collections, placing researchers into the postgenomic, big-data era. The progress in next-generation sequencing and the rise of powerful computational tools provide unprecedented insights into unexplored taxa, ecological niches and 'biosynthetic dark matter', revealing diverse and chemically distinct natural products in previously unstudied bacteria. In this Review, we discuss such sources of new chemical entities and the implications for drug discovery with a particular focus on the strategies that have emerged in recent years to identify and access novelty.
Collapse
Affiliation(s)
- Franziska Hemmerling
- Institute of Microbiology, Eidgenössische Technische Hochschule (ETH) Zürich, Zürich, Switzerland
| | - Jörn Piel
- Institute of Microbiology, Eidgenössische Technische Hochschule (ETH) Zürich, Zürich, Switzerland.
| |
Collapse
|
47
|
Santos-Aberturas J, Vior NM. Beyond Soil-Dwelling Actinobacteria: Fantastic Antibiotics and Where to Find Them. Antibiotics (Basel) 2022; 11:195. [PMID: 35203798 PMCID: PMC8868522 DOI: 10.3390/antibiotics11020195] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/27/2022] [Accepted: 01/29/2022] [Indexed: 12/10/2022] Open
Abstract
Bacterial secondary metabolites represent an invaluable source of bioactive molecules for the pharmaceutical and agrochemical industries. Although screening campaigns for the discovery of new compounds have traditionally been strongly biased towards the study of soil-dwelling Actinobacteria, the current antibiotic resistance and discovery crisis has brought a considerable amount of attention to the study of previously neglected bacterial sources of secondary metabolites. The development and application of new screening, sequencing, genetic manipulation, cultivation and bioinformatic techniques have revealed several other groups of bacteria as producers of striking chemical novelty. Biosynthetic machineries evolved from independent taxonomic origins and under completely different ecological requirements and selective pressures are responsible for these structural innovations. In this review, we summarize the most important discoveries related to secondary metabolites from alternative bacterial sources, trying to provide the reader with a broad perspective on how technical novelties have facilitated the access to the bacterial metabolic dark matter.
Collapse
Affiliation(s)
| | - Natalia M. Vior
- Department of Molecular Microbiology, John Innes Centre, Norwich NR7 4UH, UK
| |
Collapse
|
48
|
Iskandar K, Murugaiyan J, Hammoudi Halat D, Hage SE, Chibabhai V, Adukkadukkam S, Roques C, Molinier L, Salameh P, Van Dongen M. Antibiotic Discovery and Resistance: The Chase and the Race. Antibiotics (Basel) 2022; 11:182. [PMID: 35203785 PMCID: PMC8868473 DOI: 10.3390/antibiotics11020182] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 01/23/2022] [Accepted: 01/26/2022] [Indexed: 12/14/2022] Open
Abstract
The history of antimicrobial resistance (AMR) evolution and the diversity of the environmental resistome indicate that AMR is an ancient natural phenomenon. Acquired resistance is a public health concern influenced by the anthropogenic use of antibiotics, leading to the selection of resistant genes. Data show that AMR is spreading globally at different rates, outpacing all efforts to mitigate this crisis. The search for new antibiotic classes is one of the key strategies in the fight against AMR. Since the 1980s, newly marketed antibiotics were either modifications or improvements of known molecules. The World Health Organization (WHO) describes the current pipeline as bleak, and warns about the scarcity of new leads. A quantitative and qualitative analysis of the pre-clinical and clinical pipeline indicates that few antibiotics may reach the market in a few years, predominantly not those that fit the innovative requirements to tackle the challenging spread of AMR. Diversity and innovation are the mainstays to cope with the rapid evolution of AMR. The discovery and development of antibiotics must address resistance to old and novel antibiotics. Here, we review the history and challenges of antibiotics discovery and describe different innovative new leads mechanisms expected to replenish the pipeline, while maintaining a promising possibility to shift the chase and the race between the spread of AMR, preserving antibiotic effectiveness, and meeting innovative leads requirements.
Collapse
Affiliation(s)
- Katia Iskandar
- Department of Mathématiques Informatique et Télécommunications, Université Toulouse III, Paul Sabatier, INSERM, UMR 1295, 31000 Toulouse, France
- INSPECT-LB: Institut National de Santé Publique, d’Épidémiologie Clinique et de Toxicologie-Liban, Beirut 6573, Lebanon;
- Faculty of Pharmacy, Lebanese University, Beirut 6573, Lebanon
| | - Jayaseelan Murugaiyan
- Department of Biological Sciences, SRM University–AP, Amaravati 522502, India; (J.M.); (S.A.)
| | - Dalal Hammoudi Halat
- Department of Pharmaceutical Sciences, School of Pharmacy, Lebanese International University, Bekaa Campus, Beirut 1103, Lebanon
| | - Said El Hage
- Faculty of Medicine, Lebanese University, Beirut 6573, Lebanon;
| | - Vindana Chibabhai
- Division of Clinical Microbiology and Infectious Diseases, School of Pathology, University of the Witwatersrand, Johannesburg 2193, South Africa;
- Microbiology Laboratory, National Health Laboratory Service, Charlotte Maxeke Johannesburg Academic Hospital, Johannesburg 2193, South Africa
| | - Saranya Adukkadukkam
- Department of Biological Sciences, SRM University–AP, Amaravati 522502, India; (J.M.); (S.A.)
| | - Christine Roques
- Laboratoire de Génie Chimique, Department of Bioprocédés et Systèmes Microbiens, Université Paul Sabtier, Toulouse III, UMR 5503, 31330 Toulouse, France;
| | - Laurent Molinier
- Department of Medical Information, Centre Hospitalier Universitaire, INSERM, UMR 1295, Université Paul Sabatier Toulouse III, 31000 Toulouse, France;
| | - Pascale Salameh
- INSPECT-LB: Institut National de Santé Publique, d’Épidémiologie Clinique et de Toxicologie-Liban, Beirut 6573, Lebanon;
- Faculty of Medicine, Lebanese University, Beirut 6573, Lebanon;
- Department of Primary Care and Population Health, University of Nicosia Medical School, Nicosia 2408, Cyprus
| | | |
Collapse
|
49
|
Al Ayed K, Ballantine RD, Hoekstra M, Bann SJ, Wesseling CMJ, Bakker AT, Zhong Z, Li YX, Brüchle NC, van der Stelt M, Cochrane SA, Martin NI. Synthetic Studies with the Brevicidine and Laterocidine Lipopeptide Antibiotics Including Analogues with Enhanced Properties and in vivo Efficacy. Chem Sci 2022; 13:3563-3570. [PMID: 35432860 PMCID: PMC8943889 DOI: 10.1039/d2sc00143h] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 02/09/2022] [Indexed: 12/01/2022] Open
Abstract
Brevicidine and laterocidine are two recently discovered lipopeptide antibiotics with promising antibacterial activity. Possessing a macrocyclic core, multiple positive charges, and a lipidated N-terminus, these lipopeptides exhibit potent and selective activity against Gram-negative pathogens, including polymyxin-resistant isolates. Given the low amounts of brevicidine and laterocidine accessible by fermentation of the producing microorganisms, synthetic routes to these lipopeptides present an attractive alternative. We here report the convenient solid-phase syntheses of both brevicidine and laterocidine and confirm their potent anti-Gram-negative activities. The synthetic routes developed also provide convenient access to novel structural analogues of both brevicidine and laterocidine that display improved hydrolytic stability while maintaining potent antibacterial activity in both in vitro assays and in vivo infection models. Convenient solid-phase approaches are described for the synthesis of brevicidine and laterocidine. Also reported are novel analogues including a laterocidine variant with enhanced hydrolytic stability and potent in vivo antibacterial activity.![]()
Collapse
Affiliation(s)
- Karol Al Ayed
- Biological Chemistry Group, Institute of Biology, Leiden University Sylviusweg 72 2333 BE Leiden The Netherlands
| | - Ross D Ballantine
- School of Chemistry and Chemical Engineering, Queen's University Belfast David Keir Building, Stranmillis Road BT9 5AG Belfast UK
| | - Michael Hoekstra
- Biological Chemistry Group, Institute of Biology, Leiden University Sylviusweg 72 2333 BE Leiden The Netherlands
| | - Samantha J Bann
- School of Chemistry and Chemical Engineering, Queen's University Belfast David Keir Building, Stranmillis Road BT9 5AG Belfast UK
| | - Charlotte M J Wesseling
- Biological Chemistry Group, Institute of Biology, Leiden University Sylviusweg 72 2333 BE Leiden The Netherlands
| | - Alexander T Bakker
- Molecular Physiology Group, Leiden Institute of Chemistry, Leiden University Einsteinweg 55 2333 CC Leiden The Netherlands
| | - Zheng Zhong
- Department of Chemistry, The University of Hong Kong Pokfulam Road Hong Kong China
| | - Yong-Xin Li
- Department of Chemistry, The University of Hong Kong Pokfulam Road Hong Kong China
| | - Nora C Brüchle
- Biological Chemistry Group, Institute of Biology, Leiden University Sylviusweg 72 2333 BE Leiden The Netherlands
| | - Mario van der Stelt
- Molecular Physiology Group, Leiden Institute of Chemistry, Leiden University Einsteinweg 55 2333 CC Leiden The Netherlands
| | - Stephen A Cochrane
- School of Chemistry and Chemical Engineering, Queen's University Belfast David Keir Building, Stranmillis Road BT9 5AG Belfast UK
| | - Nathaniel I Martin
- Biological Chemistry Group, Institute of Biology, Leiden University Sylviusweg 72 2333 BE Leiden The Netherlands
| |
Collapse
|
50
|
Hu Y, Li H, Qu R, He T, Tang X, Chen W, Li L, Bai H, Li C, Wang W, Fu G, Luo G, Xia X, Zhang J. Lysine Stapling Screening Provides Stable and Low Toxic Cationic Antimicrobial Peptides Combating Multidrug-Resistant Bacteria In Vitro and In Vivo. J Med Chem 2021; 65:579-591. [PMID: 34968054 DOI: 10.1021/acs.jmedchem.1c01754] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Cationic antimicrobial peptides (CAMPs) are promising for treatment of multidrug-resistant (MDR) bacteria-caused infections. However, clinical application of CAMPs has been hampered mostly due to their poor proteolytic stability and hemolytic toxicity. Recently, lysine-stapled CAMPs developed by us had been proved to increase peptide stability in vitro without induction of hemolysis. Herein, the applicability of the lysine stapling strategy was further explored by using five natural or artificial CAMPs as model peptides. Lysine stapling screening was implemented to provide 13 cyclic analogues in total. Biological screening of these cyclic analogues showed that CAMPs with a better amphiphilic structure were inclined to exhibit improved antimicrobial activity, protease stability, and biocompatibility after lysine-stapling. One of the stapled analogues of BF15-a1 was found to have extended half-life in plasma, enhanced antimicrobial activity against clinically isolated MDR ESKAPE pathogens, and remained highly effective in combating MRSA infection in a mouse model.
Collapse
Affiliation(s)
- Yuchen Hu
- Innovative Drug Research Centre (IDRC), Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, People's Republic of China
| | - Hong Li
- Innovative Drug Research Centre (IDRC), Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, People's Republic of China
| | - Rui Qu
- Innovative Drug Research Centre (IDRC), Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, People's Republic of China
| | - Tong He
- Innovative Drug Research Centre (IDRC), Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, People's Republic of China
| | - Xiaomin Tang
- Innovative Drug Research Centre (IDRC), Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, People's Republic of China
| | - Wanyi Chen
- Department of Pharmacy, Chongqing University Cancer Hospital, Chongqing 400030, People's Republic of China
| | - Lixian Li
- Department of Pharmacy, Chongqing University Cancer Hospital, Chongqing 400030, People's Republic of China
| | - Hao Bai
- Department of Pharmacy, Chongqing University Cancer Hospital, Chongqing 400030, People's Republic of China
| | - Chao Li
- Department of Pharmacy, Chongqing University Cancer Hospital, Chongqing 400030, People's Republic of China
| | - Wei Wang
- Clinical Laboratory, Chongqing University Cancer Hospital, Chongqing 400030, People's Republic of China
| | - Gang Fu
- Clinical Laboratory, Chongqing University Cancer Hospital, Chongqing 400030, People's Republic of China
| | - Guangli Luo
- Clinical Laboratory, Chongqing University Cancer Hospital, Chongqing 400030, People's Republic of China
| | - Xuefeng Xia
- Innovative Drug Research Centre (IDRC), Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, People's Republic of China
| | - Jinqiang Zhang
- Innovative Drug Research Centre (IDRC), Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, People's Republic of China
| |
Collapse
|