1
|
Li C, Wu Y, Bao S, Li H, Xu Z, Yan J, Yu X, He L, Zhang T, Liu W, Hou S, Zhang Y, Xu J, Yan T, Wang T, Yan Y, Liu J. Photo-Switchable Supramolecular Interactions Regulate K + Transmembrane Transport and Cancer Cell Apoptosis. J Am Chem Soc 2025; 147:14139-14153. [PMID: 40111177 DOI: 10.1021/jacs.4c14583] [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: 03/22/2025]
Abstract
Natural channel proteins (NCPs) have numerous ion transport modes, but it remains a big challenge to replicate this trait by artificial ion transport systems. Herein, we present an azobenzene-incorporated single-chain random heteropolymers (RHPs)-derived biomimetic K+ channel P3, which can switch between three ion transport states ("ON," "Partially OFF," and "Totally OFF") in both liposomes and cancer cells. The conformational adjustments of P3 activated by light-modulating two groups of supramolecular interactions ((1) hydrogen bonding and π-π interactions; (2) host-guest interactions) realize these switches, resembling the protein mechanisms that govern activity. Underlying molecular mechanisms are the photoisomerization of azobenzene moieties in P3 and their complexation with β-cyclodextrin (β-CD), enabling the exploit of a "one stone (azobenzene moiety), two birds (supramolecular interactions)" strategy. Mechanistic investigations demonstrate that P3-induced substantial K+ efflux (a 50% drop within just 4 min) causes endoplasmic reticulum (ER) stress, intriguing Ca2+ sparks, enhanced reactive oxygen species (ROS), and finally severe mitochondria-dependent apoptosis. This NCP-like channel (P3) is expected to provide new opportunities for a deeper understanding of the internal mechanisms of NCPs, as well as for treating cancer and other diseases.
Collapse
Affiliation(s)
- Cong Li
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Hangzhou Normal University, Hangzhou 311121, China
- College of Chemistry and Chemical Engineering, Key Laboratory of Special Functional and Smart Polymer Materials of Ministry of Industry and Information Technology, Northwestern Polytechnical University, Xi'an 710129, China
| | - Yaqi Wu
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Hangzhou Normal University, Hangzhou 311121, China
- College of Chemistry and Chemical Engineering, Key Laboratory of Special Functional and Smart Polymer Materials of Ministry of Industry and Information Technology, Northwestern Polytechnical University, Xi'an 710129, China
| | - Sheng Bao
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Hangzhou Normal University, Hangzhou 311121, China
| | - Hui Li
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Hangzhou Normal University, Hangzhou 311121, China
| | - Zhengwei Xu
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Hangzhou Normal University, Hangzhou 311121, China
| | - Jing Yan
- College of Chemistry and Chemical Engineering, Key Laboratory of Special Functional and Smart Polymer Materials of Ministry of Industry and Information Technology, Northwestern Polytechnical University, Xi'an 710129, China
| | - Xiaoxuan Yu
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Hangzhou Normal University, Hangzhou 311121, China
| | - Lei He
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Hangzhou Normal University, Hangzhou 311121, China
| | - Tianlong Zhang
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Hangzhou Normal University, Hangzhou 311121, China
| | - Wang Liu
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Hangzhou Normal University, Hangzhou 311121, China
| | - Shida Hou
- College of Chemistry and Chemical Engineering, Key Laboratory of Special Functional and Smart Polymer Materials of Ministry of Industry and Information Technology, Northwestern Polytechnical University, Xi'an 710129, China
| | - Yang Zhang
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Hangzhou Normal University, Hangzhou 311121, China
| | - Jiayun Xu
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Hangzhou Normal University, Hangzhou 311121, China
| | - Tengfei Yan
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Hangzhou Normal University, Hangzhou 311121, China
| | - Tingting Wang
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Hangzhou Normal University, Hangzhou 311121, China
| | - Yi Yan
- College of Chemistry and Chemical Engineering, Key Laboratory of Special Functional and Smart Polymer Materials of Ministry of Industry and Information Technology, Northwestern Polytechnical University, Xi'an 710129, China
| | - Junqiu Liu
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Hangzhou Normal University, Hangzhou 311121, China
- College of Chemistry and Chemical Engineering, Key Laboratory of Special Functional and Smart Polymer Materials of Ministry of Industry and Information Technology, Northwestern Polytechnical University, Xi'an 710129, China
| |
Collapse
|
2
|
Zan B, Ulmschneider MB, Ulmschneider JP. The difference between MelP5 and melittin membrane poration. Sci Rep 2025; 15:7442. [PMID: 40033017 PMCID: PMC11876596 DOI: 10.1038/s41598-025-91951-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 02/21/2025] [Indexed: 03/05/2025] Open
Abstract
Melittin, a natural peptide found in bees, has been shown to induce pore formation in cell membranes. However, its artificial mutant, MelP5 can do so at concentrations 200 times lower than melittin. The mechanism of the enhanced portion ability is not fully understood. By conducting all-atom molecular dynamics (MD) simulations, we found that MelP5 forms a stable pore that is macro-molecular sized. Our results suggest that the mutation of five amino acids from melittin reduces the electrostatic repulsion between peptides and strengthens hydrophobic interactions between MelP5 and lipid tails, resulting in the formation of a stable and larger pore. Furthermore, we found that cholesterol (CHOL), which occupies 30% in mammalian cell membranes, plays a crucial role in enhancing the pore formation of MelP5. As the amount of CHOL increases, the pore becomes larger, more stable, and forms more quickly. The presence of CHOL also promotes the formation of oligomers, which further support the pore. Our findings indicate that CHOL promotes the insertion of peptides into the membrane and reduces the amount of surface state peptides, thereby stabilizing the pore. These results highlight the important role of CHOL in membrane permeabilization by MelP5 and provide new insights into the mechanism of action of membrane-active antimicrobial peptides.
Collapse
Affiliation(s)
- Bing Zan
- School of Physics and Astronomy, Shanghai Jiao Tong University, Shanghai, China
- Department of Chemistry, King's College London, London, UK
- Institute of Natural Sciences, Shanghai Jiao Tong University, Shanghai, China
| | | | - Jakob P Ulmschneider
- School of Physics and Astronomy, Shanghai Jiao Tong University, Shanghai, China.
- Institute of Natural Sciences, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
3
|
Juhaniewicz-Debinska J. Melittin-Induced Structural Transformations in DMPG and DMPS Lipid Membranes: A Langmuir Monolayer and AFM Study. Molecules 2024; 29:6064. [PMID: 39770152 PMCID: PMC11677270 DOI: 10.3390/molecules29246064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/20/2024] [Accepted: 12/21/2024] [Indexed: 01/11/2025] Open
Abstract
In this study, we explore the interactions between melittin, a cationic antimicrobial peptide, and model lipid membranes composed of the negatively charged phospholipids 1,2-dimyristoyl-sn-glycero-3-phosphoglycerol (DMPG) and 1,2-dimyristoyl-sn-glycero-3-phosphoserine (DMPS). Using the Langmuir monolayer technique and atomic force microscopy (AFM), we reveal novel insights into these interactions. Our key finding is the observation of the ripple phase in the DMPS bilayer on mica, a phenomenon not previously reported for negatively charged single bilayers. This discovery is significant given the critical role of phosphatidylserine (PS) in cancer biology and the potential of melittin as an anticancer agent. We also highlight the importance of subphase composition, as melittin interacts preferentially with lipids in the liquid-condensed phase; thus, selecting the appropriate subphase composition is crucial because it affects lipid behavior and consequently melittin interactions. Our results show that melittin incorporates into lipid monolayers in both liquid-expanded and liquid-condensed phases, enhancing membrane fluidity and disorder, but is expelled from DMPS in the solid phase. AFM imaging further reveals that melittin induces substantial structural changes in the DMPG membrane and forms the ripple phase in the DMPS bilayers. Despite these alterations, melittin does not cause pore formation or membrane rupture, suggesting strong electrostatic adsorption on the membrane surface that prevents penetration. These findings highlight the differential impacts of melittin on lipid monolayers and bilayers and underscore its potential for interacting with membranes without causing disruption.
Collapse
Affiliation(s)
- Joanna Juhaniewicz-Debinska
- Faculty of Chemistry, Biological and Chemical Research Centre, University of Warsaw, ul. Zwirki i Wigury 101, 02-089 Warsaw, Poland
| |
Collapse
|
4
|
Volovik MV, Batishchev OV. Membrane Activity of Melittin and Magainin-I at Low Peptide-to-Lipid Ratio: Different Types of Pores and Translocation Mechanisms. Biomolecules 2024; 14:1118. [PMID: 39334885 PMCID: PMC11430820 DOI: 10.3390/biom14091118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/01/2024] [Accepted: 09/03/2024] [Indexed: 09/30/2024] Open
Abstract
Antimicrobial peptides (AMPs) are believed to be a prominent alternative to the common antibiotics. However, despite decades of research, there are still no good clinical examples of peptide-based antimicrobial drugs for system application. The main reasons are loss of activity in the human body, cytotoxicity, and low selectivity. To overcome these challenges, a well-established structure-function relationship for AMPs is critical. In the present study, we focused on the well-known examples of melittin and magainin to investigate in detail the initial stages of AMP interaction with lipid membranes at low peptide-to-lipid ratio. By combining the patch-clamp technique with the bioelectrochemical method of intramembrane field compensation, we showed that these peptides interact with the membrane in different ways: melittin inserts deeper into the lipid bilayer than magainin. This difference led to diversity in pore formation. While magainin, after a threshold concentration, formed the well-known toroidal pores, allowing the translocation of the peptide through the membrane, melittin probably induced predominantly pure lipidic pores with a very low rate of peptide translocation. Thus, our results shed light on the early stages of peptide-membrane interactions and suggest new insights into the structure-function relationship of AMPs based on the depth of their membrane insertion.
Collapse
Affiliation(s)
- Marta V Volovik
- Laboratory of Bioelectrochemistry, A.N. Frumkin Institute of Physical Chemistry and Electrochemistry, Russian Academy of Sciences, 31/4 Leninskiy Prospekt, 119071 Moscow, Russia
| | - Oleg V Batishchev
- Laboratory of Bioelectrochemistry, A.N. Frumkin Institute of Physical Chemistry and Electrochemistry, Russian Academy of Sciences, 31/4 Leninskiy Prospekt, 119071 Moscow, Russia
| |
Collapse
|
5
|
Lund PM, Kristensen K, Larsen NW, Knuhtsen A, Hansen MB, Hjørringgaard CU, Eriksen AZ, Urquhart AJ, Mortensen KI, Simonsen JB, Andresen TL, Larsen JB. Tuning the double lipidation of salmon calcitonin to introduce a pore-like membrane translocation mechanism. J Colloid Interface Sci 2024; 669:198-210. [PMID: 38713958 DOI: 10.1016/j.jcis.2024.04.093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 04/10/2024] [Accepted: 04/14/2024] [Indexed: 05/09/2024]
Abstract
A widespread strategy to increase the transport of therapeutic peptides across cellular membranes has been to attach lipid moieties to the peptide backbone (lipidation) to enhance their intrinsic membrane interaction. Efforts in vitro and in vivo investigating the correlation between lipidation characteristics and peptide membrane translocation efficiency have traditionally relied on end-point read-out assays and trial-and-error-based optimization strategies. Consequently, the molecular details of how therapeutic peptide lipidation affects it's membrane permeation and translocation mechanisms remain unresolved. Here we employed salmon calcitonin as a model therapeutic peptide and synthesized nine double lipidated analogs with varying lipid chain lengths. We used single giant unilamellar vesicle (GUV) calcein influx time-lapse fluorescence microscopy to determine how tuning the lipidation length can lead to an All-or-None GUV filling mechanism, indicative of a peptide mediated pore formation. Finally, we used a GUVs-containing-inner-GUVs assay to demonstrate that only peptide analogs capable of inducing pore formation show efficient membrane translocation. Our data provided the first mechanistic details on how therapeutic peptide lipidation affects their membrane perturbation mechanism and demonstrated that fine-tuning lipidation parameters could induce an intrinsic pore-forming capability. These insights and the microscopy based workflow introduced for investigating structure-function relations could be pivotal for optimizing future peptide design strategies.
Collapse
Affiliation(s)
- Philip M Lund
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, Lyngby, Denmark; DTU Health Tech, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Kasper Kristensen
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, Lyngby, Denmark; DTU Health Tech, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Nanna W Larsen
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, Lyngby, Denmark; DTU Health Tech, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Astrid Knuhtsen
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, Lyngby, Denmark; DTU Health Tech, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Morten B Hansen
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, Lyngby, Denmark; DTU Health Tech, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Claudia U Hjørringgaard
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, Lyngby, Denmark; DTU Health Tech, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Anne Z Eriksen
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, Lyngby, Denmark; DTU Health Tech, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Andrew J Urquhart
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, Lyngby, Denmark; DTU Health Tech, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Kim I Mortensen
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, Lyngby, Denmark; DTU Health Tech, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Jens B Simonsen
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, Lyngby, Denmark; DTU Health Tech, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Thomas L Andresen
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, Lyngby, Denmark; DTU Health Tech, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark.
| | - Jannik B Larsen
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, Lyngby, Denmark; DTU Health Tech, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark.
| |
Collapse
|
6
|
Ulmschneider JP, Ulmschneider MB. Melittin can permeabilize membranes via large transient pores. Nat Commun 2024; 15:7281. [PMID: 39179607 PMCID: PMC11343860 DOI: 10.1038/s41467-024-51691-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 08/15/2024] [Indexed: 08/26/2024] Open
Abstract
Membrane active peptides are known to porate lipid bilayers, but their exact permeabilization mechanism and the structure of the nanoaggregates they form in membranes have often been difficult to determine experimentally. For many sequences at lower peptide concentrations, transient leakage is observed in experiments, suggesting the existence of transient pores. For two well-know peptides, alamethicin and melittin, we show here that molecular mechanics simulations i) can directly distinguish equilibrium poration and non-equilibrium transient leakage processes, and ii) can be used to observe the detailed pore structures and mechanism of permeabilization in both cases. Our results are in very high agreement with numerous experimental evidence for these two peptides. This suggests that molecular simulations can capture key membrane poration phenomena directly and in the future may develop to be a useful tool that can assist experimental peptide design.
Collapse
Affiliation(s)
- Jakob P Ulmschneider
- Institute of Natural Sciences and School of Physics and Astronomy, Shanghai Jiao Tong University, Shanghai, China.
| | | |
Collapse
|
7
|
Li C, Wu Y, Zhu Y, Yan J, Liu S, Xu J, Fa S, Yan T, Zhu D, Yan Y, Liu J. Molecular Motor-Driven Light-Controlled Logic-Gated K + Channel for Cancer Cell Apoptosis. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2312352. [PMID: 38301140 DOI: 10.1002/adma.202312352] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 01/11/2024] [Indexed: 02/03/2024]
Abstract
Developing artificial ion transport systems, which process complicated information and step-wise regulate properties, is essential for deeply comprehending the subtle dynamic behaviors of natural channel proteins (NCPs). Here a photo-controlled logic-gated K+ channel based on single-chain random heteropolymers containing molecular motors, exhibiting multi-core processor-like properties to step-wise control ion transport is reported. Designed with oxygen, deoxygenation, and different wavelengths of light as input signals, complicated logical circuits comprising "YES", "AND", "OR" and "NOT" gate components are established. Implementing these logical circuits with K+ transport efficiencies as output signals, multiple state transitions including "ON", "Partially OFF" and "Totally OFF" in liposomes and cancer cells are realized, further causing step-wise anticancer treatments. Dramatic K+ efflux in the "ON" state (decrease by 50% within 7 min) significantly induces cancer cell apoptosis. This integrated logic-gated strategy will be expanded toward understanding the delicate mechanism underlying NCPs and treating cancer or other diseases is expected.
Collapse
Affiliation(s)
- Cong Li
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Hangzhou Normal University, Hangzhou, 311121, China
- College of Chemistry and Chemical Engineering, Key Laboratory of Special Functional and Smart Polymer Materials of Ministry of Industry and Information Technology, Northwestern Polytechnical University, Xi'an, 710129, China
| | - Yaqi Wu
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Hangzhou Normal University, Hangzhou, 311121, China
- College of Chemistry and Chemical Engineering, Key Laboratory of Special Functional and Smart Polymer Materials of Ministry of Industry and Information Technology, Northwestern Polytechnical University, Xi'an, 710129, China
| | - Yihang Zhu
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Hangzhou Normal University, Hangzhou, 311121, China
| | - Jing Yan
- College of Chemistry and Chemical Engineering, Key Laboratory of Special Functional and Smart Polymer Materials of Ministry of Industry and Information Technology, Northwestern Polytechnical University, Xi'an, 710129, China
| | - Shengda Liu
- College of Chemistry and Environmental Engineering, Changchun University of Science and Technology, Changchun, 130022, China
| | - Jiayun Xu
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Hangzhou Normal University, Hangzhou, 311121, China
| | - Shixin Fa
- College of Chemistry and Chemical Engineering, Key Laboratory of Special Functional and Smart Polymer Materials of Ministry of Industry and Information Technology, Northwestern Polytechnical University, Xi'an, 710129, China
| | - Tengfei Yan
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Hangzhou Normal University, Hangzhou, 311121, China
| | - Dingcheng Zhu
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Hangzhou Normal University, Hangzhou, 311121, China
| | - Yi Yan
- College of Chemistry and Chemical Engineering, Key Laboratory of Special Functional and Smart Polymer Materials of Ministry of Industry and Information Technology, Northwestern Polytechnical University, Xi'an, 710129, China
| | - Junqiu Liu
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Hangzhou Normal University, Hangzhou, 311121, China
- College of Chemistry and Chemical Engineering, Key Laboratory of Special Functional and Smart Polymer Materials of Ministry of Industry and Information Technology, Northwestern Polytechnical University, Xi'an, 710129, China
| |
Collapse
|
8
|
Ahmed MBM, El-ssayad MF, Yousef SY, Salem SH. Bee venom: A potential natural alternative to conventional preservatives for prolonging the shelf-life of soft cheese 'Talaga'. Heliyon 2024; 10:e28968. [PMID: 38601605 PMCID: PMC11004823 DOI: 10.1016/j.heliyon.2024.e28968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 03/27/2024] [Accepted: 03/27/2024] [Indexed: 04/12/2024] Open
Abstract
The study aims to explore bee venom (honey-BV) as a potential natural preservative for "Tallaga" soft cheese. Characterization of the active compounds in honey-BV was conducted via chromatographic analyses. Antimicrobial efficacy against pathogenic bacteria and fungi was evaluated, and minimum inhibitory concentration (MIC) was determined. Subsequently, honey-BV was applied to Tallaga cheese at 15 mg/g concentrations. The main active ingredients identified in bee venom were apamin (2%) and melittin (48.7%). Both concentrations of bee venom (100 and 200 mg/mL) exhibited significant antifungal and antibacterial properties against tested organisms, with MIC values varied from 0.2 to 0.5 mg/mL for bacteria to 3-13 mg/mL for fungi. Application of honey-BV in Tallaga cheese resulted in complete elimination of Staphylococcal populations after 2 weeks of cold storage, with no detectable growth of molds or yeasts throughout the storage period. Additionally, a steady decrease in aerobic plate count was observed over time. In summary, honey-BV holds promise as a natural preservative for soft cheese, however, more investigation is required to optimize the concentration for economic viability, taking into account health benefits and safety considerations.
Collapse
Affiliation(s)
- Mohamed Bedair M. Ahmed
- Department of Food Toxicology and Contaminants, National Research Centre, 33 El-Bohouth St., P.O. Box: 12622, Dokki, Cairo, Egypt
| | - Mohamed Fathy El-ssayad
- Dairy Sciences Department, National Research Centre, 33 El-Bohouth St., P.O. Box: 12622, Dokki, Cairo, Egypt
| | | | - Salah H. Salem
- Department of Food Toxicology and Contaminants, National Research Centre, 33 El-Bohouth St., P.O. Box: 12622, Dokki, Cairo, Egypt
| |
Collapse
|
9
|
Larsen NW, Kostrikov S, Hansen MB, Hjørringgaard CU, Larsen NB, Andresen TL, Kristensen K. Interactions of oral permeation enhancers with lipid membranes in simulated intestinal environments. Int J Pharm 2024; 654:123957. [PMID: 38430950 DOI: 10.1016/j.ijpharm.2024.123957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 02/17/2024] [Accepted: 02/28/2024] [Indexed: 03/05/2024]
Abstract
The oral bioavailability of therapeutic peptides is generally low. To increase peptide transport across the gastrointestinal barrier, permeation enhancers are often used. Despite their widespread use, mechanistic knowledge of permeation enhancers is limited. To address this, we here investigate the interactions of six commonly used permeation enhancers with lipid membranes in simulated intestinal environments. Specifically, we study the interactions of the permeation enhancers sodium caprate, dodecyl maltoside, sodium cholate, sodium dodecyl sulfate, melittin, and penetratin with epithelial cell-like model membranes. To mimic the molecular composition of the real intestinal environment, the experiments are performed with two peptide drugs, salmon calcitonin and desB30 insulin, in fasted-state simulated intestinal fluid. Besides providing a comparison of the membrane interactions of the studied permeation enhancers, our results demonstrate that peptide drugs as well as intestinal-fluid components may substantially change the membrane activity of permeation enhancers. This highlights the importance of testing permeation enhancement in realistic physiological environments and carefully choosing a permeation enhancer for each individual peptide drug.
Collapse
Affiliation(s)
- Nanna Wichmann Larsen
- DTU Health Tech, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark; Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Serhii Kostrikov
- DTU Health Tech, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark; Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Morten Borre Hansen
- DTU Health Tech, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark; Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Claudia Ulrich Hjørringgaard
- DTU Health Tech, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark; Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Niels Bent Larsen
- DTU Health Tech, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark; Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Thomas Lars Andresen
- DTU Health Tech, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark; Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark.
| | - Kasper Kristensen
- DTU Health Tech, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark; Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark.
| |
Collapse
|
10
|
Tror S, Jeon S, Nguyen HT, Huh E, Shin K. A Self-Regenerating Artificial Cell, that is One Step Closer to Living Cells: Challenges and Perspectives. SMALL METHODS 2023; 7:e2300182. [PMID: 37246263 DOI: 10.1002/smtd.202300182] [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: 02/12/2023] [Revised: 04/29/2023] [Indexed: 05/30/2023]
Abstract
Controllable, self-regenerating artificial cells (SRACs) can be a vital advancement in the field of synthetic biology, which seeks to create living cells by recombining various biological molecules in the lab. This represents, more importantly, the first step on a long journey toward creating reproductive cells from rather fragmentary biochemical mimics. However, it is still a difficult task to replicate the complex processes involved in cell regeneration, such as genetic material replication and cell membrane division, in artificially created spaces. This review highlights recent advances in the field of controllable, SRACs and the strategies to achieve the goal of creating such cells. Self-regenerating cells start by replicating DNA and transferring it to a location where proteins can be synthesized. Functional but essential proteins must be synthesized for sustained energy generation and survival needs and function in the same liposomal space. Finally, self-division and repeated cycling lead to autonomous, self-regenerating cells. The pursuit of controllable, SRACs will enable authors to make bold advances in understanding life at the cellular level, ultimately providing an opportunity to use this knowledge to understand the nature of life.
Collapse
Affiliation(s)
- Seangly Tror
- Department of Chemistry and Institute of Biological Interfaces, Sogang University, Seoul, 04107, Republic of Korea
| | - SeonMin Jeon
- Department of Chemistry and Institute of Biological Interfaces, Sogang University, Seoul, 04107, Republic of Korea
| | - Huong Thanh Nguyen
- Department of Chemistry and Institute of Biological Interfaces, Sogang University, Seoul, 04107, Republic of Korea
| | - Eunjin Huh
- Department of Chemistry and Institute of Biological Interfaces, Sogang University, Seoul, 04107, Republic of Korea
| | - Kwanwoo Shin
- Department of Chemistry and Institute of Biological Interfaces, Sogang University, Seoul, 04107, Republic of Korea
| |
Collapse
|
11
|
Li S, Zhao Y, Wu S, Zhang X, Yang B, Tian L, Han X. Regulation of species metabolism in synthetic community systems by environmental pH oscillations. Nat Commun 2023; 14:7507. [PMID: 37980410 PMCID: PMC10657449 DOI: 10.1038/s41467-023-43398-6] [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: 03/20/2023] [Accepted: 11/07/2023] [Indexed: 11/20/2023] Open
Abstract
Constructing a synthetic community system helps scientist understand the complex interactions among species in a community and its environment. Herein, a two-species community is constructed with species A (artificial cells encapsulating pH-responsive molecules and sucrose) and species B (Saccharomyces cerevisiae), which causes the environment to exhibit pH oscillation behaviour due to the generation and dissipation of CO2. In addition, a three-species community is constructed with species A' (artificial cells containing sucrose and G6P), species B, and species C (artificial cells containing NAD+ and G6PDH). The solution pH oscillation regulates the periodical release of G6P from species A'; G6P then enters species C to promote the metabolic reaction that converts NAD+ to NADH. The location of species A' and B determines the metabolism behaviour in species C in the spatially coded three-species communities with CA'B, CBA', and A'CB patterns. The proposed synthetic community system provides a foundation to construct a more complicated microecosystem.
Collapse
Affiliation(s)
- Shubin Li
- State Key Laboratory of Urban Water Resource and Environment, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China
| | - Yingming Zhao
- State Key Laboratory of Urban Water Resource and Environment, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China
| | - Shuqi Wu
- Key Laboratory of Biomedical Engineering of Ministry of Education, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Xiangxiang Zhang
- State Key Laboratory of Urban Water Resource and Environment, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China
| | - Boyu Yang
- State Key Laboratory of Urban Water Resource and Environment, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China
| | - Liangfei Tian
- Key Laboratory of Biomedical Engineering of Ministry of Education, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China.
| | - Xiaojun Han
- State Key Laboratory of Urban Water Resource and Environment, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China.
| |
Collapse
|
12
|
Doolan JA, Williams GT, Hilton KLF, Chaudhari R, Fossey JS, Goult BT, Hiscock JR. Advancements in antimicrobial nanoscale materials and self-assembling systems. Chem Soc Rev 2022; 51:8696-8755. [PMID: 36190355 PMCID: PMC9575517 DOI: 10.1039/d1cs00915j] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Indexed: 11/21/2022]
Abstract
Antimicrobial resistance is directly responsible for more deaths per year than either HIV/AIDS or malaria and is predicted to incur a cumulative societal financial burden of at least $100 trillion between 2014 and 2050. Already heralded as one of the greatest threats to human health, the onset of the coronavirus pandemic has accelerated the prevalence of antimicrobial resistant bacterial infections due to factors including increased global antibiotic/antimicrobial use. Thus an urgent need for novel therapeutics to combat what some have termed the 'silent pandemic' is evident. This review acts as a repository of research and an overview of the novel therapeutic strategies being developed to overcome antimicrobial resistance, with a focus on self-assembling systems and nanoscale materials. The fundamental mechanisms of action, as well as the key advantages and disadvantages of each system are discussed, and attention is drawn to key examples within each field. As a result, this review provides a guide to the further design and development of antimicrobial systems, and outlines the interdisciplinary techniques required to translate this fundamental research towards the clinic.
Collapse
Affiliation(s)
- Jack A Doolan
- School of Chemistry and Forensic Science, University of Kent, Canterbury, Kent CT2 7NH, UK.
- School of Biosciences, University of Kent, Canterbury, Kent CT2 7NJ, UK.
| | - George T Williams
- School of Chemistry, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.
| | - Kira L F Hilton
- School of Chemistry and Forensic Science, University of Kent, Canterbury, Kent CT2 7NH, UK.
| | - Rajas Chaudhari
- School of Chemistry and Forensic Science, University of Kent, Canterbury, Kent CT2 7NH, UK.
| | - John S Fossey
- School of Chemistry, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.
| | - Benjamin T Goult
- School of Biosciences, University of Kent, Canterbury, Kent CT2 7NJ, UK.
| | - Jennifer R Hiscock
- School of Chemistry and Forensic Science, University of Kent, Canterbury, Kent CT2 7NH, UK.
| |
Collapse
|
13
|
Yan T, Liu S, Li C, Xu J, Yu S, Wang T, Sun H, Liu J. Flexible Single‐Chain‐Heteropolymer‐Derived Transmembrane Ion Channels with High K
+
Selectivity and Tunable pH‐Gated Characteristics. Angew Chem Int Ed Engl 2022; 61:e202210214. [DOI: 10.1002/anie.202210214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Indexed: 11/11/2022]
Affiliation(s)
- Tengfei Yan
- College of Material Chemistry and Chemical Engineering Key Laboratory of Organosilicon Chemistry and Material Technology Ministry of Education Key Laboratory of Organosilicon Material Technology of Zhejiang Province Hangzhou Normal University Hangzhou 311121 China
| | - Shengda Liu
- College of Material Chemistry and Chemical Engineering Key Laboratory of Organosilicon Chemistry and Material Technology Ministry of Education Key Laboratory of Organosilicon Material Technology of Zhejiang Province Hangzhou Normal University Hangzhou 311121 China
| | - Cong Li
- College of Material Chemistry and Chemical Engineering Key Laboratory of Organosilicon Chemistry and Material Technology Ministry of Education Key Laboratory of Organosilicon Material Technology of Zhejiang Province Hangzhou Normal University Hangzhou 311121 China
| | - Jiayun Xu
- College of Material Chemistry and Chemical Engineering Key Laboratory of Organosilicon Chemistry and Material Technology Ministry of Education Key Laboratory of Organosilicon Material Technology of Zhejiang Province Hangzhou Normal University Hangzhou 311121 China
| | - Shuangjiang Yu
- College of Material Chemistry and Chemical Engineering Key Laboratory of Organosilicon Chemistry and Material Technology Ministry of Education Key Laboratory of Organosilicon Material Technology of Zhejiang Province Hangzhou Normal University Hangzhou 311121 China
| | - Tingting Wang
- Department of Biomedical Engineering College of Design and Engineering National University of Singapore Singapore 117583 Singapore
| | - Hongcheng Sun
- College of Material Chemistry and Chemical Engineering Key Laboratory of Organosilicon Chemistry and Material Technology Ministry of Education Key Laboratory of Organosilicon Material Technology of Zhejiang Province Hangzhou Normal University Hangzhou 311121 China
| | - Junqiu Liu
- College of Material Chemistry and Chemical Engineering Key Laboratory of Organosilicon Chemistry and Material Technology Ministry of Education Key Laboratory of Organosilicon Material Technology of Zhejiang Province Hangzhou Normal University Hangzhou 311121 China
| |
Collapse
|
14
|
Miyazaki Y, Shinoda W. Cooperative antimicrobial action of melittin on lipid membranes: A coarse-grained molecular dynamics study. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2022; 1864:183955. [PMID: 35526599 DOI: 10.1016/j.bbamem.2022.183955] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 04/09/2022] [Accepted: 04/29/2022] [Indexed: 12/29/2022]
Abstract
We conducted a series of coarse-grained molecular dynamics (CG-MD) simulations to investigate the complicated actions of melittin, which is an antimicrobial peptide (AMP) derived from honey bee venom, on a lipid membrane. To accurately simulate the AMP action, we developed and used a protein CG model as an extension of the pSPICA force field (FF), which was designed to reproduce several thermodynamic quantities and structural properties. At a low peptide-to-lipid (P/L) ratio (1/102), no defect was detected. At P/L = 1/51, toroidal pore formation was observed due to collective insertion of multiple melittin peptides from the N-termini. The pore formation was initiated by a local increase in membrane curvature in the vicinity of the peptide aggregate. At a higher P/L ratio (1/26), two more modes were detected, seemingly not controlled by the P/L ratio but by a local arrangement of melittin peptides: 1. Pore formation accompanied by lipid extraction by melittin peptides:a detergent-like mechanism. 2. A rapidly formed large pore in a significantly curved membrane: bursting. Thus, we observed three pore formation modes (toroidal pore formation, lipid extraction, and bursting) depending on the peptide concentration and local arrangement. These observations were consistent with experimental observations and hypothesized melittin modes. Through this study, we found that the local arrangements and population of melittin peptides and the area expansion rate by membrane deformation were key to the initiation of and competition among the multiple pore formation mechanisms.
Collapse
Affiliation(s)
- Yusuke Miyazaki
- Department of Materials Chemistry, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan; Research Institute for Interdisciplinary Science, Okayama University, 3-1-1 Tsushima-naka, Kita-ku, Okayama 700-8530, Japan
| | - Wataru Shinoda
- Department of Materials Chemistry, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan; Research Institute for Interdisciplinary Science, Okayama University, 3-1-1 Tsushima-naka, Kita-ku, Okayama 700-8530, Japan; Department of Chemistry, Faculty of Science, Okayama University, 3-1-1 Tsushima-naka, Kita-ku, Okayama 700-8530, Japan.
| |
Collapse
|
15
|
Yan T, Liu S, Li C, Xu J, Yu S, Wang T, Sun H, Liu J. Flexible Single‐Chain‐Heteropolymer‐Derived Transmembrane Ion Channels with High K+ Selectivity and Tunable pH‐Gated Characteristics. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202210214] [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)
- Tengfei Yan
- Hangzhou Normal University College of Material, Chemistry and Chemical Engineering CHINA
| | - Shengda Liu
- Hangzhou Normal University College of Material, Chemistry and Chemical Engineering CHINA
| | - Cong Li
- Hangzhou Normal University College of Material, Chemistry and Chemical Engineering CHINA
| | - Jiayun Xu
- Hangzhou Normal University College of Material, Chemistry and Chemical Engineering CHINA
| | - Shuangjiang Yu
- Hangzhou Normal University College of Material, Chemistry and Chemical Engineering CHINA
| | - Tingting Wang
- National University of Singapore Department of Biomedical Engineering, College of Design and Engineering Singapore SINGAPORE
| | - Hongcheng Sun
- Hangzhou Normal University College of Material, Chemistry and Chemical Engineering CHINA
| | - Junqiu Liu
- Jilin University State Key Laboratory of Supramolecular Structure and Materials Qianjin Street 2699# 130012 Changchun CHINA
| |
Collapse
|
16
|
Perez-Rodriguez A, Eraso E, Quindós G, Mateo E. Antimicrobial Peptides with Anti-Candida Activity. Int J Mol Sci 2022; 23:ijms23169264. [PMID: 36012523 PMCID: PMC9409312 DOI: 10.3390/ijms23169264] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/10/2022] [Accepted: 08/13/2022] [Indexed: 02/06/2023] Open
Abstract
Mycoses are accountable for millions of infections yearly worldwide. Invasive candidiasis is the most usual, presenting a high morbidity and mortality. Candida albicans remains the prevalent etiologic agent, but the incidence of other species such as Candida parapsilosis, Candida glabrata and Candida auris keeps increasing. These pathogens frequently show a reduced susceptibility to commonly used antifungal drugs, including polyenes, triazoles and echinocandins, and the incidence of emerging multi-drug-resistant strains of these species continues to increase. Therefore, the need to search for new molecules that target these pathogenic species in a different manner is now more urgent than ever. Nature is an almost endless source of interesting new molecules that could meet this need. Among these molecules, antimicrobial peptides, present in different sources in nature, possess some advantages over conventional antifungal agents, even with their own drawbacks, and are considered as a promising pharmacological option against a wide range of microbial infections. In this review, we describe 20 antimicrobial peptides from different origins that possess an activity against Candida.
Collapse
|
17
|
Yi L, Chen S, Li G, Ren J, Zhou R, Zeng K. Prevalence of antibiotic resistance pathogens in online fresh-cut fruit from Chongqing, China and controlling Enterococcus faecalis by bacteriocin GF-15. Lebensm Wiss Technol 2022. [DOI: 10.1016/j.lwt.2022.113678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
18
|
Kaushik V, Tiwari M, Tiwari V. Interaction of RecA mediated SOS response with bacterial persistence, biofilm formation, and host response. Int J Biol Macromol 2022; 217:931-943. [PMID: 35905765 DOI: 10.1016/j.ijbiomac.2022.07.176] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/20/2022] [Accepted: 07/22/2022] [Indexed: 11/28/2022]
Abstract
Antibiotics have a primary mode of actions, and most of them have a common secondary mode of action via reactive species (ROS and RNS) mediated DNA damage. Bacteria have been able to tolerate this DNA damage by SOS (Save-Our-Soul) response. RecA is the universal essential key protein of the DNA damage mediated SOS repair in various bacteria including ESKAPE pathogens. In addition, antibiotics also triggers activation of various other bacterial mechanisms such as biofilm formation, host dependent responses, persister subpopulation formation. These supporting the survival of bacteria in unfriendly natural conditions i.e. antibiotic presence. This review highlights the detailed mechanism of RecA mediated SOS response as well as role of RecA-LexA interaction in SOS response. The review also focuses on inter-connection between DNA damage repair pathway (like SOS response) with other survival mechanisms of bacteria such as host mediated RecA induction, persister-SOS interplay, and biofilm-SOS interplay. This understanding of inter-connection of SOS response with different other survival mechanisms will prove beneficial in targeting the SOS response for prevention and development of therapeutics against recalcitrant bacterial infections. The review also covers the significance of RecA as a promising potent therapeutic target for hindering bacterial SOS response in prevailing successful treatments of bacterial infections and enhancing the conventional antibiotic efficiency.
Collapse
Affiliation(s)
- Vaishali Kaushik
- Department of Biochemistry, Central University of Rajasthan, Ajmer 305817, India
| | - Monalisa Tiwari
- Department of Biochemistry, Central University of Rajasthan, Ajmer 305817, India
| | - Vishvanath Tiwari
- Department of Biochemistry, Central University of Rajasthan, Ajmer 305817, India.
| |
Collapse
|
19
|
Blair JMA, Zeth K, Bavro VN, Sancho-Vaello E. The role of bacterial transport systems in the removal of host antimicrobial peptides in Gram-negative bacteria. FEMS Microbiol Rev 2022; 46:6617596. [PMID: 35749576 PMCID: PMC9629497 DOI: 10.1093/femsre/fuac032] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 05/23/2022] [Accepted: 06/22/2022] [Indexed: 01/09/2023] Open
Abstract
Antibiotic resistance is a global issue that threatens our progress in healthcare and life expectancy. In recent years, antimicrobial peptides (AMPs) have been considered as promising alternatives to the classic antibiotics. AMPs are potentially superior due to their lower rate of resistance development, since they primarily target the bacterial membrane ('Achilles' heel' of the bacteria). However, bacteria have developed mechanisms of AMP resistance, including the removal of AMPs to the extracellular space by efflux pumps such as the MtrCDE or AcrAB-TolC systems, and the internalization of AMPs to the cytoplasm by the Sap transporter, followed by proteolytic digestion. In this review, we focus on AMP transport as a resistance mechanism compiling all the experimental evidence for the involvement of efflux in AMP resistance in Gram-negative bacteria and combine this information with the analysis of the structures of the efflux systems involved. Finally, we expose some open questions with the aim of arousing the interest of the scientific community towards the AMPs-efflux pumps interactions. All the collected information broadens our understanding of AMP removal by efflux pumps and gives some clues to assist the rational design of AMP-derivatives as inhibitors of the efflux pumps.
Collapse
Affiliation(s)
- Jessica M A Blair
- College of Medical and Dental Sciences, Institute of Microbiology and Infection, University of Birmingham, Edgbaston, Birmingham, B15 2TT, United Kingdom
| | - Kornelius Zeth
- Department of Science and Environment, Roskilde University, Universitetsvej 1, 4000 Roskilde, Denmark
| | - Vassiliy N Bavro
- School of Life Sciences, University of Essex, Colchester, CO4 3SQ, United Kingdom
| | - Enea Sancho-Vaello
- Corresponding author. College of Medical and Dental Sciences, Institute of Microbiology and Infection, University of Birmingham, Edgbaston, Birmingham, B15 2TT, United Kingdom. E-mail:
| |
Collapse
|
20
|
Wichmann N, Lund PM, Hansen MB, Hjørringgaard CU, Larsen JB, Kristensen K, Andresen TL, Simonsen JB. Applying flow cytometry to identify the modes of action of membrane-active peptides in a label-free and high-throughput fashion. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2022; 1864:183820. [PMID: 34813768 DOI: 10.1016/j.bbamem.2021.183820] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 10/29/2021] [Accepted: 11/12/2021] [Indexed: 06/13/2023]
Abstract
Membrane-active peptides (MAPs) have several potential therapeutic uses, including as antimicrobial drugs. Many traditional methods used to evaluate the membrane interactions of MAPs have limited applicability. Low-throughput methods, such as microscopy, provide detailed information but often rely on fluorophore-labeled MAPs, and high-throughput assays, such as the calcein release assay, cannot assess the mechanism behind the disruption of vesicular-based lipid membranes. Here we present a flow cytometric assay that provides detailed information about the peptide-lipid membrane interactions on single artificial lipid vesicles while being high-throughput (1000-2000 vesicles/s) and based on label-free MAPs. We synthesized and investigated six MAPs with different modes of action to evaluate the versatility of the assay. The assay is based on the flow cytometric readouts from artificial lipid vesicles, including the fluorescence from membrane-anchored and core-encapsulated fluorophores, and the vesicle concentration. From these parameters, we were able to distinguish between MAPs that induce vesicle solubilization, permeation (pores/membrane distortion), and aggregation or fusion. Our flow cytometry findings have been verified by traditional methods, including the calcein release assay, dynamic light scattering, and fluorescence microscopy on giant unilamellar vesicles. We envision that the presented flow cytometric assay can be used for various types of peptide-lipid membrane studies, e.g. to identify new antibiotics. Moreover, the assay can easily be expanded to derive additional valuable information.
Collapse
Affiliation(s)
- Nanna Wichmann
- Department of Health Technology, Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Philip M Lund
- Department of Health Technology, Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Morten B Hansen
- Department of Health Technology, Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Claudia U Hjørringgaard
- Department of Health Technology, Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Jannik B Larsen
- Department of Health Technology, Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Kasper Kristensen
- Department of Health Technology, Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Thomas L Andresen
- Department of Health Technology, Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark.
| | - Jens B Simonsen
- Department of Health Technology, Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark.
| |
Collapse
|
21
|
Liu X, Stenhammar J, Wennerström H, Sparr E. Vesicles Balance Osmotic Stress with Bending Energy That Can Be Released to Form Daughter Vesicles. J Phys Chem Lett 2022; 13:498-507. [PMID: 35005979 PMCID: PMC8785185 DOI: 10.1021/acs.jpclett.1c03369] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 12/28/2021] [Indexed: 06/14/2023]
Abstract
The bending energy of the lipid membrane is central to biological processes involving vesicles, such as endocytosis and exocytosis. To illustrate the role of bending energy in these processes, we study the response of single-component giant unilamellar vesicles (GUVs) subjected to external osmotic stress by glucose addition. For osmotic pressures exceeding 0.15 atm, an abrupt shape change from spherical to prolate occurs, showing that the osmotic pressure is balanced by the free energy of membrane bending. After equilibration, the external glucose solution was exchanged for pure water, yielding rapid formation of monodisperse daughter vesicles inside the GUVs through an endocytosis-like process. Our theoretical analysis shows that this process requires significant free energies stored in the deformed membrane to be kinetically allowed. The results indicate that bending energies stored in GUVs are much higher than previously implicated, with potential consequences for vesicle fusion/fission and the osmotic regulation in living cells.
Collapse
Affiliation(s)
- Xiaoyan Liu
- Physical Chemistry, Lund University, 221 00 Lund, Sweden
| | | | | | - Emma Sparr
- Physical Chemistry, Lund University, 221 00 Lund, Sweden
| |
Collapse
|
22
|
Matsuzaki K. Elucidation of Complex Dynamic Intermolecular Interactions in Membranes. Chem Pharm Bull (Tokyo) 2022; 70:1-9. [PMID: 34980725 DOI: 10.1248/cpb.c21-00815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Biomembranes composed of various proteins and lipids play important roles in cellular functions, such as signal transduction and substance transport. In addition, some bioactive peptides and pathogenic proteins target membrane proteins and lipids to exert their effects. Therefore, an understanding of dynamic and complex intermolecular interactions among these membrane constituents is needed to elucidate their mechanisms. This review summarizes the major research carried out in the author's laboratory on how lipids and their inhomogeneous distributions regulate the structures and functions of antimicrobial peptides and Alzheimer's amyloid β-protein. Also, how to detect transmembrane helix-helix and membrane protein-protein interactions and how they are modulated by lipids are discussed.
Collapse
|
23
|
Yan T, Liu S, Xu J, Sun H, Yu S, Liu J. Unimolecular Helix-Based Transmembrane Nanochannel with a Smallest Luminal Cavity of 1 Å Expressing High Proton Selectivity and Transport Activity. NANO LETTERS 2021; 21:10462-10468. [PMID: 34860025 DOI: 10.1021/acs.nanolett.1c03858] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Natural protein channels have evolved with exquisite structures to transport ions selectively and rapidly. Learning from nature to construct biomimetic artificial channels is always challenging. Herein we present a unimolecular transmembrane proton channel by quinoline-derived helix, which exhibited highly selective and ultrafast proton transport behaviors. This helix-based channel possesses a small luminal cavity of 1 Å in diameter, which could efficiently reject the permeation of cations, anions or water molecules but only permits the translocation of protons owing to the size effect. The proton flow rate exceeded 107 H+ s-1 channel-1 and reached the same magnitude with gramicidin A. Mechanism investigation revealed that the directionally arrayed NH-chain inside the synthetic channel played a pivotal role during the proton flux. This work not only presented a helix-based channel with the smallest observable nanopore, but also unveiled an unexplored pathway for realizing efficient transport of protons via the consecutive NH-chain.
Collapse
Affiliation(s)
- Tengfei Yan
- College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121, China
| | - Shengda Liu
- College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121, China
| | - Jiayun Xu
- College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121, China
| | - Hongcheng Sun
- College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121, China
| | - Shuangjiang Yu
- College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121, China
| | - Junqiu Liu
- College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121, China
- College of Chemistry, State Key Laboratory of Supramolecular Structure and Materials, Jilin University, Changchun 130012, China
| |
Collapse
|
24
|
Rocha-Roa C, Orjuela JD, Leidy C, Cossio P, Aponte-Santamaría C. Cardiolipin prevents pore formation in phosphatidylglycerol bacterial membrane models. FEBS Lett 2021; 595:2701-2714. [PMID: 34633077 DOI: 10.1002/1873-3468.14206] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/17/2021] [Accepted: 10/04/2021] [Indexed: 12/12/2022]
Abstract
Several antimicrobial peptides, including magainin and the human cathelicidin LL-37, act by forming pores in bacterial membranes. Bacteria such as Staphylococcus aureus modify their membrane's cardiolipin composition to resist such types of perturbations that compromise their membrane stability. Here, we used molecular dynamic simulations to quantify the role of cardiolipin on the formation of pores in simple bacterial-like membrane models composed of phosphatidylglycerol and cardiolipin mixtures. Cardiolipin modified the structure and ordering of the lipid bilayer, making it less susceptible to mechanical changes. Accordingly, the free-energy barrier for the formation of a transmembrane pore and its kinetic instability augmented by increasing the cardiolipin concentration. This is attributed to the unfavorable positioning of cardiolipin near the formed pore, due to its small polar head and bulky hydrophobic body. Overall, our study demonstrates how cardiolipin prevents membrane-pore formation and this constitutes a plausible mechanism used by bacteria to act against stress perturbations and, thereby, gain resistance to antimicrobial agents.
Collapse
Affiliation(s)
- Cristian Rocha-Roa
- Biophysics of Tropical Diseases, Max Planck Tandem Group, University of Antioquia, Medellín, Colombia
| | - Juan David Orjuela
- Max Planck Tandem Group in Computational Biophysics, Universidad de los Andes, Bogotá, Colombia
- Department of Biomedical Engineering, Universidad de los Andes, Bogotá, Colombia
| | - Chad Leidy
- Biophysics Group, Department of Physics, Universidad de los Andes, Bogotá, Colombia
| | - Pilar Cossio
- Biophysics of Tropical Diseases, Max Planck Tandem Group, University of Antioquia, Medellín, Colombia
- Department of Theoretical Biophysics, Max Planck Institute of Biophysics, Frankfurt, Germany
| | | |
Collapse
|
25
|
Sun L, Hristova K, Wimley WC. Membrane-selective nanoscale pores in liposomes by a synthetically evolved peptide: implications for triggered release. NANOSCALE 2021; 13:12185-12197. [PMID: 34190297 PMCID: PMC9265991 DOI: 10.1039/d1nr03084a] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Peptides that form nanoscale pores in lipid bilayers have potential applications in triggered release, but only if their selectivity for target synthetic membranes over bystander biomembranes can be optimized. Previously, we identified a novel family of α-helical pore-forming peptides called "macrolittins", which release macromolecular cargoes from phosphatidylcholine (PC) liposomes at concentrations as low as 1 peptide per 1000 lipids. In this work, we show that macrolittins have no measurable cytolytic activity against multiple human cell types even at high peptide concentration. This unprecedented selectivity for PC liposomes over cell plasma membranes is explained, in part, by the sensitivity of macrolittin activity to physical chemical properties of the bilayer hydrocarbon core. In the presence of cells, macrolittins release all vesicle-entrapped cargoes (proteins and small molecule drugs) which are then readily uptaken by cells. Triggered release occurs without any direct effect of the peptide on the cells, and without vesicle-vesicle or vesicle-cell interactions.
Collapse
Affiliation(s)
- Leisheng Sun
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, Louisiana 70112, USA.
| | | | | |
Collapse
|
26
|
Pirtskhalava M, Vishnepolsky B, Grigolava M, Managadze G. Physicochemical Features and Peculiarities of Interaction of AMP with the Membrane. Pharmaceuticals (Basel) 2021; 14:471. [PMID: 34067510 PMCID: PMC8156082 DOI: 10.3390/ph14050471] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/25/2021] [Accepted: 04/29/2021] [Indexed: 02/06/2023] Open
Abstract
Antimicrobial peptides (AMPs) are anti-infectives that have the potential to be used as a novel and untapped class of biotherapeutics. Modes of action of antimicrobial peptides include interaction with the cell envelope (cell wall, outer- and inner-membrane). A comprehensive understanding of the peculiarities of interaction of antimicrobial peptides with the cell envelope is necessary to perform a rational design of new biotherapeutics, against which working out resistance is hard for microbes. In order to enable de novo design with low cost and high throughput, in silico predictive models have to be invoked. To develop an efficient predictive model, a comprehensive understanding of the sequence-to-function relationship is required. This knowledge will allow us to encode amino acid sequences expressively and to adequately choose the accurate AMP classifier. A shared protective layer of microbial cells is the inner, plasmatic membrane. The interaction of AMP with a biological membrane (native and/or artificial) has been comprehensively studied. We provide a review of mechanisms and results of interactions of AMP with the cell membrane, relying on the survey of physicochemical, aggregative, and structural features of AMPs. The potency and mechanism of AMP action are presented in terms of amino acid compositions and distributions of the polar and apolar residues along the chain, that is, in terms of the physicochemical features of peptides such as hydrophobicity, hydrophilicity, and amphiphilicity. The survey of current data highlights topics that should be taken into account to come up with a comprehensive explanation of the mechanisms of action of AMP and to uncover the physicochemical faces of peptides, essential to perform their function. Many different approaches have been used to classify AMPs, including machine learning. The survey of knowledge on sequences, structures, and modes of actions of AMP allows concluding that only possessing comprehensive information on physicochemical features of AMPs enables us to develop accurate classifiers and create effective methods of prediction. Consequently, this knowledge is necessary for the development of design tools for peptide-based antibiotics.
Collapse
Affiliation(s)
- Malak Pirtskhalava
- Ivane Beritashvili Center of Experimental Biomedicine, Tbilisi 0160, Georgia; (B.V.); (M.G.); (G.M.)
| | | | | | | |
Collapse
|
27
|
Zhou J, Wan C, Cheng J, Huang H, Lovell JF, Jin H. Delivery Strategies for Melittin-Based Cancer Therapy. ACS APPLIED MATERIALS & INTERFACES 2021; 13:17158-17173. [PMID: 33847113 DOI: 10.1021/acsami.1c03640] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Melittin (MLT) has been studied preclinically as an anticancer agent based on its broad lytic effects in multiple tumor types. However, unsatisfactory tissue distribution, hemolysis, rapid metabolism, and limited specificity are critical obstacles that limit the translation of MLT. Emerging drug delivery strategies hold promise for targeting, controlled drug release, reduced side effects, and ultimately improved treatment efficiency. In this review, we discuss recent advances in the use of diverse carriers to deliver MLT, with an emphasis on the design and mechanisms of action. We further outline the opportunities for MLT-based cancer immunotherapy.
Collapse
Affiliation(s)
- Jie Zhou
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, P. R. China
| | - Chao Wan
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, P. R. China
| | - Jing Cheng
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, P. R. China
| | - Hao Huang
- Guo Life Science Center, Wuhan Shengrun Biotechnology Co. Ltd, Wuhan 430075, P.R. China
| | - Jonathan F Lovell
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, New York 14260, United States
| | - Honglin Jin
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, P. R. China
| |
Collapse
|
28
|
Huynh L, Velásquez J, Rabara R, Basu S, Nguyen HB, Gupta G. Rational design of antimicrobial peptides targeting Gram-negative bacteria. Comput Biol Chem 2021; 92:107475. [PMID: 33813188 DOI: 10.1016/j.compbiolchem.2021.107475] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 03/02/2021] [Accepted: 03/09/2021] [Indexed: 01/16/2023]
Abstract
Membrane-targeting host antimicrobial peptides (AMPs) can kill or inhibit the growth of Gram-negative bacteria. However, the evolution of resistance among microbes poses a substantial barrier to the long-term utility of the host AMPs. Combining experiment and molecular dynamics simulations, we show that terminal carboxyl capping enhances both membrane insertion and antibacterial activity of an AMP called P1. Furthermore, we show that a bacterial strain with evolved resistance to this peptide becomes susceptible to P1 variants with either backbone capping or lysine-to-arginine substitutions. Our results suggest that cocktails of closely related AMPs may be useful in overcoming evolved resistance.
Collapse
Affiliation(s)
- Loan Huynh
- New Mexico Consortium, Los Alamos, NM, 87544, USA
| | | | - Roel Rabara
- New Mexico Consortium, Los Alamos, NM, 87544, USA
| | | | - Hau B Nguyen
- Los Alamos National Laboratory, Los Alamos, NM 87545, USA
| | - Goutam Gupta
- New Mexico Consortium, Los Alamos, NM, 87544, USA.
| |
Collapse
|
29
|
Guo X, Rao J, Yan T, Zhang B, Yang W, Sun W, Xie J. Feleucin-K3 Analogue with an α-(4-Pentenyl)-Ala Substitution at the Key Site Has More Potent Antimicrobial and Antibiofilm Activities in Vitro and in Vivo. ACS Infect Dis 2021; 7:64-78. [PMID: 33296183 DOI: 10.1021/acsinfecdis.0c00545] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The development of antimicrobial compounds is now regarded as an urgent problem. Antimicrobial peptides (AMPs) have great potential to become novel antimicrobial drugs. Feleucin-K3 is an α-helical cationic AMP isolated from the skin secretion of the Asian bombinid toad species Bombina orientalis and has antimicrobial activity. In our previous studies, amino acid scanning of Feleucin-K3 was performed to determine the key site affecting its activity. In this study, we investigated and synthesized a series of analogues that have either a natural or an unnatural hydrophobic amino acid substitution at the fourth amino acid residue of Feleucin-K3. Among these analogues, Feleucin-K59 (K59), which has an α-(4-pentenyl)-Ala substitution, was shown to have increased antimicrobial activity against both standard and drug-resistant strains of clinical common bacteria, improved stability, no hemolytic activity at antimicrobial concentrations, and no resistance. In addition, K59 has potent antibiofilm activity in vitro. More importantly, K59 showed better antimicrobial and antibiofilm activities against drug-resistant bacteria in in vivo experiments in mice than traditional antibiotics. In this preliminary study of the mechanism of action, we found that K59 could rapidly kill bacteria by a dual-action mechanism of disrupting the cell membrane and binding to intracellular DNA, thus making it difficult for bacteria to develop resistance.
Collapse
Affiliation(s)
- Xiaomin Guo
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, China
| | - Jing Rao
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, China
| | - Tiantian Yan
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, China
| | - Bangzhi Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, China
| | - Wenle Yang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, China
| | - Wangsheng Sun
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, China
| | - Junqiu Xie
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, China
| |
Collapse
|
30
|
Martínez-Calle M, Parra-Ortiz E, Cruz A, Olmeda B, Pérez-Gil J. Towards the Molecular Mechanism of Pulmonary Surfactant Protein SP-B: At the Crossroad of Membrane Permeability and Interfacial Lipid Transfer. J Mol Biol 2020; 433:166749. [PMID: 33309854 DOI: 10.1016/j.jmb.2020.166749] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 11/14/2020] [Accepted: 12/03/2020] [Indexed: 11/17/2022]
Abstract
Pulmonary surfactant is a lipid-protein complex that coats the alveolar air-liquid interface, enabling the proper functioning of lung mechanics. The hydrophobic surfactant protein SP-B, in particular, plays an indispensable role in promoting the rapid adsorption of phospholipids into the interface. For this, formation of SP-B ring-shaped assemblies seems to be important, as oligomerization could be required for the ability of the protein to generate membrane contacts and to mediate lipid transfer among surfactant structures. SP-B, together with the other hydrophobic surfactant protein SP-C, also promotes permeability of surfactant membranes to polar molecules although the molecular mechanisms underlying this property, as well as its relevance for the surface activity of the protein, remain undefined. In this work, the contribution of SP-B and SP-C to surfactant membrane permeability has been further investigated, by evaluation of the ability of differently-sized fluorescent polar probes to permeate through giant vesicles with different lipid/protein composition. Our results are consistent with the generation by SP-B of pores with defined size in surfactant membranes. Furthermore, incubation of surfactant with an anti-SP-B antibody not only blocked membrane permeability but also affected lipid transfer into the air-water interface, as observed in a captive bubble surfactometer device. Our findings include the identification of SP-C and anionic phospholipids as modulators required for maintaining native-like permeability features in pulmonary surfactant membranes. Proper permeability through membrane assemblies could be crucial to complement the overall role of surfactant in maintaining alveolar equilibrium, beyond its biophysical function in stabilizing the respiratory air-liquid interface.
Collapse
Affiliation(s)
- Marta Martínez-Calle
- Department of Biochemistry, Faculty of Biology, Complutense University, Madrid, Spain; Research Institute "Hospital 12 de Octubre (imas12)", Complutense University, Madrid, Spain
| | - Elisa Parra-Ortiz
- Department of Biochemistry, Faculty of Biology, Complutense University, Madrid, Spain
| | - Antonio Cruz
- Department of Biochemistry, Faculty of Biology, Complutense University, Madrid, Spain; Research Institute "Hospital 12 de Octubre (imas12)", Complutense University, Madrid, Spain
| | - Barbara Olmeda
- Department of Biochemistry, Faculty of Biology, Complutense University, Madrid, Spain; Research Institute "Hospital 12 de Octubre (imas12)", Complutense University, Madrid, Spain.
| | - Jesús Pérez-Gil
- Department of Biochemistry, Faculty of Biology, Complutense University, Madrid, Spain; Research Institute "Hospital 12 de Octubre (imas12)", Complutense University, Madrid, Spain
| |
Collapse
|
31
|
Nishimura T, de Campo L, Iwase H, Akiyoshi K. Determining the Hydration in the Hydrophobic Layer of Permeable Polymer Vesicles by Neutron Scattering. Macromolecules 2020. [DOI: 10.1021/acs.macromol.0c01261] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Tomoki Nishimura
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Liliana de Campo
- Australian Centre for Neutron Scattering, Australian Nuclear Science and Technology Organisation (ANSTO), New Illawarra Road, Lucas Heights, NSW 2234, Australia
| | - Hiroki Iwase
- Research Center for Neutron Science and Technology, Comprehensive Research Organization for Science and Society (CROSS), Tokai, Naka, Ibaraki 319-1106, Japan
| | - Kazunari Akiyoshi
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| |
Collapse
|
32
|
Anti-fungal properties and mechanisms of melittin. Appl Microbiol Biotechnol 2020; 104:6513-6526. [PMID: 32500268 DOI: 10.1007/s00253-020-10701-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/15/2020] [Accepted: 05/24/2020] [Indexed: 12/17/2022]
Abstract
Many fungal diseases remain poorly addressed by public health authorities, despite posing a substantial threat to humans, animals, and plants. More worryingly, few classes of anti-fungals have been developed to combat fungal infections thus far. These medications also have certain drawbacks in terms of toxicity, spectrum of activity, and pharmacokinetic properties. Hence, there is a dire need for discovery of novel anti-fungal agents. Melittin, the main constituent in the venom of European honeybee Apis mellifera, has attracted considerable attention among researchers owing to its potential therapeutic applications. To our knowledge, there has been no review pertinent to anti-fungal properties of melittin, prompting us to synopsize the results of experimental investigations with a special emphasis upon underlying mechanisms. In this respect, melittin inhibits a broad spectrum of fungal genera including Aspergillus, Botrytis, Candida, Colletotrichum, Fusarium, Malassezia, Neurospora, Penicillium, Saccharomyces, Trichoderma, Trichophyton, and Trichosporon. Melittin hinders fungal growth by several mechanisms such as membrane permeabilization, apoptosis induction by reactive oxygen species-mediated mitochondria/caspase-dependent pathway, inhibition of (1,3)-β-D-glucan synthase, and alterations in fungal gene expression. Overall, melittin will definitely open up new avenues for various biomedical applications, from medicine to agriculture. KEYPOINTS: • Venom-derived peptides have potential for development of anti-microbial agents. • Many fungal pathogens are susceptible to melittin at micromolar concentrations. • Melittin possesses multi-target mechanism of action against fungal cells.
Collapse
|
33
|
Juhaniewicz-Dębińska J, Dziubak D, Sęk S. Physicochemical Characterization of Daptomycin Interaction with Negatively Charged Lipid Membranes. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2020; 36:5324-5335. [PMID: 32340456 PMCID: PMC7588137 DOI: 10.1021/acs.langmuir.0c00716] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Daptomycin is known as an effective antibiotic lipopeptide which shows activity against the number of Gram-positive pathogens. Its primary target is the bacterial cell membrane. However, the detailed mechanism of daptomycin action is still subject to debate. In this paper, we have investigated the interactions between lipopeptide and model lipid films composed of negatively charged phosphatidylglycerols and cardiolipin. In order to evaluate the effect of daptomycin on the molecular organization and the properties of lipid assemblies, we have used surface pressure measurements and electrochemical methods combined with atomic force microscopy, quartz crystal microbalance, and surface-enhanced infrared absorption spectroscopy. Our results indicate that daptomycin interaction with the lipid membrane is complex. It involves daptomycin aggregation and partial insertion, which in turn affect the charge distribution on both sides of the membrane and may result in a gradient of water chemical potential. The latter can drive the flux of water across the membrane.
Collapse
|
34
|
Lee MW, de Anda J, Kroll C, Bieniossek C, Bradley K, Amrein KE, Wong GCL. How do cyclic antibiotics with activity against Gram-negative bacteria permeate membranes? A machine learning informed experimental study. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183302. [PMID: 32311341 DOI: 10.1016/j.bbamem.2020.183302] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 03/26/2020] [Accepted: 03/30/2020] [Indexed: 12/30/2022]
Abstract
All antibiotics have to engage bacterial amphiphilic barriers such as the lipopolysaccharide-rich outer membrane or the phospholipid-based inner membrane in some manner, either by disrupting them outright and/or permeating them and thereby allow the antibiotic to get into bacteria. There is a growing class of cyclic antibiotics, many of which are of bacterial origin, that exhibit activity against Gram-negative bacteria, which constitute an urgent problem in human health. We examine a diverse collection of these cyclic antibiotics, both natural and synthetic, which include bactenecin, polymyxin B, octapeptin, capreomycin, and Kirshenbaum peptoids, in order to identify what they have in common when they interact with bacterial lipid membranes. We find that they virtually all have the ability to induce negative Gaussian curvature (NGC) in bacterial membranes, the type of curvature geometrically required for permeation mechanisms such as pore formation, blebbing, and budding. This is interesting since permeation of membranes is a function usually ascribed to antimicrobial peptides (AMPs) from innate immunity. As prototypical test cases of cyclic antibiotics, we analyzed amino acid sequences of bactenecin, polymyxin B, and capreomycin using our recently developed machine-learning classifier trained on α-helical AMP sequences. Although the original classifier was not trained on cyclic antibiotics, a modified classifier approach correctly predicted that bactenecin and polymyxin B have the ability to induce NGC in membranes, while capreomycin does not. Moreover, the classifier was able to recapitulate empirical structure-activity relationships from alanine scans in polymyxin B surprisingly well. These results suggest that there exists some common ground in the sequence design of hybrid cyclic antibiotics and linear AMPs.
Collapse
Affiliation(s)
- Michelle W Lee
- Department of Bioengineering, Department of Chemistry, California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, United States
| | - Jaime de Anda
- Department of Bioengineering, Department of Chemistry, California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, United States
| | - Carsten Kroll
- Roche Pharma Research and Early Development Pharmaceutical Science, Roche, Innovation Center Basel, F. Hoffmann-La Roche Ltd, 4070 Basel, Switzerland
| | - Christoph Bieniossek
- Roche Pharma Research and Early Development Pharmaceutical Science, Roche, Innovation Center Basel, F. Hoffmann-La Roche Ltd, 4070 Basel, Switzerland
| | - Kenneth Bradley
- Roche Pharma Research and Early Development Pharmaceutical Science, Roche, Innovation Center Basel, F. Hoffmann-La Roche Ltd, 4070 Basel, Switzerland
| | - Kurt E Amrein
- Roche Pharma Research and Early Development Pharmaceutical Science, Roche, Innovation Center Basel, F. Hoffmann-La Roche Ltd, 4070 Basel, Switzerland
| | - Gerard C L Wong
- Department of Bioengineering, Department of Chemistry, California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, United States.
| |
Collapse
|
35
|
Liposomal membrane permeability assessment by fluorescence techniques: Main permeabilizing agents, applications and challenges. Int J Pharm 2020; 580:119198. [PMID: 32169353 DOI: 10.1016/j.ijpharm.2020.119198] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 02/19/2020] [Accepted: 03/02/2020] [Indexed: 01/08/2023]
Abstract
Liposomes are lipid vesicles made of one or multiple lipid bilayers surrounding an internal aqueous core. They are broadly employed as models to study membrane structure and properties. Among these properties, liposome membrane permeability is crucial and widely assessed by fluorescence techniques. The first part of this review is devoted to describe the various techniques used for membrane permeability assessment. Attention is paid to fluorescence techniques based on vesicle leakage of self-quenching probes, dye/quencher pair or cation/ligand pair. Secondly, the membrane-active agents inducing membrane permeabilization is presented and details on their mechanisms of action are given. Emphasis is also laid on the intrinsic and extrinsic factors that can modulate the membrane permeability. Hence, a suitable liposomal membrane should be formulated according to the aim of the study and its application.
Collapse
|
36
|
Felizatti AP, Zeraik AE, Basso LG, Kumagai PS, Lopes JL, Wallace B, Araujo AP, DeMarco R. Interactions of amphipathic α-helical MEG proteins from Schistosoma mansoni with membranes. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183173. [DOI: 10.1016/j.bbamem.2019.183173] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 12/19/2019] [Accepted: 12/23/2019] [Indexed: 02/04/2023]
|
37
|
Sabapathy T, Deplazes E, Mancera RL. Revisiting the Interaction of Melittin with Phospholipid Bilayers: The Effects of Concentration and Ionic Strength. Int J Mol Sci 2020; 21:E746. [PMID: 31979376 PMCID: PMC7037773 DOI: 10.3390/ijms21030746] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 01/17/2020] [Accepted: 01/21/2020] [Indexed: 12/21/2022] Open
Abstract
Melittin is an anti-microbial peptide (AMP) and one of the most studied membrane-disrupting peptides. There is, however, a lack of accurate measurements of the concentration-dependent kinetics and affinity of binding of melittin to phospholipid membranes. In this study, we used surface plasmon resonance spectroscopy to determine the concentration-dependent effect on the binding of melittin to 1-palmitoyl-2-oleoyl-glycero-3-phosphocholine (POPC) bilayers in vesicles. Three concentration ranges were considered, and when combined, covered two orders of magnitudes (0.04 µM to 8 µM), corresponding to concentrations relevant to the membrane-disrupting and anti-microbial activities of melittin. Binding kinetics data were analysed using a 1:1 Langmuir-binding model and a two-state reaction model. Using in-depth quantitative analysis, we characterised the effect of peptide concentration, the addition of NaCl at physiological ionic strength and the choice of kinetic binding model on the reliability of the calculated kinetics and affinity of binding parameters. The apparent binding affinity of melittin for POPC bilayers was observed to decrease with increasing peptide/lipid (P/L) ratio, primarily due to the marked decrease in the association rate. At all concentration ranges, the two-state reaction model provided a better fit to the data and, thus, a more reliable estimate of binding affinity. Addition of NaCl significantly reduced the signal response during the association phase; however, no substantial effect on the binding affinity of melittin to the POPC bilayers was observed. These findings based on POPC bilayers could have important implications for our understanding of the mechanism of action of melittin on more complex model cell membranes of higher physiological relevance.
Collapse
Affiliation(s)
- Thiru Sabapathy
- School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, GPO Box U1987, Perth, WA 6845, Australia; (T.S.); (E.D.)
| | - Evelyne Deplazes
- School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, GPO Box U1987, Perth, WA 6845, Australia; (T.S.); (E.D.)
- School of Life Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Ricardo L. Mancera
- School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, GPO Box U1987, Perth, WA 6845, Australia; (T.S.); (E.D.)
| |
Collapse
|
38
|
Memariani H, Memariani M, Moravvej H, Shahidi-Dadras M. Melittin: a venom-derived peptide with promising anti-viral properties. Eur J Clin Microbiol Infect Dis 2020; 39:5-17. [PMID: 31422545 PMCID: PMC7224078 DOI: 10.1007/s10096-019-03674-0] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 08/08/2019] [Indexed: 12/23/2022]
Abstract
Despite tremendous advances in the development of anti-viral therapeutics, viral infections remain a chief culprit accounting for ongoing morbidity and mortality worldwide. Natural products, in particular animal venoms, embody a veritable cornucopia of exotic constituents, suggesting an immensurable source of anti-infective drugs. In this context, melittin, the principal constituent in the venom of the European honeybee Apis mellifera, has been demonstrated to exert anti-cancer, anti-inflammatory, anti-diabetic, anti-infective, and adjuvant properties. To our knowledge, there is no review appertaining to effects of melittin against viruses, prompting us to synopsize experimental investigations on its anti-viral activity throughout the past decades. Accumulating evidence indicates that melittin curbs infectivity of a diverse array of viruses including coxsackievirus, enterovirus, influenza A viruses, human immunodeficiency virus (HIV), herpes simplex virus (HSV), Junín virus (JV), respiratory syncytial virus (RSV), vesicular stomatitis virus (VSV), and tobacco mosaic virus (TMV). However, medication safety, different routes of administrations, and molecular mechanisms behind the anti-viral activity of melittin should be scrutinized in future studies.
Collapse
Affiliation(s)
- Hamed Memariani
- Skin Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mojtaba Memariani
- Skin Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Hamideh Moravvej
- Skin Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
39
|
Lee TH, Hall K, Aguilar MI. The Effect of Charge on Melittin-Induced Changes in Membrane Structure and Morphology. Aust J Chem 2020. [DOI: 10.1071/ch19500] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The binding of melittin to a range of phospholipid bilayers was studied using dual polarisation interferometry and atomic force microscopy. The phospholipid model membranes included zwitterionic dimyristylphosphatidylcholine (DMPC), together with mixtures of DMPC/dimyristylphosphatidylglycerol (DMPG) and DMPC/DMPG/cholesterol. Melittin caused significant disruption on all bilayers, but differences in morphological changes during binding were different on each membrane. Overall, the results demonstrate that the process of membrane disruption follows distinct structural changes for different lipid mixtures irrespective of the strength of binding to the membrane surface.
Collapse
|
40
|
Belluati A, Mikhalevich V, Yorulmaz Avsar S, Daubian D, Craciun I, Chami M, Meier WP, Palivan CG. How Do the Properties of Amphiphilic Polymer Membranes Influence the Functional Insertion of Peptide Pores? Biomacromolecules 2019; 21:701-715. [DOI: 10.1021/acs.biomac.9b01416] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Andrea Belluati
- Department of Chemistry, University of Basel, Mattenstrasse 24a, BPR 1096, 4058 Basel, Switzerland
| | - Viktoria Mikhalevich
- Department of Chemistry, University of Basel, Mattenstrasse 24a, BPR 1096, 4058 Basel, Switzerland
| | - Saziye Yorulmaz Avsar
- Department of Chemistry, University of Basel, Mattenstrasse 24a, BPR 1096, 4058 Basel, Switzerland
| | - Davy Daubian
- Department of Chemistry, University of Basel, Mattenstrasse 24a, BPR 1096, 4058 Basel, Switzerland
| | - Ioana Craciun
- Department of Chemistry, University of Basel, Mattenstrasse 24a, BPR 1096, 4058 Basel, Switzerland
| | - Mohamed Chami
- BioEM Lab, Biozentrum, University of Basel, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Wolfgang P. Meier
- Department of Chemistry, University of Basel, Mattenstrasse 24a, BPR 1096, 4058 Basel, Switzerland
| | - Cornelia G. Palivan
- Department of Chemistry, University of Basel, Mattenstrasse 24a, BPR 1096, 4058 Basel, Switzerland
| |
Collapse
|
41
|
Quantitative accounting of dye leakage and photobleaching in single lipid vesicle measurements: Implications for biomacromolecular interaction analysis. Colloids Surf B Biointerfaces 2019; 182:110338. [DOI: 10.1016/j.colsurfb.2019.06.067] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Revised: 06/08/2019] [Accepted: 06/28/2019] [Indexed: 02/05/2023]
|
42
|
Effects of Peptide Charge, Orientation, and Concentration on Melittin Transmembrane Pores. Biophys J 2019; 114:2865-2874. [PMID: 29925023 DOI: 10.1016/j.bpj.2018.05.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 03/21/2018] [Accepted: 05/02/2018] [Indexed: 11/21/2022] Open
Abstract
Melittin is a short cationic peptide that exerts cytolytic effects on bacterial and eukaryotic cells. Experiments suggest that in zwitterionic membranes, melittin forms transmembrane toroidal pores supported by four to eight peptides. A recently constructed melittin variant with a reduced cationic charge, MelP5, is active at 10-fold lower concentrations. In previous work, we performed molecular dynamics simulations on the microsecond timescale to examine the supramolecular pore structure of a melittin tetramer in zwitterionic and partially anionic membranes. We now extend that study to include the effects of peptide charge, initial orientation, and number of monomers on the pore formation and stabilization processes. Our results show that parallel transmembrane orientations of melittin and MelP5 are more consistent with experimental data. Whereas a MelP5 parallel hexamer forms a large stable pore during the 5-μs simulation time, a melittin hexamer and an octamer are not fully stable, with several monomers dissociating during the simulation time. Interaction-energy analysis shows that this difference in behavior between melittin and MelP5 is not due to stronger electrostatic repulsion between neighboring melittin peptides but to peptide-lipid interactions that disfavor the isolated MelP5 transmembrane monomer. The ability of melittin monomers to diffuse freely in the 1,2-dimyristoyl-SN-glycero-3-phosphocholine membrane leads to dynamic pores with varying molecularity.
Collapse
|
43
|
Li T, Liu Q, Wang D, Li J. Characterization and antimicrobial mechanism of CF-14, a new antimicrobial peptide from the epidermal mucus of catfish. FISH & SHELLFISH IMMUNOLOGY 2019; 92:881-888. [PMID: 31291603 DOI: 10.1016/j.fsi.2019.07.015] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 06/28/2019] [Accepted: 07/05/2019] [Indexed: 06/09/2023]
Abstract
In this study, we identified a novel antibacterial peptide, RIVELTLPRVSVRL-NH2 (named CF-14), derived from the epidermal mucus of catfish and characterized its antimicrobial activity. Analysis of antimicrobial activity and hemolytic activity of CF-14 revealed broad spectrum, high levels of antimicrobial activity and low toxicity to eukaryotic cells. CF-14 remained stable at pH values ranging from 4.0 to 12.0 and remained bioactive when exposed to high temperature. CD analysis indicated that CF-14 forms a random coil in PBS buffer and an α-helical conformation in the membrane-mimetic 2.5% SDS micelle. Additionally, the antibacterial mechanism of CF-14 against Shewanella putrefaciens was investigated. Membrane permeability experiments confirmed that CF-14 could increase cell wall membrane permeability and cause nucleotide leakage. Moreover, observations performed using scanning electron and confocal microscopy indicated that CF-14 could penetrate into the cell membranes of S. putrefaciens and accumulate in bacterial cells, but did not break down cell membranes. Further, electrophoresis analysis demonstrated that CF-14 possesses DNA-binding affinity. The results provide a substantial basis for future application of CF-14, a novel cell-penetrating peptide (CPP) derived from catfish.
Collapse
Affiliation(s)
- Tingting Li
- Key Laboratory of Biotechnology and Bioresources Utilization (Dalian Minzu University), Ministry of Education, Dalian Liaoning, 116029, China.
| | - Quanwei Liu
- College of Food Science and Technology, Bohai University, National & Local Joint Engineering Research Center of Storage, Processing and Safety Control Technology for Fresh Agricultural and Aquatic Products, JinzhouLiaoning, 121013, China.
| | - Dangfeng Wang
- College of Food Science and Technology, Bohai University, National & Local Joint Engineering Research Center of Storage, Processing and Safety Control Technology for Fresh Agricultural and Aquatic Products, JinzhouLiaoning, 121013, China.
| | - Jianrong Li
- College of Food Science and Technology, Bohai University, National & Local Joint Engineering Research Center of Storage, Processing and Safety Control Technology for Fresh Agricultural and Aquatic Products, JinzhouLiaoning, 121013, China.
| |
Collapse
|
44
|
Hammond K, Lewis H, Faruqui N, Russell C, Hoogenboom BW, Ryadnov MG. Helminth Defense Molecules as Design Templates for Membrane Active Antibiotics. ACS Infect Dis 2019; 5:1471-1479. [PMID: 31117348 DOI: 10.1021/acsinfecdis.9b00157] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
A design template for membrane active antibiotics against microbial and tumor cells is described. The template is an amino acid sequence that combines the properties of helminth defense molecules, which are not cytolytic, with the properties of host-defense peptides, which disrupt microbial membranes. Like helminth defense molecules, the template folds into an amphipathic helix in both mammalian host and microbial phospholipid membranes. Unlike these molecules, the template exhibits antimicrobial and anticancer properties that are comparable to those of antimicrobial and anticancer antibiotics. The selective antibiotic activity of the template builds upon a functional synergy between three distinctive faces of the helix, which is in contrast to two faces of membrane-disrupting amphipathic structures. This synergy enables the template to adapt pore formation mechanisms according to the nature of the target membrane, inducing the lysis of microbial and tumor cells.
Collapse
Affiliation(s)
- Katharine Hammond
- National Physical Laboratory, Hampton Road, Teddington TW11 0LW, United Kingdom
- Department of Physics & Astronomy, University College London, Gower Street, London WC1E 6BT, United Kingdom
- London Centre for Nanotechnology, University College London, Gordon Street, London WC1H 0AH, United Kingdom
| | - Helen Lewis
- National Physical Laboratory, Hampton Road, Teddington TW11 0LW, United Kingdom
| | - Nilofar Faruqui
- National Physical Laboratory, Hampton Road, Teddington TW11 0LW, United Kingdom
| | - Craig Russell
- National Physical Laboratory, Hampton Road, Teddington TW11 0LW, United Kingdom
| | - Bart W. Hoogenboom
- Department of Physics & Astronomy, University College London, Gower Street, London WC1E 6BT, United Kingdom
- London Centre for Nanotechnology, University College London, Gordon Street, London WC1H 0AH, United Kingdom
| | - Maxim G. Ryadnov
- National Physical Laboratory, Hampton Road, Teddington TW11 0LW, United Kingdom
- Department of Physics, King’s College London, Strand Lane, London WC2R, United Kingdom
| |
Collapse
|
45
|
Pandidan S, Mechler A. Nano-viscosimetry analysis of the membrane disrupting action of the bee venom peptide melittin. Sci Rep 2019; 9:10841. [PMID: 31346251 PMCID: PMC6658469 DOI: 10.1038/s41598-019-47325-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 07/10/2019] [Indexed: 12/30/2022] Open
Abstract
Melittin is one of the most studied α-helical cationic membrane disrupting peptides. It is the main component of bee venom, however it is considered an antimicrobial peptide for its ability to kill bacteria. Melittin is believed to act by opening large toroidal pores in the plasma membrane of the targeted cells/bacteria, although this is questioned by some authors. Little is known, however, about the molecular mechanism leading to this activity. In this study the mechanism of action of melittin was studied by dye leakage and quartz crystal microbalance fingerprinting analysis in biomimetic model membranes. The results revealed the existence of multiple stages in the membrane disrupting action with characteristic differences between different membrane types. In bacterial-mimetic (charged) lipid mixtures the viscoelastic fingerprints suggest a surface-acting mechanism, whereas in mammalian-mimetic (neutral) membranes melittin appears to penetrate the bilayer already at low concentrations. In domain-forming mixed membranes melittin shows a preference for the domain containing predominantly zwitterionic lipids. The results confirm membrane poration but are inconsistent with the insertion-to-toroidal pore pathway. Therefore hypotheses of the two membrane disrupting pathways were developed, describing the membrane disruption as either surface tension modulation leading to toroidal pore formation, or linear aggregation leading to fissure formation in the membrane.
Collapse
Affiliation(s)
- Sara Pandidan
- La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia
| | - Adam Mechler
- La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia.
| |
Collapse
|
46
|
Abstract
Membrane permeabilizing peptides (MPPs) are as ubiquitous as the lipid bilayer membranes they act upon. Produced by all forms of life, most membrane permeabilizing peptides are used offensively or defensively against the membranes of other organisms. Just as nature has found many uses for them, translational scientists have worked for decades to design or optimize membrane permeabilizing peptides for applications in the laboratory and in the clinic ranging from antibacterial and antiviral therapy and prophylaxis to anticancer therapeutics and drug delivery. Here, we review the field of membrane permeabilizing peptides. We discuss the diversity of their sources and structures, the systems and methods used to measure their activities, and the behaviors that are observed. We discuss the fact that "mechanism" is not a discrete or a static entity for an MPP but rather the result of a heterogeneous and dynamic ensemble of structural states that vary in response to many different experimental conditions. This has led to an almost complete lack of discrete three-dimensional active structures among the thousands of known MPPs and a lack of useful or predictive sequence-structure-function relationship rules. Ultimately, we discuss how it may be more useful to think of membrane permeabilizing peptides mechanisms as broad regions of a mechanistic landscape rather than discrete molecular processes.
Collapse
Affiliation(s)
- Shantanu Guha
- Department of Biochemistry and Molecular Biology Tulane University School of Medicine , New Orleans , Louisiana 70112 , United States
| | - Jenisha Ghimire
- Department of Biochemistry and Molecular Biology Tulane University School of Medicine , New Orleans , Louisiana 70112 , United States
| | - Eric Wu
- Department of Biochemistry and Molecular Biology Tulane University School of Medicine , New Orleans , Louisiana 70112 , United States
| | - William C Wimley
- Department of Biochemistry and Molecular Biology Tulane University School of Medicine , New Orleans , Louisiana 70112 , United States
| |
Collapse
|
47
|
Miyazaki Y, Okazaki S, Shinoda W. Free energy analysis of membrane pore formation process in the presence of multiple melittin peptides. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2019; 1861:1409-1419. [PMID: 30885804 DOI: 10.1016/j.bbamem.2019.03.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 03/01/2019] [Accepted: 03/04/2019] [Indexed: 11/30/2022]
Abstract
Understanding the molecular mechanism underlying pore formation in lipid membranes by antimicrobial peptides is of great importance in biological sciences as well as in drug design applications. Melittin has been widely studied as a pore forming peptide, though the molecular mechanism for pore formation is still illusive. We examined the free energy barrier for the creation of a pore in lipid membranes with and without multiple melittin peptides. It was found that six melittin peptides significantly stabilized a pore, though a small barrier (a few kBT) for the formation still existed. With five melittin peptides or fewer, the pore formation barrier was much higher, though the established pore was in a local energy minimum. Although seven melittins effectively reduced the free energy barrier, a single melittin peptide left the pore after a long time MD simulation probably because of the overcrowded environment around the bilayer pore. Thus, it is highly selective for the number of melittin peptides to stabilize the membrane pore, as was also suggested by the line tension evaluations. The free energy cost required to insert a single melittin into the membrane is too high to explain the one-by-one insertion mechanism for pore formation, which also supports the collective melittin mechanism for pore formation.
Collapse
Affiliation(s)
- Yusuke Miyazaki
- Department of Materials Chemistry, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8603, Japan
| | - Susumu Okazaki
- Department of Materials Chemistry, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8603, Japan
| | - Wataru Shinoda
- Department of Materials Chemistry, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8603, Japan.
| |
Collapse
|
48
|
Memariani H, Memariani M, Shahidi-Dadras M, Nasiri S, Akhavan MM, Moravvej H. Melittin: from honeybees to superbugs. Appl Microbiol Biotechnol 2019; 103:3265-3276. [DOI: 10.1007/s00253-019-09698-y] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 01/11/2019] [Accepted: 02/11/2019] [Indexed: 02/08/2023]
|
49
|
Tissera MJE, Disalvo EA, Martini MF, Cutró AC. Filling gaps in the knowledge of melittin on lipid membranes. Colloids Surf A Physicochem Eng Asp 2019. [DOI: 10.1016/j.colsurfa.2018.10.055] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
50
|
Awasthi N, Kopec W, Wilkosz N, Jamróz D, Hub JS, Zatorska M, Petka R, Nowakowska M, Kepczynski M. Molecular Mechanism of Polycation-Induced Pore Formation in Biomembranes. ACS Biomater Sci Eng 2018; 5:780-794. [PMID: 33405839 DOI: 10.1021/acsbiomaterials.8b01495] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Polycations are an attractive class of macromolecules with promising applications as drug/gene carriers and biocides. The chemical structure and concentration of a polycation determine its interaction with cellular membranes and, hence, are crucial parameters for designing efficient nontoxic polycations. However, the interaction of polycations with biomembranes at the molecular level and the corresponding free-energy landscape is not well understood. In this work, we investigate the molecular mechanism of interaction between a strong polycation substituted with alkyl moieties and zwitterionic membranes via long-time-scale all-atom molecular dynamics simulations and free-energy calculations combined with Langmuir monolayer, atomic force microscopy, and calcein-release experimental measurements. We found that the membrane activity of the polycation and its ability to induce pores in the membranes can be attributed to the polycation-induced changes in the bilayer organization, such as reduced membrane thickness, increased disorder of the acyl chains, reduced packing, and electrostatic field gradients between membrane leaflets. These changes facilitate the penetration of water into the membrane and the formation of aqueous defects/pores. The calculated free-energy profiles indicate that the polycation lowers the nucleation barrier for pore opening and the free energy for pore formation in a concentration-dependent manner. Above the critical coverage of the membrane, the polycation nucleates spontaneous pores in zwitterionic membranes. Our work demonstrates the potential of combining enhanced sampling methods in MD simulations with experiments for a quantitative description of various events in the polycation-membrane interaction cycle, such as strong adsorption on the membrane due to hydrophobic and electrostatic interactions, and pore formation.
Collapse
Affiliation(s)
- Neha Awasthi
- Institute for Microbiology and Genetics, Georg-August-Universität, Justus-von-Liebig Weg 11, 37077 Göttingen, Germany
| | - Wojciech Kopec
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Kraków, Poland
| | - Natalia Wilkosz
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Kraków, Poland
| | - Dorota Jamróz
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Kraków, Poland
| | - Jochen S Hub
- Institute for Microbiology and Genetics, Georg-August-Universität, Justus-von-Liebig Weg 11, 37077 Göttingen, Germany
| | - Maria Zatorska
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Kraków, Poland
| | - Rafał Petka
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Kraków, Poland
| | - Maria Nowakowska
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Kraków, Poland
| | - Mariusz Kepczynski
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Kraków, Poland
| |
Collapse
|