1
|
Islam MZ, Hossain F, Yamazaki M. Single-cell analysis of antimicrobial compound-induced cell death of bacterial cells. J Antimicrob Chemother 2025:dkaf116. [PMID: 40238567 DOI: 10.1093/jac/dkaf116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2025] Open
Abstract
Due to the stochasticity of metabolic reactions and cell cycles of bacterial cells, it is necessary to examine the antibacterial activities of antimicrobial compounds (AMCs) such as antibiotics and antimicrobial peptides (AMPs) at the single-cell level. Here, we review recent studies of the bactericidal activities of AMCs at the single-cell level. First, we discuss recent investigations of the interaction of various AMPs with single bacterial cells, as monitored in real time using optical microscopy. This strategy provides information on AMP-induced membrane damage in single cells [e.g. the onset time of damage to the cell membrane (CM) and outer membrane of single cells]. The rate of AMP-induced CM damage is estimated as the fraction of cells with CM damage [Pdamage (t)] at a specific interaction time t. Second, we discuss the use of single-cell analysis of the bactericidal activity of AMCs. The fraction of dead cells after the exposure to AMCs for time t is determined as the fraction of the microcolonies containing only one cell [Psingle (t)]. For some AMPs, the Pdamage (t) and Psingle (t) values are similar, indicating that AMP-induced CM damage is the direct cause of cell death. Third, we discuss single-cell analysis of the processes and mechanisms of antibiotic-induced cell death. For example, fluoroquinolones and aminoglycosides are observed to induce cytoplasmic condensation and cell lysis, leading to cell death. Based on these studies, we provide our perspective on future investigations using single-cell analysis to assess the processes and the mechanisms of the bactericidal activities of AMCs.
Collapse
Affiliation(s)
- Md Zahidul Islam
- Nanomaterials Research Division, Research Institute of Electronics, Shizuoka University, Shizuoka 422-8529, Japan
- Department of Biotechnology and Genetic Engineering, Jahangirnagar University, Savar, Dhaka 1342, Bangladesh
| | - Farzana Hossain
- Nanomaterials Research Division, Research Institute of Electronics, Shizuoka University, Shizuoka 422-8529, Japan
- Department of Biochemistry and Microbiology, School of Health and Life Sciences, North South University, Bashundhara, Dhaka 1229, Bangladesh
| | - Masahito Yamazaki
- Nanomaterials Research Division, Research Institute of Electronics, Shizuoka University, Shizuoka 422-8529, Japan
- Integrated Bioscience Section, Graduate School of Science and Technology, Shizuoka University, Shizuoka 422-8529, Japan
- Department of Science, Graduate School of Integrated Science and Technology, Shizuoka University, 836 Oya, Suruga-ku, Shizuoka 422-8529, Japan
| |
Collapse
|
2
|
Kato R, Miyazawa K, Imura T, Minamikawa T. Toward nanoscale structural and chemical analysis of microbial surfaces. Biosci Biotechnol Biochem 2025; 89:489-495. [PMID: 39577857 DOI: 10.1093/bbb/zbae176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 11/18/2024] [Indexed: 11/24/2024]
Abstract
Microbial surfaces play a critical role in various biological processes, including cell adhesion and biofilm formation. Understanding these surfaces at the nanoscale is essential for both fundamental and applied microbiology. This review explores recent advancements in nanoscale structural and chemical analyses of microbial surfaces, with a focus on vibrational spectroscopy, such as Raman spectroscopy, infrared spectroscopy, and atomic force microscopy. The review also discusses current challenges of these techniques, including variability in sample preparation and the reproducibility of data, and highlights future directions in nanoscale analysis that could lead to new insights in microbial physiology, antimicrobial resistance, and biofilm research.
Collapse
Affiliation(s)
- Ryo Kato
- Department of Systems Innovation, Graduate School of Engineering Science, Osaka University, Osaka, Japan
| | - Keisuke Miyazawa
- Faculty of Frontier Engineering, Kanazawa University, Kanazawa, Japan
- Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa, Japan
| | - Takumi Imura
- Department of Systems Innovation, Graduate School of Engineering Science, Osaka University, Osaka, Japan
| | - Takeo Minamikawa
- Department of Systems Innovation, Graduate School of Engineering Science, Osaka University, Osaka, Japan
| |
Collapse
|
3
|
McArthur SJ, Umeda K, Kodera N. Nano-Scale Video Imaging of Motility Machinery by High-Speed Atomic Force Microscopy. Biomolecules 2025; 15:257. [PMID: 40001560 PMCID: PMC11852755 DOI: 10.3390/biom15020257] [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: 01/17/2025] [Revised: 02/05/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Motility is a vital aspect of many forms of life, with a wide range of highly conserved as well as highly unique systems adapted to the needs of various organisms and environments. While many motility systems are well studied using structural techniques like X-ray crystallography and electron microscopy, as well as fluorescence microscopy methodologies, it is difficult to directly determine the relationship between the shape and movement of a motility system due to a notable gap in spatiotemporal resolution. Bridging this gap as well as understanding the dynamic molecular movements that underpin motility mechanisms has been challenging. The advent of high-speed atomic force microscopy (HS-AFM) has provided a new window into understanding these nano-scale machines and the dynamic processes underlying motility. In this review, we highlight some of the advances in this field, ranging from reconstituted systems and purified higher-order supramolecular complexes to live cells, in both prokaryotic and eukaryotic contexts.
Collapse
Affiliation(s)
- Steven John McArthur
- WPI Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan;
| | - Kenichi Umeda
- WPI Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan;
- Precursory Research for Embryonic Science and Technology (PRESTO), Japan and Japan Science and Technology Agency (JST), 4-1-8 Honcho, Kawaguchi 332-0012, Japan
| | - Noriyuki Kodera
- WPI Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan;
| |
Collapse
|
4
|
Li X, Sang Z, Zhao X, Wen Y. Metabolic engineering of Streptomyces roseosporus for increased production of clinically important antibiotic daptomycin. Microb Biotechnol 2024; 17:e70038. [PMID: 39487765 PMCID: PMC11530997 DOI: 10.1111/1751-7915.70038] [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: 08/12/2024] [Accepted: 10/10/2024] [Indexed: 11/04/2024] Open
Abstract
Daptomycin (DAP), a novel cyclic lipopeptide antibiotic produced by Streptomyces roseosporus, is clinically important for treatment of infections caused by multidrug-resistant Gram-positive pathogens, but the low yield hampers its large-scale industrial production. Here, we describe a combination metabolic engineering strategy for constructing a DAP high-yielding strain. Initially, we enhanced aspartate (Asp) precursor supply in S. roseosporus wild-type (WT) strain by separately inhibiting Asp degradation and competitive pathway genes using CRISPRi and overexpressing Asp synthetic pathway genes using strong promoter kasOp*. The resulting strains all showed increased DAP titre. Combined inhibition of acsA4, pta, pyrB, and pyrC increased DAP titre to 167.4 μg/mL (73.5% higher than WT value). Co-overexpression of aspC, gdhA, ppc, and ecaA led to DAP titre 168 μg/mL (75.7% higher than WT value). Concurrently, we constructed a chassis strain favourable for DAP production by abolishing by-product production (i.e., deleting a 21.1 kb region of the red pigment biosynthetic gene cluster (BGC)) and engineering the DAP BGC (i.e., replacing its native dptEp with kasOp*). Titre for the resulting chassis strain reached 185.8 μg/mL. Application of our Asp precursor supply strategies to the chassis strain further increased DAP titre to 302 μg/mL (2.1-fold higher than WT value). Subsequently, we cloned the engineered DAP BGC and duplicated it in the chassis strain, leading to DAP titre 274.6 μg/mL. The above strategies, in combination, resulted in maximal DAP titre 350.7 μg/mL (2.6-fold higher than WT value), representing the highest reported DAP titre in shake-flask fermentation. These findings provide an efficient combination strategy for increasing DAP production and can also be readily applied in the overproduction of other Asp-related antibiotics.
Collapse
Affiliation(s)
- Xingwang Li
- State Key Laboratory of Animal Biotech Breeding and College of Biological SciencesChina Agricultural UniversityBeijingChina
| | - Ziwei Sang
- State Key Laboratory of Animal Biotech Breeding and College of Biological SciencesChina Agricultural UniversityBeijingChina
| | - Xuejin Zhao
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of MicrobiologyChinese Academy of SciencesBeijingChina
| | - Ying Wen
- State Key Laboratory of Animal Biotech Breeding and College of Biological SciencesChina Agricultural UniversityBeijingChina
| |
Collapse
|
5
|
Melcrová A, Klein C, Roos WH. Membrane-Active Antibiotics Affect Domains in Bacterial Membranes as the First Step of Their Activity. NANO LETTERS 2024; 24:11800-11807. [PMID: 39145544 PMCID: PMC11440642 DOI: 10.1021/acs.nanolett.4c01873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 08/07/2024] [Accepted: 08/07/2024] [Indexed: 08/16/2024]
Abstract
The need to combat antimicrobial resistance is becoming more and more pressing. Here we investigate the working mechanism of a small cationic agent, N-alkylamide 3d, by conventional and high-speed atomic force microscopy. We show that N-alkylamide 3d interacts with the membrane of Staphylococcus aureus, where it changes the organization and dynamics of lipid domains. After this initial step, supramolecular structures of the antimicrobial agent attach on top of the affected membrane gradually, covering it entirely. These results demonstrate that lateral domains in the bacterial membranes might be affected by small antimicrobial agents more often than anticipated. At the same time, we show a new dual-step activity of N-alkylamide 3d that not only destroys the lateral membrane organization but also effectively covers the whole membrane with aggregates. This final step could render the membrane inaccessible from the outside and possibly prevent signaling and waste disposal of living bacteria.
Collapse
Affiliation(s)
- Adéla Melcrová
- Molecular
Biophysics, Zernike Institute for Advanced Materials, Rijksuniversiteit Groningen, 9712 AG Groningen, The Netherlands
| | - Christiaan Klein
- Molecular
Biophysics, Zernike Institute for Advanced Materials, Rijksuniversiteit Groningen, 9712 AG Groningen, The Netherlands
| | - Wouter H. Roos
- Molecular
Biophysics, Zernike Institute for Advanced Materials, Rijksuniversiteit Groningen, 9712 AG Groningen, The Netherlands
| |
Collapse
|
6
|
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
|
7
|
Asaftei M, Lucidi M, Anton SR, Trompeta AF, Hristu R, Tranca DE, Fiorentis E, Cirtoaje C, Lazar V, Stanciu GA, Cincotti G, Ayala P, Charitidis CA, Holban A, Visca P, Stanciu SG. Antibacterial Interactions of Ethanol-Dispersed Multiwalled Carbon Nanotubes with Staphylococcus aureus and Pseudomonas aeruginosa. ACS OMEGA 2024; 9:33751-33764. [PMID: 39130555 PMCID: PMC11307305 DOI: 10.1021/acsomega.4c03044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 06/19/2024] [Accepted: 06/20/2024] [Indexed: 08/13/2024]
Abstract
Infectious diseases are acknowledged as one of the leading causes of death worldwide. Statistics show that the annual death toll caused by bacterial infections has reached 14 million, most of which are caused by drug-resistant strains. Bacterial antibiotic resistance is currently regarded as a compelling problem with dire consequences, which motivates the urgent identification of alternative ways of fighting bacteria. Various types of nanomaterials have been reported to date as efficient antibacterial solutions. Among these, carbon-based nanomaterials, such as carbon nanodots, carbon graphene oxide, and carbon nanotubes (CNTs), have been shown to be effective in killing a wide panel of pathogenic bacteria. With this study, we aim to provide additional insights into this topic of research by investigating the antibacterial activity of a specific type of multiwalled CNTs, with diameters from 50 to 150 nm, against two representative opportunistic pathogens, i.e., the Gram-positive bacterium Staphylococcus aureus and the Gram-negative bacterium Pseudomonas aeruginosa, both included among the top antibiotic-resistant pathogens. We also test the synergistic effect of CNTs with different antibiotics commonly used in the treatment of infections caused by S. aureus and/or P. aeruginosa. Additionally, a novel approach for quantitatively analyzing bacterial aggregation in brightfield microscopy images was implemented. This method was utilized to assess the effectiveness of CNTs, either alone or in combination with antibiotics, in dispersing bacterial aggregates. Finally, atomic force microscopy coupled with a newly devised image analysis pipeline was used to examine any potential morphological changes in bacterial cells following exposure to CNTs and antibiotics.
Collapse
Affiliation(s)
- Mihaela Asaftei
- Center
for Microscopy-Microanalysis and Information Processing, National University of Science and Technology Politehnica
Bucharest, 313 Splaiul Independentei, 060042 Bucharest, Romania
- Department
of Microbiology and Immunology, Faculty of Biology, Research Institute
of the University of Bucharest, University
of Bucharest, 060101 Bucharest, Romania
| | - Massimiliano Lucidi
- Department
of Science, Roma Tre University, Viale G. Marconi 446, 00146 Rome, Italy
- NBFC,
National Biodiversity Future Center, Piazza Marina 61, 90133 Palermo, Italy
| | - Stefan Razvan Anton
- Center
for Microscopy-Microanalysis and Information Processing, National University of Science and Technology Politehnica
Bucharest, 313 Splaiul Independentei, 060042 Bucharest, Romania
| | - Aikaterini-Flora Trompeta
- Research
Lab of Advanced, Composite, Nano-Materials and Nanotechnology (R-NanoLab),
School of Chemical Engineering, National
Technical University of Athens, 9 Heroon Polytechniou, 15773 Athens, Greece
| | - Radu Hristu
- Center
for Microscopy-Microanalysis and Information Processing, National University of Science and Technology Politehnica
Bucharest, 313 Splaiul Independentei, 060042 Bucharest, Romania
| | - Denis E. Tranca
- Center
for Microscopy-Microanalysis and Information Processing, National University of Science and Technology Politehnica
Bucharest, 313 Splaiul Independentei, 060042 Bucharest, Romania
| | - Efstathios Fiorentis
- Center
for Microscopy-Microanalysis and Information Processing, National University of Science and Technology Politehnica
Bucharest, 313 Splaiul Independentei, 060042 Bucharest, Romania
| | - Cristina Cirtoaje
- Center
for Microscopy-Microanalysis and Information Processing, National University of Science and Technology Politehnica
Bucharest, 313 Splaiul Independentei, 060042 Bucharest, Romania
| | - Veronica Lazar
- Department
of Microbiology and Immunology, Faculty of Biology, Research Institute
of the University of Bucharest, University
of Bucharest, 060101 Bucharest, Romania
| | - George A. Stanciu
- Center
for Microscopy-Microanalysis and Information Processing, National University of Science and Technology Politehnica
Bucharest, 313 Splaiul Independentei, 060042 Bucharest, Romania
| | - Gabriella Cincotti
- Department
of Engineering, Roma Tre University, Viale G. Marconi 446, 00146 Rome, Italy
| | - Paola Ayala
- Faculty
of Physics, University of Vienna, Boltzmanngasse 5, A-1090 Vienna, Austria
| | - Costas A. Charitidis
- Research
Lab of Advanced, Composite, Nano-Materials and Nanotechnology (R-NanoLab),
School of Chemical Engineering, National
Technical University of Athens, 9 Heroon Polytechniou, 15773 Athens, Greece
| | - Alina Holban
- Department
of Microbiology and Immunology, Faculty of Biology, Research Institute
of the University of Bucharest, University
of Bucharest, 060101 Bucharest, Romania
| | - Paolo Visca
- Department
of Science, Roma Tre University, Viale G. Marconi 446, 00146 Rome, Italy
| | - Stefan G. Stanciu
- Center
for Microscopy-Microanalysis and Information Processing, National University of Science and Technology Politehnica
Bucharest, 313 Splaiul Independentei, 060042 Bucharest, Romania
| |
Collapse
|
8
|
Chen Y, Jiang H, Sun Z, Liu F, Su M. Hydantoin derivative dimers as broad-spectrum antimicrobial agents against ESKAPE pathogens with enhanced killing rate and stability. RSC Med Chem 2024; 15:2340-2350. [PMID: 39026634 PMCID: PMC11253853 DOI: 10.1039/d4md00374h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 05/29/2024] [Indexed: 07/20/2024] Open
Abstract
A new series of hydantoin derivative dimers as potential broad-spectrum antibiotic agents is designed and synthesized to combat ESKAPE pathogens. As membrane-active antimicrobial agents, in addition to cationic charged and hydrophobic groups that mimic AMPs (antimicrobial peptides), hydantoin backbones and aromatic linkers increased the rigidity and lipophilicity of the designed compounds, thus improving the stability and bactericidal killing rate. After whole cell phenotypic screening against eight bacterial strains, including MRSA (methicillin-resistant S. aureus), compound 18 was chosen as the lead compound with overall excellent broad-spectrum antibacterial activity (GM = 7.32 μg mL-1) and good selectivity. Kill-kinetic studies of compound 18 showed that the bacterial growth of both Gram-positive and Gram-negative was completely inhibited within one hour, which demonstrated excellent sterilization efficiency of 18. Furthermore, drug resistance and mechanism studies showed that compound 18 exhibited a steady antibacterial performance during 25 passages and could disrupt bacterial cell membrane integrity and cause cell death. Along with the facile synthesis procedures in solution, this series of hydantoin derivative dimer compounds could be an appealing next generation of antibiotic agents to combat emergent drug resistance.
Collapse
Affiliation(s)
- Yating Chen
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Department of Medicinal Chemistry, College of Pharmaceutical Sciences, Soochow University 199 Ren-Ai Road Suzhou Jiangsu 215123 China
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, College of Pharmaceutical Sciences, Soochow University 199 Ren-Ai Road Suzhou Jiangsu 215123 People's Republic of China
| | - Huiqin Jiang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Department of Medicinal Chemistry, College of Pharmaceutical Sciences, Soochow University 199 Ren-Ai Road Suzhou Jiangsu 215123 China
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, College of Pharmaceutical Sciences, Soochow University 199 Ren-Ai Road Suzhou Jiangsu 215123 People's Republic of China
| | - Zibin Sun
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Department of Medicinal Chemistry, College of Pharmaceutical Sciences, Soochow University 199 Ren-Ai Road Suzhou Jiangsu 215123 China
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, College of Pharmaceutical Sciences, Soochow University 199 Ren-Ai Road Suzhou Jiangsu 215123 People's Republic of China
| | - Feng Liu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Department of Medicinal Chemistry, College of Pharmaceutical Sciences, Soochow University 199 Ren-Ai Road Suzhou Jiangsu 215123 China
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, College of Pharmaceutical Sciences, Soochow University 199 Ren-Ai Road Suzhou Jiangsu 215123 People's Republic of China
| | - Ma Su
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Department of Medicinal Chemistry, College of Pharmaceutical Sciences, Soochow University 199 Ren-Ai Road Suzhou Jiangsu 215123 China
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, College of Pharmaceutical Sciences, Soochow University 199 Ren-Ai Road Suzhou Jiangsu 215123 People's Republic of China
| |
Collapse
|
9
|
Ando T, Fukuda S, Ngo KX, Flechsig H. High-Speed Atomic Force Microscopy for Filming Protein Molecules in Dynamic Action. Annu Rev Biophys 2024; 53:19-39. [PMID: 38060998 DOI: 10.1146/annurev-biophys-030722-113353] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
Structural biology is currently undergoing a transformation into dynamic structural biology, which reveals the dynamic structure of proteins during their functional activity to better elucidate how they function. Among the various approaches in dynamic structural biology, high-speed atomic force microscopy (HS-AFM) is unique in the ability to film individual molecules in dynamic action, although only topographical information is acquirable. This review provides a guide to the use of HS-AFM for biomolecular imaging and showcases several examples, as well as providing information on up-to-date progress in HS-AFM technology. Finally, we discuss the future prospects of HS-AFM in the context of dynamic structural biology in the upcoming era.
Collapse
Affiliation(s)
- Toshio Ando
- Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa, Japan;
| | - Shingo Fukuda
- Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa, Japan;
| | - Kien X Ngo
- Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa, Japan;
| | - Holger Flechsig
- Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa, Japan;
| |
Collapse
|
10
|
Lüchtefeld I, Pivkin IV, Gardini L, Zare-Eelanjegh E, Gäbelein C, Ihle SJ, Reichmuth AM, Capitanio M, Martinac B, Zambelli T, Vassalli M. Dissecting cell membrane tension dynamics and its effect on Piezo1-mediated cellular mechanosensitivity using force-controlled nanopipettes. Nat Methods 2024; 21:1063-1073. [PMID: 38802520 PMCID: PMC11166569 DOI: 10.1038/s41592-024-02277-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 04/10/2024] [Indexed: 05/29/2024]
Abstract
The dynamics of cellular membrane tension and its role in mechanosensing, which is the ability of cells to respond to physical stimuli, remain incompletely understood, mainly due to the lack of appropriate tools. Here, we report a force-controlled nanopipette-based method that combines fluidic force microscopy with fluorescence imaging for precise manipulation of the cellular membrane tension while monitoring the impact on single-cell mechanosensitivity. The force-controlled nanopipette enables control of the indentation force imposed on the cell cortex as well as of the aspiration pressure applied to the plasma membrane. We show that this setup can be used to concurrently monitor the activation of Piezo1 mechanosensitive ion channels via calcium imaging. Moreover, the spatiotemporal behavior of the tension propagation is assessed with the fluorescent membrane tension probe Flipper-TR, and further dissected using molecular dynamics modeling. Finally, we demonstrate that aspiration and indentation act independently on the cellular mechanobiological machinery, that indentation induces a local pre-tension in the membrane, and that membrane tension stays confined by links to the cytoskeleton.
Collapse
Affiliation(s)
- Ines Lüchtefeld
- Laboratory for Biosensors and Bioelectronics, ETH Zürich, Zurich, Switzerland.
| | - Igor V Pivkin
- Institute of Computing, Università della Svizzera Italiana, Lugano, Switzerland.
- Swiss Institute of Bioinformatics, Lausanne, Switzerland.
| | - Lucia Gardini
- National Institute of Optics, National Research Council, Florence, Italy
- European Laboratory for Non-Linear Spectroscopy, University of Florence, Florence, Italy
| | | | | | - Stephan J Ihle
- Laboratory for Biosensors and Bioelectronics, ETH Zürich, Zurich, Switzerland
| | - Andreas M Reichmuth
- Laboratory for Biosensors and Bioelectronics, ETH Zürich, Zurich, Switzerland
| | - Marco Capitanio
- European Laboratory for Non-Linear Spectroscopy, University of Florence, Florence, Italy
- Physics and Astronomy Department, University of Florence, Florence, Italy
| | - Boris Martinac
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia
| | - Tomaso Zambelli
- Laboratory for Biosensors and Bioelectronics, ETH Zürich, Zurich, Switzerland.
| | - Massimo Vassalli
- James Watt School of Engineering, University of Glasgow, Glasgow, UK.
| |
Collapse
|
11
|
Su M, Su Y. Recent Advances in Amphipathic Peptidomimetics as Antimicrobial Agents to Combat Drug Resistance. Molecules 2024; 29:2492. [PMID: 38893366 PMCID: PMC11173824 DOI: 10.3390/molecules29112492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 05/21/2024] [Accepted: 05/22/2024] [Indexed: 06/21/2024] Open
Abstract
The development of antimicrobial drugs with novel structures and clear mechanisms of action that are active against drug-resistant bacteria has become an urgent need of safeguarding human health due to the rise of bacterial drug resistance. The discovery of AMPs and the development of amphipathic peptidomimetics have lay the foundation for novel antimicrobial agents to combat drug resistance due to their overall strong antimicrobial activities and unique membrane-active mechanisms. To break the limitation of AMPs, researchers have invested in great endeavors through various approaches in the past years. This review summarized the recent advances including the development of antibacterial small molecule peptidomimetics and peptide-mimic cationic oligomers/polymers, as well as mechanism-of-action studies. As this exciting interdisciplinary field is continuously expanding and growing, we hope this review will benefit researchers in the rational design of novel antimicrobial peptidomimetics in the future.
Collapse
Affiliation(s)
- Ma Su
- College of Pharmaceutical Sciences, Soochow University, 199 Ren-Ai Road, Suzhou 215123, China
| | - Yongxiang Su
- College of Chemistry and Environmental Engineering, Jiaozuo University, Ren-Min Road, Jiaozuo 454000, China;
| |
Collapse
|
12
|
Srivastava D, Patra N. Elucidating Daptomycin's Antibacterial Efficacy: Insights into the Tripartite Complex with Lipid II and Phospholipids in Bacterial Septum Membrane. J Phys Chem B 2024; 128:4414-4427. [PMID: 38690887 DOI: 10.1021/acs.jpcb.4c00332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2024]
Abstract
This study elucidated the mechanism of formation of a tripartite complex containing daptomycin (Dap), lipid II, and phospholipid phosphatidylglycerol in the bacterial septum membrane, which was previously reported as the cause of the antibacterial action of Dap against gram-positive bacteria via molecular dynamics and enhanced sampling methods. Others have suggested that this transient complex ushers in the inhibition of cell wall synthesis by obstructing the downstream polymerization and cross-linking processes involving lipid II, which is absent in the presence of cardiolipin lipid in the membrane. In this work, we observed that the complex was stabilized by Ca2+-mediated electrostatic interactions between Dap and lipid head groups, hydrophobic interaction, hydrogen bonds, and salt bridges between the lipopeptide and lipids and was associated with Dap concentration-dependent membrane depolarization, thinning of the bilayer, and increased lipid tail disorder. Residues Orn6 and Kyn13, along with the DXDG motif, made simultaneous contact with constituent lipids, hence playing a crucial role in the formation of the complex. Incorporating cardiolipin into the membrane model led to its competitively displacing lipid II away from the Dap, reducing the lifetime of the complex and the nonexistence of lipid tail disorder and membrane depolarization. No evidence of water permeation inside the membrane hydrophobic interior was noted in all of the systems studied. Additionally, it was shown that using hydrophobic contacts between Dap and lipids as collective variables for enhanced sampling gave rise to a free energy barrier for the translocation of the lipopeptide. A better understanding of Dap's antibacterial mechanism, as studied through this work, will help develop lipopeptide-based antibiotics for rising Dap-resistant bacteria.
Collapse
Affiliation(s)
- Diship Srivastava
- Department of Chemistry and Chemical Biology, Indian Institute of Technology (ISM) Dhanbad, Dhanbad 826004, India
| | - Niladri Patra
- Department of Chemistry and Chemical Biology, Indian Institute of Technology (ISM) Dhanbad, Dhanbad 826004, India
| |
Collapse
|
13
|
Taylor SD, Moreira R. Daptomycin: Mechanism of action, mechanisms of resistance, synthesis and structure-activity relationships. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 212:163-234. [PMID: 40122645 DOI: 10.1016/bs.pmbts.2024.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
Daptomycin is a cyclic lipodepsipeptide antibiotic that is a mainstay for the treatment of serious infections caused by Gram-positive bacteria, including methicillin-resistant Streptococcus aureus and vancomycin resistant enterococci. It is one of the so-called last-resort antibiotics that are used to tackle life-threatening infections that do not respond to first-line treatments. However, resistance to daptomycin is eroding its clinical efficacy motivating the design and/or discovery of analogues that overcome resistance. The strategy of antibiotic analogue synthesis has been used to overcome bacterial resistance to many classes of antibiotics such as the β-lactams. Pursuing this strategy with daptomycin requires a detailed understanding of daptomycin's action mechanism and synthesis. Here, we discuss the action mechanism of daptomycin in a holistic manner and expand this discussion to rationalize conferred modes of resistance. Synthetic efforts, both chemical and biological, are discussed in detail and the structure-activity relationship emanating from these works is distilled into a usable model that can guide the design of new daptomycin analogues.
Collapse
Affiliation(s)
- Scott D Taylor
- Department of Chemistry, University of Waterloo, Waterloo, ON, Canada.
| | - Ryan Moreira
- Department of Chemistry and Howard Hughes Medical Institute, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| |
Collapse
|
14
|
Zhang S, Cui M, Liu D, Fu B, Shi T, Wang Y, Sun C, Wu C. Tigecycline Sensitivity Reduction in Escherichia coli Due to Widely Distributed tet(A) Variants. Microorganisms 2023; 11:3000. [PMID: 38138144 PMCID: PMC10745318 DOI: 10.3390/microorganisms11123000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 12/14/2023] [Accepted: 12/16/2023] [Indexed: 12/24/2023] Open
Abstract
Despite scattered studies that have reported mutations in the tet(A) gene potentially linked to tigecycline resistance in clinical pathogens, the detailed function and epidemiology of these tet(A) variants remains limited. In this study, we analyzed 64 Escherichia coli isolates derived from MacConkey plates supplemented with tigecycline (2 μg/mL) and identified five distinct tet(A) variants that account for reduced sensitivity to tigecycline. In contrast to varied tigecycline MICs (0.25 to 16 μg/mL) of the 64 tet(A)-variant-positive E. coli isolates, gene function analysis confirmed that the five tet(A) variants exhibited a similar capacity to reduce tigecycline sensitivity in DH5α carrying pUC19. Among the observed seven non-synonymous mutations, the V55M mutation was unequivocally validated for its positive role in conferring tigecycline resistance. Interestingly, the variability in tigecycline MICs among the E. coli strains did not correlate with tet(A) gene expression. Instead, a statistically significant reduction in intracellular tigecycline concentrations was noted in strains displaying higher MICs. Genomic analysis of 30 representative E. coli isolates revealed that tet(A) variants predominantly resided on plasmids (n = 14) and circular intermediates (n = 13). Within China, analysis of a well-characterized E. coli collection isolated from pigs and chickens in 2018 revealed the presence of eight tet(A) variants in 103 (4.2%, 95% CI: 3.4-5.0%) isolates across 13 out of 17 tested Chinese provinces or municipalities. Globally, BLASTN analysis identified 21 tet(A) variants in approximately 20.19% (49,423/244,764) of E. coli genomes in the Pathogen Detection database. These mutant tet(A) genes have been widely disseminated among E. coli isolates from humans, food animals, and the environment sectors, exhibiting a growing trend in tet(A) variants over five decades. Our findings underscore the urgency of addressing tigecycline resistance and the underestimated role of tet(A) mutations in this context.
Collapse
Affiliation(s)
- Shan Zhang
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (S.Z.); (D.L.); (B.F.); (T.S.); (Y.W.)
- Key Laboratory of Animal Antimicrobial Resistance Surveillance, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Mingquan Cui
- China Institute of Veterinary Drug Control, Beijing 100081, China;
| | - Dejun Liu
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (S.Z.); (D.L.); (B.F.); (T.S.); (Y.W.)
- Key Laboratory of Animal Antimicrobial Resistance Surveillance, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Bo Fu
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (S.Z.); (D.L.); (B.F.); (T.S.); (Y.W.)
- Key Laboratory of Animal Antimicrobial Resistance Surveillance, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Tingxuan Shi
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (S.Z.); (D.L.); (B.F.); (T.S.); (Y.W.)
- Key Laboratory of Animal Antimicrobial Resistance Surveillance, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Yang Wang
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (S.Z.); (D.L.); (B.F.); (T.S.); (Y.W.)
- Key Laboratory of Animal Antimicrobial Resistance Surveillance, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Chengtao Sun
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (S.Z.); (D.L.); (B.F.); (T.S.); (Y.W.)
- Key Laboratory of Animal Antimicrobial Resistance Surveillance, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Congming Wu
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (S.Z.); (D.L.); (B.F.); (T.S.); (Y.W.)
- Key Laboratory of Animal Antimicrobial Resistance Surveillance, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| |
Collapse
|
15
|
Fukuda S, Ando T. Technical advances in high-speed atomic force microscopy. Biophys Rev 2023; 15:2045-2058. [PMID: 38192344 PMCID: PMC10771405 DOI: 10.1007/s12551-023-01171-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 11/19/2023] [Indexed: 01/10/2024] Open
Abstract
It has been 30 years since the outset of developing high-speed atomic force microscopy (HS-AFM), and 15 years have passed since its establishment in 2008. This advanced microscopy is capable of directly visualizing individual biological macromolecules in dynamic action and has been widely used to answer important questions that are inaccessible by other approaches. The number of publications on the bioapplications of HS-AFM has rapidly increased in recent years and has already exceeded 350. Although less visible than these biological studies, efforts have been made for further technical developments aimed at enhancing the fundamental performance of HS-AFM, such as imaging speed, low sample disturbance, and scan size, as well as expanding its functionalities, such as correlative microscopy, temperature control, buffer exchange, and sample manipulations. These techniques can expand the range of HS-AFM applications. After summarizing the key technologies underlying HS-AFM, this article focuses on recent technical advances and discusses next-generation HS-AFM.
Collapse
Affiliation(s)
- Shingo Fukuda
- Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kakuma-Machi, Kanazawa, 920-1192 Japan
| | - Toshio Ando
- Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kakuma-Machi, Kanazawa, 920-1192 Japan
| |
Collapse
|
16
|
Guan D, Li J, Chen F, Li J, Bian X, Yu Y, Feng X, Lan L, Huang W. A facile and selective derivatization approach on kynurenine-NH 2 in daptomycin, leading to the discovery of hexakynomycin to combat multidrug-resistant Gram-positive pathogens especially daptomycin-resistant bacteria. Eur J Med Chem 2023; 259:115638. [PMID: 37482019 DOI: 10.1016/j.ejmech.2023.115638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/03/2023] [Accepted: 07/12/2023] [Indexed: 07/25/2023]
Abstract
Wide-spread use of daptomycin unavoidably resulted in the emergence of daptomycin-resistant pathogens. In the hunt for more active daptomycin derivatives through medicinal chemistry studies, we established a concise semisynthetic approach to modify the L-Kyn13 on daptomycin specifically and effectively. Here, 19 novel derivatives with certain diversity were designed and synthesized to perform a comprehensive SAR study on this underestimated position. The optimal compound, termed "hexakynomycin", as the new generation of daptomycin-based antibiotic candidate exhibited 4->125-fold higher activity against methicillin-susceptible S. aureus (MSSA), methicillin-resistant S. aureus (MRSA), vancomycin-intermediate resistant S. aureus (VISA), and vancomycin-resistant Enterococci (VRE), including daptomycin-resistant strains, compared with that of daptomycin. Greater membrane binding capacity rendered hexakynomycin better activity and special antibiotic property. Hexakynomycin also demonstrated a better pharmacokinetic profile, good safety features and good pharmacodynamics properties. This work provided an effective modification strategy aiming at daptomycin which provided significant insights and showed great promise for the next generation of daptomycin derivatives.
Collapse
Affiliation(s)
- Dongliang Guan
- State Key Laboratory of Drug Research, Center for Biotherapeutics Discovery Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No.555 Zuchongzhi Rd, Pudong, Shanghai, 201203, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong, 264117, China; University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China.
| | - Jian Li
- State Key Laboratory of Drug Research, Center for Biotherapeutics Discovery Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No.555 Zuchongzhi Rd, Pudong, Shanghai, 201203, China; School of Pharmaceutical Science and Technology, Hangzhou Institute of Advanced Study, Hangzhou, 310024, China
| | - Feifei Chen
- State Key Laboratory of Drug Research, Center for Biotherapeutics Discovery Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No.555 Zuchongzhi Rd, Pudong, Shanghai, 201203, China; School of Pharmaceutical Science and Technology, Hangzhou Institute of Advanced Study, Hangzhou, 310024, China
| | - Jiaqi Li
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan, 410082, China
| | - Xiaolei Bian
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong, 264117, China
| | - Yue Yu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan, 410082, China
| | - Xinxin Feng
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan, 410082, China.
| | - Lefu Lan
- School of Pharmaceutical Science and Technology, Hangzhou Institute of Advanced Study, Hangzhou, 310024, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China.
| | - Wei Huang
- State Key Laboratory of Drug Research, Center for Biotherapeutics Discovery Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No.555 Zuchongzhi Rd, Pudong, Shanghai, 201203, China; School of Pharmaceutical Science and Technology, Hangzhou Institute of Advanced Study, Hangzhou, 310024, China; University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China.
| |
Collapse
|
17
|
Chen EHL, Wang CH, Liao YT, Chan FY, Kanaoka Y, Uchihashi T, Kato K, Lai L, Chang YW, Ho MC, Chen RPY. Visualizing the membrane disruption action of antimicrobial peptides by cryo-electron tomography. Nat Commun 2023; 14:5464. [PMID: 37673860 PMCID: PMC10482868 DOI: 10.1038/s41467-023-41156-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/24/2023] [Indexed: 09/08/2023] Open
Abstract
The abuse of antibiotics has led to the emergence of multidrug-resistant microbial pathogens, presenting a pressing challenge in global healthcare. Membrane-disrupting antimicrobial peptides (AMPs) combat so-called superbugs via mechanisms different than conventional antibiotics and have good application prospects in medicine, agriculture, and the food industry. However, the mechanism-of-action of AMPs has not been fully characterized at the cellular level due to a lack of high-resolution imaging technologies that can capture cellular-membrane disruption events in the hydrated state. Previously, we reported PepD2M, a de novo-designed AMP with potent and wide-spectrum bactericidal and fungicidal activity. In this study, we use cryo-electron tomography (cryo-ET) and high-speed atomic force microscopy (HS-AFM) to directly visualize the pepD2M-induced disruption of the outer and inner membranes of the Gram-negative bacterium Escherichia coli, and compared with a well-known pore-forming peptide, melittin. Our high-resolution cryo-ET images reveal how pepD2M disrupts the E. coli membrane using a carpet/detergent-like mechanism. Our studies reveal the direct membrane-disrupting consequence of AMPs on the bacterial membrane by cryo-ET, and this information provides critical insights into the mechanisms of this class of antimicrobial agents.
Collapse
Affiliation(s)
- Eric H-L Chen
- Institute of Biological Chemistry, Academia Sinica, Taipei, 11529, Taiwan
| | - Chun-Hsiung Wang
- Institute of Biological Chemistry, Academia Sinica, Taipei, 11529, Taiwan
| | - Yi-Ting Liao
- Institute of Biological Chemistry, Academia Sinica, Taipei, 11529, Taiwan
| | - Feng-Yueh Chan
- Department of Physics, Nagoya University, Nagoya, 464-8602, Japan
| | - Yui Kanaoka
- Department of Physics, Nagoya University, Nagoya, 464-8602, Japan
| | - Takayuki Uchihashi
- Department of Physics, Nagoya University, Nagoya, 464-8602, Japan
- Institute for Glyco-core Research (iGCORE), Nagoya University, Nagoya, 464-8602, Japan
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Aichi, 444-8787, Japan
| | - Koichi Kato
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Aichi, 444-8787, Japan
- Institute for Molecular Science, National Institutes of Natural Sciences, Okazaki, Aichi, 444-8787, Japan
- Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, 467-8603, Japan
| | - Longsheng Lai
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104-6059, USA
| | - Yi-Wei Chang
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104-6059, USA
| | - Meng-Chiao Ho
- Institute of Biological Chemistry, Academia Sinica, Taipei, 11529, Taiwan.
- Institute of Biochemical Sciences, National Taiwan University, Taipei, 10617, Taiwan.
- Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, 100, Taiwan.
| | - Rita P-Y Chen
- Institute of Biological Chemistry, Academia Sinica, Taipei, 11529, Taiwan.
- Institute of Biochemical Sciences, National Taiwan University, Taipei, 10617, Taiwan.
| |
Collapse
|
18
|
Melcrová A, Maity S, Melcr J, de Kok NAW, Gabler M, van der Eyden J, Stensen W, Svendsen JSM, Driessen AJM, Marrink SJ, Roos WH. Lateral membrane organization as target of an antimicrobial peptidomimetic compound. Nat Commun 2023; 14:4038. [PMID: 37419980 PMCID: PMC10328936 DOI: 10.1038/s41467-023-39726-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 06/20/2023] [Indexed: 07/09/2023] Open
Abstract
Antimicrobial resistance is one of the leading concerns in medical care. Here we study the mechanism of action of an antimicrobial cationic tripeptide, AMC-109, by combining high speed-atomic force microscopy, molecular dynamics, fluorescence assays, and lipidomic analysis. We show that AMC-109 activity on negatively charged membranes derived from Staphylococcus aureus consists of two crucial steps. First, AMC-109 self-assembles into stable aggregates consisting of a hydrophobic core and a cationic surface, with specificity for negatively charged membranes. Second, upon incorporation into the membrane, individual peptides insert into the outer monolayer, affecting lateral membrane organization and dissolving membrane nanodomains, without forming pores. We propose that membrane domain dissolution triggered by AMC-109 may affect crucial functions such as protein sorting and cell wall synthesis. Our results indicate that the AMC-109 mode of action resembles that of the disinfectant benzalkonium chloride (BAK), but with enhanced selectivity for bacterial membranes.
Collapse
Affiliation(s)
- Adéla Melcrová
- Molecular Biophysics, Zernike Institute for Advanced Materials, Rijksuniversiteit Groningen, Groningen, the Netherlands
| | - Sourav Maity
- Molecular Biophysics, Zernike Institute for Advanced Materials, Rijksuniversiteit Groningen, Groningen, the Netherlands
| | - Josef Melcr
- Molecular Dynamics, Groningen Biomolecular Sciences & Biotechnology Institute, Rijksuniversiteit Groningen, Groningen, the Netherlands
| | - Niels A W de Kok
- Molecular Microbiology, Groningen Biomolecular Sciences & Biotechnology Institute, Rijksuniversiteit Groningen, Groningen, the Netherlands
| | - Mariella Gabler
- Molecular Biophysics, Zernike Institute for Advanced Materials, Rijksuniversiteit Groningen, Groningen, the Netherlands
| | - Jonne van der Eyden
- Molecular Biophysics, Zernike Institute for Advanced Materials, Rijksuniversiteit Groningen, Groningen, the Netherlands
| | - Wenche Stensen
- Department of Chemistry, UiT Arctic University of Norway, Tromsø, Norway
| | - John S M Svendsen
- Department of Chemistry, UiT Arctic University of Norway, Tromsø, Norway
| | - Arnold J M Driessen
- Molecular Microbiology, Groningen Biomolecular Sciences & Biotechnology Institute, Rijksuniversiteit Groningen, Groningen, the Netherlands
| | - Siewert J Marrink
- Molecular Biophysics, Zernike Institute for Advanced Materials, Rijksuniversiteit Groningen, Groningen, the Netherlands
- Molecular Dynamics, Groningen Biomolecular Sciences & Biotechnology Institute, Rijksuniversiteit Groningen, Groningen, the Netherlands
| | - Wouter H Roos
- Molecular Biophysics, Zernike Institute for Advanced Materials, Rijksuniversiteit Groningen, Groningen, the Netherlands.
| |
Collapse
|
19
|
Ding J, Zeng S, Wang Y, Yin X, Zhang B, Zhang B, Xu S, Zhang Y, Zheng J, Fan J, Wang M. Metal coordinating-induced self-assembly of cyclic lipopeptides into high-performance antimicrobial supramolecules. Food Chem 2023; 422:136203. [PMID: 37121207 DOI: 10.1016/j.foodchem.2023.136203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 12/10/2022] [Accepted: 04/17/2023] [Indexed: 05/02/2023]
Abstract
This study designed a green hydrothermally-chelating approach to generate robust antimicrobial complexes via metal-coordinated supramolecular self-assembly of cyclic lipopeptides (CLs). The metal ion (Ca2+ and Zn2+)-coordinated CL (Ca/CL or Zn/CL complex; 1 mg/mL) demonstrated potent antibacterial activity against fungi (A. niger) and bacteria (E. coli and S. aureus) respectively, and in particular, completely suppressed the microbial resistance. Further physicochemical and spectal analysis showed that this coordination approach led to CL with enhanced hydrophobic and intermolecular electrostatic interactions, forming β-sheet-rich secondary structures allowing the complexes easily contact with and destroy the membrane of microorganisms. Practical application experiments validated that the Ca/CL and Zn/CL complexes strongly avoided table grape and fresh tomato from the contamination of pathogen. The findings of this study laid foundation for the utilization of metal ions to improve the biological activity of natural antimicrobial peptides.
Collapse
Affiliation(s)
- Jinglin Ding
- College of Biological Sciences and Technology, Beijing Key Laboratory of Food Processing and Safety in Forestry, Beijing Forestry University, Beijing 100083, China
| | - Shufan Zeng
- College of Biological Sciences and Technology, Beijing Key Laboratory of Food Processing and Safety in Forestry, Beijing Forestry University, Beijing 100083, China
| | - Yueqing Wang
- College of Biological Sciences and Technology, Beijing Key Laboratory of Food Processing and Safety in Forestry, Beijing Forestry University, Beijing 100083, China
| | - Xiaoyu Yin
- College of Biological Sciences and Technology, Beijing Key Laboratory of Food Processing and Safety in Forestry, Beijing Forestry University, Beijing 100083, China
| | - Bo Zhang
- College of Biological Sciences and Technology, Beijing Key Laboratory of Food Processing and Safety in Forestry, Beijing Forestry University, Beijing 100083, China
| | - Bolin Zhang
- College of Biological Sciences and Technology, Beijing Key Laboratory of Food Processing and Safety in Forestry, Beijing Forestry University, Beijing 100083, China
| | - Shandong Xu
- College of Science, Beijing Forestry University, Beijing 100083, China
| | - Yanyan Zhang
- Food Science and Engineering College, Beijing University of Agriculture, Beijing 102206, China
| | - Jiangfu Zheng
- College of Environmental Science and Engineering, Hunan University, Changsha 410082, China
| | - Junfeng Fan
- College of Biological Sciences and Technology, Beijing Key Laboratory of Food Processing and Safety in Forestry, Beijing Forestry University, Beijing 100083, China.
| | - Mengze Wang
- School of Food & Wine, Ningxia University, Yinchuan 750021, China.
| |
Collapse
|
20
|
Sreedharan SM, Rishi N, Singh R. Microbial Lipopeptides: Properties, Mechanics and Engineering for Novel Lipopeptides. Microbiol Res 2023; 271:127363. [PMID: 36989760 DOI: 10.1016/j.micres.2023.127363] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 12/04/2022] [Accepted: 03/11/2023] [Indexed: 03/17/2023]
Abstract
Microorganisms produce active surface agents called lipopeptides (LPs) which are amphiphilic in nature. They are cyclic or linear compounds and are predominantly isolated from Bacillus and Pseudomonas species. LPs show antimicrobial activity towards various plant pathogens and act by inhibiting the growth of these organisms. Several mechanisms are exhibited by LPs, such as cell membrane disruption, biofilm production, induced systematic resistance, improving plant growth, inhibition of spores, etc., making them suitable as biocontrol agents and highly advantageous for industrial utilization. The biosynthesis of lipopeptides involves large multimodular enzymes referred to as non-ribosomal peptide synthases. These enzymes unveil a broad range of engineering approaches through which lipopeptides can be overproduced and new LPs can be generated asserting high efficacy. Such approaches involve several synthetic biology systems and metabolic engineering techniques such as promotor engineering, enhanced precursor availability, condensation domain engineering, and adenylation domain engineering. Finally, this review provides an update of the applications of lipopeptides in various fields.
Collapse
|
21
|
Seeing the unseen: High-resolution AFM imaging captures antibiotic action in bacterial membranes. Nat Commun 2022; 13:6196. [PMID: 36271086 PMCID: PMC9587010 DOI: 10.1038/s41467-022-33839-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 10/04/2022] [Indexed: 12/24/2022] Open
|
22
|
Yeh SL, Narasimhalu N, Vom Steeg LG, Muthami J, LeConey S, He Z, Pitcher M, Cassady H, Morley VJ, Cho SH, Bator C, Koshani R, Woods RJ, Hickner M, Read AF, Sheikhi A. Ion Exchange Biomaterials to Capture Daptomycin and Prevent Resistance Evolution in Off-Target Bacterial Populations. ACS APPLIED MATERIALS & INTERFACES 2022; 14:42864-42875. [PMID: 36103577 DOI: 10.1021/acsami.2c14894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Daptomycin (DAP), a cyclic anionic lipopeptide antibiotic, is among the last resorts to treat multidrug-resistant Gram-positive bacterial infections, caused by vancomycin-resistant Enterococcus faecium or methicillin-resistant Staphylococcus aureus. DAP is administered intravenously, and via biliary excretion, ∼5-10% of the intravenous DAP dose arrives in the gastrointestinal (GI) tract where it drives resistance evolution in the off-target populations of E. faecium bacteria. Previously, we have shown in vivo that the oral administration of cholestyramine, an ion exchange biomaterial (IXB) sorbent, prevents DAP treatment from enriching DAP resistance in the populations of E. faecium shed from mice. Here, we investigate the biomaterial-DAP interfacial interactions to uncover the antibiotic removal mechanisms. The IXB-mediated DAP capture from aqueous media was measured in controlled pH/electrolyte solutions and in the simulated intestinal fluid (SIF) to uncover the molecular and colloidal mechanisms of DAP removal from the GI tract. Our findings show that the IXB electrostatically adsorbs the anionic antibiotic via a time-dependent diffusion-controlled process. Unsteady-state diffusion-adsorption mass balance describes the dynamics of adsorption well, and the maximum removal capacity is beyond the electric charge stoichiometric ratio because of DAP self-assembly. This study may open new opportunities for optimizing cholestyramine adjuvant therapy to prevent DAP resistance, as well as designing novel biomaterials to remove off-target antibiotics from the GI tract.
Collapse
Affiliation(s)
- Shang-Lin Yeh
- Department of Chemical Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Naveen Narasimhalu
- Department of Chemical Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Landon G Vom Steeg
- Department of Biology, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Joy Muthami
- Department of Chemical Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Sean LeConey
- Department of Chemical Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Zeming He
- Department of Chemical Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Mica Pitcher
- Department of Chemical Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
- Department of Chemistry, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Harrison Cassady
- Department of Materials Science and Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Valerie J Morley
- Department of Biology, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Sung Hyun Cho
- Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Carol Bator
- Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Roya Koshani
- Department of Chemical Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Robert J Woods
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Michael Hickner
- Department of Chemical Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
- Department of Materials Science and Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Andrew F Read
- Department of Biology, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
- Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Amir Sheikhi
- Department of Chemical Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| |
Collapse
|
23
|
Chu S, Hu W, Zhang K, Hui F. Breeding of High Daptomycin-Producing Strain by Streptomycin Resistance Superposition. Pol J Microbiol 2022; 71:463-471. [PMID: 36185027 PMCID: PMC9608166 DOI: 10.33073/pjm-2022-041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 08/02/2022] [Indexed: 11/24/2022] Open
Abstract
Daptomycin is a cyclolipopeptide antibiotic produced by Streptomyces roseosporus. It is widely used to treat drug-resistant bacterial infections; however, daptomycin yield in wild strains is very low. To improve the daptomycin production by the strain BNCC 342432, a modified method of ribosome engineering with superposition of streptomycin resistance was adopted in this study. The highest-yield mutant strain SR-2620 was obtained by increasing streptomycin resistance of BNCC 342432, and achieved daptomycin production of 38.5 mg/l in shake-flask fermentation, 1.79-fold higher than the parent strain and its heredity stability was stable. The morphological characteristics of the two strains were significantly different, and the 440th base G of the rpsL gene in the mutant strain was deleted, which resulted in a frameshift mutation. Our results demonstrate that gradually increasing strain resistance to streptomycin was an effective breeding method to improve daptomycin yield in S. roseosporus.
Collapse
Affiliation(s)
- Shuaibei Chu
- College of Life Science and Agricultural Engineering, Nanyang Normal University, Nanyang, China
| | - Wenting Hu
- College of Life Science and Agricultural Engineering, Nanyang Normal University, Nanyang, China
| | - Kaihong Zhang
- College of Life Science and Agricultural Engineering, Nanyang Normal University, Nanyang, China
| | - Fengli Hui
- College of Life Science and Agricultural Engineering, Nanyang Normal University, Nanyang, China,Research Center of Henan Provincial Agricultural Biomass Resource Engineering and Technology, Nanyang, China, E-mail:
| |
Collapse
|
24
|
Chen Q, Zhu J, Li X, Wen Y. Transcriptional Regulator DasR Represses Daptomycin Production through Both Direct and Cascade Mechanisms in Streptomyces roseosporus. Antibiotics (Basel) 2022; 11:antibiotics11081065. [PMID: 36009934 PMCID: PMC9404778 DOI: 10.3390/antibiotics11081065] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/02/2022] [Accepted: 08/04/2022] [Indexed: 11/16/2022] Open
Abstract
Daptomycin, produced by Streptomyces roseosporus, is a clinically important cyclic lipopeptide antibiotic used for the treatment of human infections caused by drug-resistant Gram-positive pathogens. In contrast to most Streptomyces antibiotic biosynthetic gene clusters (BGCs), daptomycin BGC has no cluster-situated regulator (CSR) genes. DasR, a GntR-family transcriptional regulator (TR) widely present in the genus, was shown to regulate antibiotic production in model species S. coelicolor by binding to promoter regions of CSR genes. New findings reported here reveal that DasR pleiotropically regulates production of daptomycin and reddish pigment, and morphological development in S. roseosporus. dasR deletion enhanced daptomycin production and morphological development, but reduced pigment production. DasR inhibited daptomycin production by directly repressing dpt structural genes and global regulatory gene adpA (whose product AdpA protein activates daptomycin production and morphological development). DasR-protected regions on dptEp and adpAp contained a 16 nt sequence similar to the consensus DasR-binding site dre in S. coelicolor. AdpA was shown to target dpt structural genes and dptR2 (which encodes a DeoR-family TR required for daptomycin production). A 10 nt sequence similar to the consensus AdpA-binding site was found on target promoter regions dptAp and dptR2p. This is the first demonstration that DasR regulates antibiotic production both directly and through a cascade mechanism. The findings expand our limited knowledge of the regulatory network underlying daptomycin production, and will facilitate methods for construction of daptomycin overproducers.
Collapse
Affiliation(s)
- Qiong Chen
- State Key Laboratory of Agrobiotechnology and College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Jianya Zhu
- Institute of Fisheries Research, Beijing Academy of Agriculture and Forestry Sciences, Beijing 100068, China
| | - Xingwang Li
- State Key Laboratory of Agrobiotechnology and College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Ying Wen
- State Key Laboratory of Agrobiotechnology and College of Biological Sciences, China Agricultural University, Beijing 100193, China
- Correspondence: ; Tel.: +86-10-62732715
| |
Collapse
|
25
|
Mescola A, Ragazzini G, Facci P, Alessandrini A. The potential of AFM in studying the role of the nanoscale amphipathic nature of (lipo)-peptides interacting with lipid bilayers. NANOTECHNOLOGY 2022; 33:432001. [PMID: 35830770 DOI: 10.1088/1361-6528/ac80c9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 07/13/2022] [Indexed: 06/15/2023]
Abstract
Antimicrobial peptides (AMPs) and lipopeptides (LPs) represent very promising molecules to fight resistant bacterial infections due to their broad-spectrum of activity, their first target, i.e. the bacterial membrane, and the rapid bactericidal action. For both types of molecules, the action mechanism starts from the membrane of the pathogen agents, producing a disorganization of their phase structure or the formation of pores of different size altering their permeability. This mechanism of action is based on physical interactions more than on a lock-and-key recognition event and it is difficult for the pathogens to rapidly develop an effective resistance. Very small differences in the sequence of both AMPs and LPs might lead to very different effects on the target membrane. Therefore, a correct understanding of their mechanism of action is required with the aim of developing new synthetic peptides, analogues of the natural ones, with specific and more powerful bactericidal activity. Atomic force microscopy (AFM), with its high resolution and the associated force spectroscopy resource, provides a valuable technique to investigate the reorganization of lipid bilayers exposed to antimicrobial or lipopeptides. Here, we present AFM results obtained by ours and other groups on the action of AMPs and LPs on supported lipid bilayers (SLBs) of different composition. We also consider data obtained by fluorescence microscopy to compare the AFM data with another technique which can be used on different lipid bilayer model systems such as SLBs and giant unilamellar vesicles. The outcomes here presented highlight the powerful of AFM-based techniques in detecting nanoscale peptide-membrane interactions and strengthen their use as an exceptional complementary tool toin vivoinvestigations. Indeed, the combination of these approaches can help decipher the mechanisms of action of different antimicrobials and lipopeptides at both the micro and nanoscale levels, and to design new and more efficient antimicrobial compounds.
Collapse
Affiliation(s)
- Andrea Mescola
- CNR-Nanoscience Institute-S3, Via Campi 213/A, I-41125, Modena, Italy
| | - Gregorio Ragazzini
- Department of Physics, Informatics and Mathematics, University of Modena and Reggio Emilia, Via Campi 213/A, I-41125, Modena, Italy
| | - Paolo Facci
- CNR-Ibf, Via De Marini 6, I-16149, Genova, Italy
| | - Andrea Alessandrini
- CNR-Nanoscience Institute-S3, Via Campi 213/A, I-41125, Modena, Italy
- Department of Physics, Informatics and Mathematics, University of Modena and Reggio Emilia, Via Campi 213/A, I-41125, Modena, Italy
| |
Collapse
|
26
|
Nakamuro T, Kamei K, Sun K, Bode JW, Harano K, Nakamura E. Time-Resolved Atomistic Imaging and Statistical Analysis of Daptomycin Oligomers with and without Calcium Ions. J Am Chem Soc 2022; 144:13612-13622. [PMID: 35857028 DOI: 10.1021/jacs.2c03949] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Daptomycin (DP) is effective against multiple drug-resistant Gram-positive pathogens because of its distinct mechanism of action. An accepted mechanism includes Ca2+-triggered aggregation of the DP molecule to form oligomers. DP and its oligomers have so far defied structural analysis at a molecular level. We studied the ability of DP molecule to aggregate by itself in water, the effects of Ca2+ ions to promote the aggregation, and the connectivity of the DP molecules in the oligomers by the combined use of dynamic light scattering in water and atomic-resolution cinematographic imaging of DP molecules captured on a carbon nanotube on which the DP molecule is installed as a fishhook. We found that the DP molecule aggregates weakly into dimers, trimers, and tetramers in water, and strongly in the presence of calcium ions, and that the tetramer is the largest oligomer in homogeneous aqueous solution. The dimer remains as the major species, and we propose a face-to-face stacked structure based on dynamic imaging using millisecond and angstrom resolution transmission electron microscopy. The tetramer in its cyclic form is the largest oligomer observed, while the trimer forms in its linear form. The study has shown that the DP molecule has an intrinsic property of forming tetramers in water, which is enhanced by the presence of calcium ions. Such experimental structural information will serve as a platform for future drug design. The data also illustrate the utility of cinematographic recording for the study of self-organization processes.
Collapse
Affiliation(s)
- Takayuki Nakamuro
- Department of Chemistry, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Ko Kamei
- Department of Chemistry, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Keyi Sun
- Department of Chemistry, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Jeffrey W Bode
- Laboratorium für Organische Chemie, Department of Chemistry and Applied Biosciences, ETH Zürich, Zürich 8093, Switzerland
| | - Koji Harano
- Department of Chemistry, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Eiichi Nakamura
- Department of Chemistry, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
27
|
Moreira R, Taylor SD. Establishing the Structure-Activity Relationship between Phosphatidylglycerol and Daptomycin. ACS Infect Dis 2022; 8:1674-1686. [PMID: 35793519 DOI: 10.1021/acsinfecdis.2c00262] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Daptomycin is a clinical antibiotic used to treat serious infections caused by Gram-positive bacteria. Although there is debate about the action mechanism of daptomycin, it is known that daptomycin requires both calcium and phosphatidylglycerol (PG) to exert its antibacterial effect. Despite the importance and uniqueness of the interaction of daptomycin with PG, very little is known about this interaction or the nascent daptomycin-PG complex. In this work, we establish a structure-activity relationship between daptomycin and PG through the synthesis of PG analogues. In total, nine PGs were synthesized using a divergent approach employing phosphoramidite chemistry. The interaction between daptomycin and these PGs was studied using fluorescence, circular dichroism, and isothermal titration calorimetry. It was determined that daptomycin is highly sensitive to the modification of the headgroup of PG and both hydroxyl groups influence membrane binding, oligomerization, and backbone structure. Methylation of each hydroxyl in the headgroup suggests that the binding pocket envelops both hydroxyl groups. A PG acyl tail chain length of at least 7-8 carbons is required for stoichiometric binding at micromolar peptide concentrations. Daptomycin binds to PG having 8-carbon, linear, unsaturated acyl groups (C8PGs) at the micromolar concentration and interacts with C8PG in essentially the same manner as when the PG is incorporated into a liposome, and thus, preassembly of individual PG moieties is not a prerequisite for binding, structural transition, and oligomerization.
Collapse
Affiliation(s)
- Ryan Moreira
- Department of Chemistry, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada
| | - Scott D Taylor
- Department of Chemistry, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada
| |
Collapse
|
28
|
van de Lagemaat M, Stockbroekx V, Geertsema-Doornbusch GI, Dijk M, Carniello V, Woudstra W, van der Mei HC, Busscher HJ, Ren Y. A Comparison of the Adaptive Response of Staphylococcus aureus vs. Streptococcus mutans and the Development of Chlorhexidine Resistance. Front Microbiol 2022; 13:861890. [PMID: 35694293 PMCID: PMC9186159 DOI: 10.3389/fmicb.2022.861890] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 04/11/2022] [Indexed: 11/14/2022] Open
Abstract
Antimicrobials with nonselective antibacterial efficacy such as chlorhexidine can be effective in reducing biofilm, but bear the risk of inducing resistance in specific bacteria. In clinical practice, bacteria such as Staphylococcus aureus have been found resistant to chlorhexidine, but other bacteria, including Streptococcus mutans, have largely remained susceptible to chlorhexidine despite its widespread use in oral healthcare. Here, we aim to forward a possible reason as to why S. aureus can acquire resistance against chlorhexidine, while S. mutans remains susceptible to chlorhexidine. Measurement of surface-enhanced fluorescence indicated that chlorhexidine caused gradual, but irreversible deformation to adhering green fluorescent S. aureus due to irreparable damage to the cell wall. Concurrently, the metabolic activity of adhering staphylococci was higher than of planktonic bacteria, suggesting efflux mechanisms may have been activated upon cell wall deformation, impeding the buildup of a high chlorhexidine concentration in the cytoplasm and therewith stimulating the development of chlorhexidine resistance in S. aureus. Exposure of S. mutans to chlorhexidine caused immediate, but reversible deformation in adhering streptococci, indicative of rapid self-repair of cell wall damage done by chlorhexidine. Due to cell wall self-repair, S. mutans will be unable to effectively reduce the chlorhexidine concentration in the cytoplasm causing solidification of the cytoplasm. In line, no increased metabolic activity was observed in S. mutans during exposure to chlorhexidine. Therewith, self-repair is suicidal and prevents the development of a chlorhexidine-resistant progeny in S. mutans.
Collapse
Affiliation(s)
- Marieke van de Lagemaat
- University of Groningen and University Medical Center Groningen, Department of Orthodontics, Groningen, Netherlands
| | - Valerie Stockbroekx
- University of Groningen and University Medical Center Groningen, Department of Orthodontics, Groningen, Netherlands
| | - Gésinda I. Geertsema-Doornbusch
- University of Groningen and University Medical Center Groningen, Department of Biomedical Engineering, Groningen, Netherlands
| | - Melissa Dijk
- University of Groningen and University Medical Center Groningen, Department of Orthodontics, Groningen, Netherlands
| | - Vera Carniello
- University of Groningen and University Medical Center Groningen, Department of Biomedical Engineering, Groningen, Netherlands
| | - Willem Woudstra
- University of Groningen and University Medical Center Groningen, Department of Biomedical Engineering, Groningen, Netherlands
| | - Henny C. van der Mei
- University of Groningen and University Medical Center Groningen, Department of Biomedical Engineering, Groningen, Netherlands
- *Correspondence: Henny C. van der Mei,
| | - Henk J. Busscher
- University of Groningen and University Medical Center Groningen, Department of Biomedical Engineering, Groningen, Netherlands
| | - Yijin Ren
- University of Groningen and University Medical Center Groningen, Department of Orthodontics, Groningen, Netherlands
| |
Collapse
|
29
|
Simpson JD, Ray A, Koehler M, Mohammed D, Alsteens D. Atomic force microscopy applied to interrogate nanoscale cellular chemistry and supramolecular bond dynamics for biomedical applications. Chem Commun (Camb) 2022; 58:5072-5087. [PMID: 35315846 DOI: 10.1039/d1cc07200e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Understanding biological interactions at a molecular level grants valuable information relevant to improving medical treatments and outcomes. Among the suite of technologies available, Atomic Force Microscopy (AFM) is unique in its ability to quantitatively probe forces and receptor-ligand interactions in real-time. The ability to assess the formation of supramolecular bonds and intermediates in real-time on surfaces and living cells generates important information relevant to understanding biological phenomena. Combining AFM with fluorescence-based techniques allows for an unprecedented level of insight not only concerning the formation and rupture of bonds, but understanding medically relevant interactions at a molecular level. As the ability of AFM to probe cells and more complex models improves, being able to assess binding kinetics, chemical topographies, and garner spectroscopic information will likely become key to developing further improvements in fields such as cancer, nanomaterials, and virology. The rapid response to the COVID-19 crisis, producing information regarding not just receptor affinities, but also strain-dependent efficacy of neutralizing nanobodies, demonstrates just how viable and integral to the pre-clinical development of information AFM techniques are in this era of medicine.
Collapse
Affiliation(s)
- Joshua D Simpson
- Louvain Institute of Biomolecular Science and Technology, Université Catholique de Louvain, Louvain-la-Neuve 1348, Belgium.
| | - Ankita Ray
- Louvain Institute of Biomolecular Science and Technology, Université Catholique de Louvain, Louvain-la-Neuve 1348, Belgium.
| | - Melanie Koehler
- Louvain Institute of Biomolecular Science and Technology, Université Catholique de Louvain, Louvain-la-Neuve 1348, Belgium.
| | - Danahe Mohammed
- Louvain Institute of Biomolecular Science and Technology, Université Catholique de Louvain, Louvain-la-Neuve 1348, Belgium.
| | - David Alsteens
- Louvain Institute of Biomolecular Science and Technology, Université Catholique de Louvain, Louvain-la-Neuve 1348, Belgium.
| |
Collapse
|
30
|
Barnawi G, Noden M, Goodyear J, Marlyn J, Schneider O, Beriashvili D, Schulz S, Moreira R, Palmer M, Taylor SD. Discovery of Highly Active Derivatives of Daptomycin by Assessing the Effect of Amino Acid Substitutions at Positions 8 and 11 on a Daptomycin Analogue. ACS Infect Dis 2022; 8:778-789. [PMID: 35317552 DOI: 10.1021/acsinfecdis.1c00483] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Daptomycin is an important antibiotic used for treating serious infections caused by Gram-positive bacteria including methicillin-resistant Staphylococcus aureus (MRSA) and vancomycin-resistant enterococci. Establishing structure-activity relationships of daptomycin is important for developing new daptomycin-based antibiotics with expanded clinical applications and for tackling the ever-increasing problem of antimicrobial resistance. Toward this end, Dap-K6-E12-W13, an active analogue of daptomycin in which the uncommon amino acids in daptomycin are replaced with their common counterparts, was used as a model system for studying the effect of amino acid variation at positions 8 and 11 on in vitro biological activity against a model organism, Bacillus subtilis, and calcium-dependent insertion into model membranes. None of the new peptides were more active than Dap-K6-E12-W13; however, substitution at positions 8 and/or 11 with cationic residues resulted in little or no loss of activity, and some of these analogues were able to insert into model membranes at lower calcium ion concentrations than the parent peptide. Incorporation of these cationic residues into positions 8 and/or 11 of daptomycin itself yielded some derivatives that exhibited lower minimum inhibitory concentrations than daptomycin against B. subtilis 1046 as well as comparable and sometimes superior activity against clinical isolates of MRSA.
Collapse
Affiliation(s)
- Ghufran Barnawi
- Department of Chemistry, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada
| | - Michael Noden
- Department of Chemistry, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada
| | - Jeremy Goodyear
- Department of Chemistry, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada
| | - Julian Marlyn
- Department of Chemistry, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada
| | - Olivia Schneider
- Department of Chemistry, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada
| | - David Beriashvili
- Department of Chemistry, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada
| | - Sarah Schulz
- Department of Chemistry, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada
| | - Ryan Moreira
- Department of Chemistry, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada
| | - Michael Palmer
- Department of Chemistry, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada
| | - Scott D. Taylor
- Department of Chemistry, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada
| |
Collapse
|
31
|
Moreira R, Taylor SD. The Chiral Target of Daptomycin Is the 2
R
,2′
S
Stereoisomer of Phosphatidylglycerol. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202114858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Ryan Moreira
- Deptartment of Chemistry University of Waterloo 200 University Ave. West Waterloo Ontario Canada
| | - Scott D. Taylor
- Deptartment of Chemistry University of Waterloo 200 University Ave. West Waterloo Ontario Canada
| |
Collapse
|
32
|
Moreira R, Taylor SD. The Chiral Target of Daptomycin Is the 2R,2'S Stereoisomer of Phosphatidylglycerol. Angew Chem Int Ed Engl 2021; 61:e202114858. [PMID: 34843157 DOI: 10.1002/anie.202114858] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Indexed: 12/12/2022]
Abstract
Daptomycin (dap) is an important antibiotic that interacts with the bacterial membrane lipid phosphatidylglycerol (PG) in a calcium-dependent manner. The enantiomer of dap (ent-dap) was synthesized and was found to be 85-fold less active than dap against B. subtilis, indicating that dap interacts with a chiral target as part of its mechanism of action. Using liposomes containing enantiopure PG, we demonstrate that the binding of dap to PG, the structural transition that occurs upon dap binding to PG, and the subsequent oligomerization of dap, depends upon the configuration of PG, and that dap prefers the 1,2-diacyl-sn-glycero-3-phospho-1'-sn-glycerol stereoisomer (2R,2'S configuration). Ent-dap has a lower affinity for 2R,2'S liposomes than dap and cannot oligomerize to the same extent as dap, which accounts for why ent-dap is less active than dap. To our knowledge, this is the first example whereby the activity of an antibiotic depends upon the configuration of a lipid head group.
Collapse
Affiliation(s)
- Ryan Moreira
- Deptartment of Chemistry, University of Waterloo, 200 University Ave. West, Waterloo, Ontario, Canada
| | - Scott D Taylor
- Deptartment of Chemistry, University of Waterloo, 200 University Ave. West, Waterloo, Ontario, Canada
| |
Collapse
|
33
|
Ji CH, Kim H, Je HW, Kwon H, Lee D, Kang HS. Top-down synthetic biology approach for titer improvement of clinically important antibiotic daptomycin in Streptomyces roseosporus. Metab Eng 2021; 69:40-49. [PMID: 34737068 DOI: 10.1016/j.ymben.2021.10.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/09/2021] [Accepted: 10/29/2021] [Indexed: 12/21/2022]
Abstract
Secondary metabolites are produced at low titers by native producers due to tight regulations of their productions in response to environmental conditions. Synthetic biology provides a rational engineering principle for transcriptional optimization of secondary metabolite BGCs (biosynthetic gene clusters). Here, we demonstrate the use of synthetic biology principles for the development of a high-titer strain of the clinically important antibiotic daptomycin. Due to the presence of large NRPS (non-ribosomal peptide synthetase) genes with multiple direct repeats, we employed a top-down approach that allows transcriptional optimization of genes in daptomycin BGC with the minimum inputs of synthetic DNAs. The repeat-free daptomycin BGC was created through partial codon-reprogramming of a NRPS gene and cloned into a shuttle BAC vector, allowing BGC refactoring in a host with a powerful recombination system. Then, transcriptions of functionally divided operons were sequentially optimized through three rounds of DBTL (design-build-test-learn) cycles that resulted in up to ~2300% improvement in total lipopeptide titers compared to the wild-type strain. Upon decanoic acid feeding, daptomycin accounted for ∼ 40% of total lipopeptide production. To the best of our knowledge, this is the highest improvement of daptomycin titer ever achieved through genetic engineering of S. roseosporus. The top-down engineering approach we describe here could be used as a general strategy for the development of high-titer industrial strains of secondary metabolites produced by BGCs containing genes of large multi-modular NRPS and PKS enzymes.
Collapse
Affiliation(s)
- Chang-Hun Ji
- Department of Biomedical Science and Engineering, Konkuk University, Seoul, 05029, Republic of Korea
| | - Hiyoung Kim
- Department of Biomedical Science and Engineering, Konkuk University, Seoul, 05029, Republic of Korea
| | - Hyun-Woo Je
- Department of Biomedical Science and Engineering, Konkuk University, Seoul, 05029, Republic of Korea
| | - Haeun Kwon
- Department of Plant Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Dongho Lee
- Department of Plant Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Hahk-Soo Kang
- Department of Biomedical Science and Engineering, Konkuk University, Seoul, 05029, Republic of Korea.
| |
Collapse
|
34
|
Kotsogianni I, Wood TM, Alexander FM, Cochrane SA, Martin NI. Binding Studies Reveal Phospholipid Specificity and Its Role in the Calcium-Dependent Mechanism of Action of Daptomycin. ACS Infect Dis 2021; 7:2612-2619. [PMID: 34406007 PMCID: PMC8438661 DOI: 10.1021/acsinfecdis.1c00316] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Multidrug-resistant bacteria pose a serious global health threat as antibiotics are increasingly losing their clinical efficacy. A molecular level understanding of the mechanism of action of antimicrobials plays a key role in developing new agents to combat the threat of antimicrobial resistance. Daptomycin, the only clinically used calcium-dependent lipopeptide antibiotic, selectively disrupts Gram-positive bacterial membranes to illicit its bactericidal effect. In this study, we use isothermal titration calorimetry to further characterize the structural features of the target bacterial phospholipids that drive daptomycin binding. Our studies reveal that daptomycin shows a clear preference for the phosphoglycerol headgroup. Furthermore, unlike other calcium-dependent lipopeptide antibiotics, calcium binding by daptomycin is strongly dependent on the presence of phosphatidylglycerol. These investigations provide new insights into daptomycin's phospholipid specificity and calcium binding behavior.
Collapse
Affiliation(s)
- Ioli Kotsogianni
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, Sylviusweg 72, 2333 BE Leiden, The Netherlands
| | - Thomas M. Wood
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, Sylviusweg 72, 2333 BE Leiden, The Netherlands
| | - Francesca M. Alexander
- School of Chemistry and Chemical Engineering, David Keir Building, Stranmillis Road, Queen’s University Belfast, Belfast, BT9 5AG, United Kingdom
| | - Stephen A. Cochrane
- School of Chemistry and Chemical Engineering, David Keir Building, Stranmillis Road, Queen’s University Belfast, Belfast, BT9 5AG, United Kingdom
| | - Nathaniel I. Martin
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, Sylviusweg 72, 2333 BE Leiden, The Netherlands
| |
Collapse
|
35
|
Zhao X, Meng X, Ragauskas AJ, Lai C, Ling Z, Huang C, Yong Q. Unlocking the secret of lignin-enzyme interactions: Recent advances in developing state-of-the-art analytical techniques. Biotechnol Adv 2021; 54:107830. [PMID: 34480987 DOI: 10.1016/j.biotechadv.2021.107830] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 08/07/2021] [Accepted: 08/29/2021] [Indexed: 02/08/2023]
Abstract
Bioconversion of renewable lignocellulosics to produce liquid fuels and chemicals is one of the most effective ways to solve the problem of fossil resource shortage, energy security, and environmental challenges. Among the many biorefinery pathways, hydrolysis of lignocellulosics to fermentable monosaccharides by cellulase is arguably the most critical step of lignocellulose bioconversion. In the process of enzymatic hydrolysis, the direct physical contact between enzymes and cellulose is an essential prerequisite for the hydrolysis to occur. However, lignin is considered one of the most recalcitrant factors hindering the accessibility of cellulose by binding to cellulase unproductively, which reduces the saccharification rate and yield of sugars. This results in high costs for the saccharification of carbohydrates. The various interactions between enzymes and lignin have been explored from different perspectives in literature, and a basic lignin inhibition mechanism has been proposed. However, the exact interaction between lignin and enzyme as well as the recently reported promotion of some types of lignin on enzymatic hydrolysis is still unclear at the molecular level. Multiple analytical techniques have been developed, and fully unlocking the secret of lignin-enzyme interactions would require a continuous improvement of the currently available analytical techniques. This review summarizes the current commonly used advanced research analytical techniques for investigating the interaction between lignin and enzyme, including quartz crystal microbalance with dissipation (QCM-D), surface plasmon resonance (SPR), attenuated total reflectance-Fourier transform infrared (ATR-FTIR) spectroscopy, atomic force microscopy (AFM), nuclear magnetic resonance (NMR) spectroscopy, fluorescence spectroscopy (FLS), and molecular dynamics (MD) simulations. Interdisciplinary integration of these analytical methods is pursued to provide new insight into the interactions between lignin and enzymes. This review will serve as a resource for future research seeking to develop new methodologies for a better understanding of the basic mechanism of lignin-enzyme binding during the critical hydrolysis process.
Collapse
Affiliation(s)
- Xiaoxue Zhao
- Co-Innovation Center for Efficient Processing and Utilization of Forest Resources, Department of Bioengineering, Nanjing Forestry University, Nanjing 210037, China
| | - Xianzhi Meng
- Department of Chemical and Biomolecular Engineering, University of Tennessee, Knoxville, TN 37996, USA
| | - Arthur J Ragauskas
- Department of Chemical and Biomolecular Engineering, University of Tennessee, Knoxville, TN 37996, USA; Center for Renewable Carbon, Department of Forestry, Wildlife and Fisheries, University of Tennessee, Knoxville, TN 37996, USA; Joint Institute for Biological Sciences, Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, TN 37831, USA
| | - Chenhuan Lai
- Co-Innovation Center for Efficient Processing and Utilization of Forest Resources, Department of Bioengineering, Nanjing Forestry University, Nanjing 210037, China
| | - Zhe Ling
- Co-Innovation Center for Efficient Processing and Utilization of Forest Resources, Department of Bioengineering, Nanjing Forestry University, Nanjing 210037, China
| | - Caoxing Huang
- Co-Innovation Center for Efficient Processing and Utilization of Forest Resources, Department of Bioengineering, Nanjing Forestry University, Nanjing 210037, China.
| | - Qiang Yong
- Co-Innovation Center for Efficient Processing and Utilization of Forest Resources, Department of Bioengineering, Nanjing Forestry University, Nanjing 210037, China.
| |
Collapse
|
36
|
Látrová K, Havlová N, Večeřová R, Pinkas D, Bogdanová K, Kolář M, Fišer R, Konopásek I, Do Pham DD, Rejman D, Mikušová G. Outer membrane and phospholipid composition of the target membrane affect the antimicrobial potential of first- and second-generation lipophosphonoxins. Sci Rep 2021; 11:10446. [PMID: 34001940 PMCID: PMC8129119 DOI: 10.1038/s41598-021-89883-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 04/27/2021] [Indexed: 02/03/2023] Open
Abstract
Lipophosphonoxins (LPPOs) are small modular synthetic antibacterial compounds that target the cytoplasmic membrane. First-generation LPPOs (LPPO I) exhibit an antimicrobial activity against Gram-positive bacteria; however they do not exhibit any activity against Gram-negatives. Second-generation LPPOs (LPPO II) also exhibit broadened activity against Gram-negatives. We investigated the reasons behind this different susceptibility of bacteria to the two generations of LPPOs using model membranes and the living model bacteria Bacillus subtilis and Escherichia coli. We show that both generations of LPPOs form oligomeric conductive pores and permeabilize the bacterial membrane of sensitive cells. LPPO activity is not affected by the value of the target membrane potential, and thus they are also active against persister cells. The insensitivity of Gram-negative bacteria to LPPO I is probably caused by the barrier function of the outer membrane with LPS. LPPO I is almost incapable of overcoming the outer membrane in living cells, and the presence of LPS in liposomes substantially reduces their activity. Further, the antimicrobial activity of LPPO is also influenced by the phospholipid composition of the target membrane. A higher proportion of phospholipids with neutral charge such as phosphatidylethanolamine or phosphatidylcholine reduces the LPPO permeabilizing potential.
Collapse
Affiliation(s)
- Klára Látrová
- Department of Genetics and Microbiology, Faculty of Science, Charles University, Viničná 5, 128 00, Prague 2, Czech Republic
| | - Noemi Havlová
- Department of Genetics and Microbiology, Faculty of Science, Charles University, Viničná 5, 128 00, Prague 2, Czech Republic
| | - Renata Večeřová
- Department of Microbiology, Faculty of Medicine and Dentistry, Palacký University Olomouc, Hněvotínská 3, 775 15, Olomouc, Czech Republic
| | - Dominik Pinkas
- Department of Genetics and Microbiology, Faculty of Science, Charles University, Viničná 5, 128 00, Prague 2, Czech Republic
| | - Kateřina Bogdanová
- Department of Microbiology, Faculty of Medicine and Dentistry, Palacký University Olomouc, Hněvotínská 3, 775 15, Olomouc, Czech Republic
| | - Milan Kolář
- Department of Microbiology, Faculty of Medicine and Dentistry, Palacký University Olomouc, Hněvotínská 3, 775 15, Olomouc, Czech Republic
| | - Radovan Fišer
- Department of Genetics and Microbiology, Faculty of Science, Charles University, Viničná 5, 128 00, Prague 2, Czech Republic
| | - Ivo Konopásek
- Department of Genetics and Microbiology, Faculty of Science, Charles University, Viničná 5, 128 00, Prague 2, Czech Republic
| | - Duy Dinh Do Pham
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences v.v.i., Flemingovo nám. 2, 166 10, Prague 6, Czech Republic
| | - Dominik Rejman
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences v.v.i., Flemingovo nám. 2, 166 10, Prague 6, Czech Republic.
| | - Gabriela Mikušová
- Department of Genetics and Microbiology, Faculty of Science, Charles University, Viničná 5, 128 00, Prague 2, Czech Republic.
| |
Collapse
|