1
|
Sharifian Gh M, Norouzi F, Sorci M, Zaidi TS, Pier GB, Achimovich A, Ongwae GM, Liang B, Ryan M, Lemke M, Belfort G, Gadjeva M, Gahlmann A, Pires MM, Venter H, Harris TE, Laurie GW. Lacritin cleavage-potentiated targeting of iron - respiratory reciprocity promotes bacterial death. J Biol Chem 2025; 301:108455. [PMID: 40154612 DOI: 10.1016/j.jbc.2025.108455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/15/2025] [Accepted: 03/20/2025] [Indexed: 04/01/2025] Open
Abstract
Discovering new bacterial signaling pathways offers unique antibiotic strategies. With current antibiotic classes targeting cell wall synthesis, depolarizing the inner membrane, altering the bacterial metabolome or inhibiting replication or transcription pathways, manipulation of transporters to limit bacterial respiration and thereby pathogenesis has been a decades-long quest. Here we report an inhibitor of multiple bacterial transporters. The inhibitor is the bactericidal N-104 endogenous cleavage fragment of the prosecretory mitogen lacritin. Lacritin is now known to be widely distributed in plasma, cerebral spinal fluid, tears, and saliva. With the bactericidal mechanism determined to be nonlytic by surface plasmon resonance as confirmed by lack of SYTOX Orange entry, we performed an unbiased resistance screen of 3884 Escherichia coli gene knockout strains revealing a complex N-104 polypharmacology. Validation in the virulent Pseudomonas aeruginosa strain PA14-one of three WHO Priority 1: Critical list species-focused on an approach that sequentially couples three transporters and downstream transcription to lethally suppress respiration. By targeting the outer membrane YaiW, cationic N-104 translocates into the periplasm where it ligates inner membrane transporters FeoB and PotH, respectively, to suppress both ferrous iron and polyamine uptake. With FeoB favoring an anaerobic environment, N-104 promotes the expression of genes regulating anaerobic respiration while largely suppressing those involved in aerobic respiration-a strategy counterproductive under aerobic conditions. This mechanism is innate to the surface of the eye and is enhanced by synergistic coupling with tear thrombin fragment GKY20 as tested on antibiotic-resistant clinical isolates.
Collapse
Affiliation(s)
| | - Fatemeh Norouzi
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia, USA
| | - Mirco Sorci
- Howard P. Isermann Department of Chemical and Biological Engineering and the Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York, USA
| | - Tanweer S Zaidi
- Division of Infectious Disease, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Gerald B Pier
- Division of Infectious Disease, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Alecia Achimovich
- Department of Chemistry, University of Virginia, Charlottesville, Virginia, USA
| | - George M Ongwae
- Department of Chemistry, University of Virginia, Charlottesville, Virginia, USA
| | - Binyong Liang
- Department of Physiology, University of Virginia, Charlottesville, Virginia, USA
| | - Margaret Ryan
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia, USA
| | - Michael Lemke
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia, USA
| | - Georges Belfort
- Howard P. Isermann Department of Chemical and Biological Engineering and the Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York, USA
| | - Mihaela Gadjeva
- Division of Infectious Disease, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Andreas Gahlmann
- Department of Chemistry, University of Virginia, Charlottesville, Virginia, USA
| | - Marcos M Pires
- Department of Chemistry, University of Virginia, Charlottesville, Virginia, USA
| | - Henrietta Venter
- Sansom Institute for Health Research, University of South Australia, Adelaide, Australia
| | - Thurl E Harris
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia, USA
| | - Gordon W Laurie
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia, USA; Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA; Department of Ophthalmology, University of Virginia, Charlottesville, Virginia, USA.
| |
Collapse
|
2
|
Pandidan S, Mechler A. Nano-viscosimetry analysis of membrane disrupting peptide magainin2 interactions with model membranes. Biophys Chem 2025; 318:107390. [PMID: 39798207 DOI: 10.1016/j.bpc.2025.107390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 01/08/2025] [Accepted: 01/08/2025] [Indexed: 01/15/2025]
Abstract
The rapid spread of antibiotic-resistant strains of bacteria has created an urgent need for new alternative antibiotic agents. Membrane disrupting antimicrobial peptides (AMPs): short amino acid sequences with bactericidal and fungicidal activity that kill pathogens by permeabilizing their plasma membrane may offer a solution for this global health crisis. Magainin 2 is an AMP secreted by the African clawed frog (Xenopus laevis) that is described as a toroidal pore former membrane disrupting AMP. Magainin 2 is one of the most thoroughly studied AMPs, yet its mechanism of action is still largely hypothetical: visual evidence of the pore formation is lacking, and the molecular mechanism leading to pore formation is still debated. In the present study, quartz crystal microbalance (QCM) based viscoelastic fingerprinting analysis supported by dye leakage experiments and atomic force microscopy (AFM) imaging was used to glean deeper insights into the mechanism of action. The effect of membrane charge, acyl chain unsaturation and cholesterol concentration were also investigated. The results show lipid specific disruptive mechanism of magainin 2. QCM nano-viscometry measurements revealed the presence of distinct stages in the mechanism of magainin 2 action that, with dye leakage data, confirm the existence of an initial transient pore stage that may result in peptide flip-flop between the outer and inner membrane leaflets. There is evidence of a further mechanistic stage at high peptide concentrations that is consistent with membrane collapse into a peptide-lipid mixed phase that is distinct from the transient pore formation. The results confirm some of the earliest hypotheses about magainin 2 action, while also highlighting the membrane modulating effect of this peptide.
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
|
3
|
Le Han H, Pham PTV, Kim SG, Chan SS, Khoo KS, Chew KW, Show PL, Tran TNT, Nguyen HTV, Nguyen PTD. Isolation and Characterization of Antimicrobial Peptides Isolated from Brevibacillus halotolerans 7WMA2 for the Activity Against Multidrug-Resistant Pathogens. Mol Biotechnol 2024; 66:3618-3627. [PMID: 38042757 DOI: 10.1007/s12033-023-00963-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 10/26/2023] [Indexed: 12/04/2023]
Abstract
Multidrug resistance to pathogens has posed a severe threat to public health. The threat could be addressed by antimicrobial peptides (AMPs) with broad-spectrum suppression. In this study, Brevibacillus halotolerans 7WMA2, isolated from marine sediment, produced AMPs against Gram-positive and Gram-negative bacteria. The AMPs were precipitated by ammonium sulfate 30% (w/v) from culture broth and dialyzed by a 1 kDa membrane. Tryptone Soy Agar (TSA) was used for the cultivation and resulted in the largest bacteria-inhibiting zones under aerobic conditions at 25 °C, 48 h. An SDS-PAGE gel overlay test revealed that strain 7WMA2 could produce AMPs of 5-10 kDa and showed no degradation when held at 121 °C for 30 min at a wide pH 2-12 range. The AMPs did not cause toxicity to HeLa cells with concentrations up to 500 µg/mL while increasing the arbitrary unit up to eight times. The study showed that the AMPs produced were unique, with broad-spectrum antimicrobial ability.
Collapse
Affiliation(s)
- Ho Le Han
- The University of Danang, University of Science and Technology, 54 Nguyen Luong Bang St., Danang, 550000, Viet Nam.
| | | | - Song-Gun Kim
- Biological Resource Center/Korean Collection for Type Cultures (KCTC), Korea Research Institute of Bioscience and Biotechnology, Jeongeup, 56212, Republic of Korea
- University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
| | - Sook Sin Chan
- Institut Sains Biologi, Fakulti Sains, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Kuan Shiong Khoo
- Department of Chemical Engineering and Material Science, Yuan Ze University, Taoyuan, Taiwan.
| | - Kit Wayne Chew
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 62 Nanyang Drive, Singapore, 637459, Singapore
| | - Pau Loke Show
- Department of Chemical Engineering, Khalifa University, Shakhbout Bin Sultan St, Zone 1, Abu Dhabi, United Arab Emirates
| | - Thi Ngoc Thu Tran
- The University of Danang, University of Technology and Education, 48 Cao Thang st, Danang, 550000, Viet Nam
| | | | - Phuong Thi Dong Nguyen
- The University of Danang, University of Technology and Education, 48 Cao Thang st, Danang, 550000, Viet Nam.
| |
Collapse
|
4
|
Simm C, Lee TH, Weerasinghe H, Walsh D, Nakou IT, Shankar M, Tse WC, Zhang Y, Inman R, Mulder RJ, Harrison F, Aguilar MI, Challis GL, Traven A. Gladiolin produced by pathogenic Burkholderia synergizes with amphotericin B through membrane lipid rearrangements. mBio 2024; 15:e0261124. [PMID: 39422464 PMCID: PMC11559049 DOI: 10.1128/mbio.02611-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024] Open
Abstract
Amphotericin B (AmpB) is an effective but toxic antifungal drug. Thus, improving its activity/toxicity relationship is of interest. AmpB disrupts fungal membranes by two proposed mechanisms: ergosterol sequestration from the membrane and pore formation. Whether these two mechanisms operate in conjunction and how they could be potentiated remains to be fully understood. Here, we report that gladiolin, a polyketide antibiotic produced by Burkholderia gladioli, is a strong potentiator of AmpB and acts synergistically against Cryptococcus and Candida species, including drug-resistant C. auris. Gladiolin also synergizes with AmpB against drug-resistant fungal biofilms, while exerting no mammalian cytotoxicity. To explain the mechanism of synergy, we show that gladiolin interacts with membranes via a previously unreported binding mode for polyketides. Moreover, gladiolin modulates lipid binding by AmpB and, in combination, causes faster and more pronounced lipid rearrangements relative to AmpB alone which include membrane thinning consistent with ergosterol extraction, areas of thickening, pore formation, and increased membrane destruction. These biophysical data provide evidence of a functional interaction between gladiolin and AmpB at the membrane interface. The data further indicate that the two proposed AmpB mechanisms (ergosterol sequestration and pore formation) act in conjunction to disrupt membranes, and that gladiolin synergizes by enhancing both mechanisms. Collectively, our findings shed light on AmpB's mechanism of action and characterize gladiolin as an AmpB potentiator, showing an antifungal mechanism distinct from its proposed antibiotic activity. We shed light on the synergistic mechanism at the membrane, and provide insights into potentiation strategies to improve AmpB's activity/toxicity relationship. IMPORTANCE Amphotericin B (AmpB) is one of the oldest antifungal drugs in clinical use. It is an effective therapeutic, but it comes with toxicity issues due to the similarities between its fungal target (the membrane lipid ergosterol) and its mammalian counterpart (cholesterol). One strategy to improve its activity/toxicity relationship is by combinatorial therapy with potentiators, which would enable a lower therapeutic dose of AmpB. Here, we report on the discovery of the antibiotic gladiolin as a potentiator of AmpB against several priority human fungal pathogens and fungal biofilms, with no increased toxicity against mammalian cells. We show that gladiolin potentiates AmpB by increasing and accelerating membrane damage. Our findings also provide insights into the on-going debate about the mechanism of action of AmpB by indicating that both proposed mechanisms, extraction of ergosterol from membranes and pore formation, are potentiated by gladiolin.
Collapse
Affiliation(s)
- Claudia Simm
- Department of Biochemistry and Molecular Biology and the Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Centre to Impact AMR, Monash University, Clayton, Victoria, Australia
| | - Tzong-Hsien Lee
- Department of Biochemistry and Molecular Biology and the Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Harshini Weerasinghe
- Department of Biochemistry and Molecular Biology and the Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Centre to Impact AMR, Monash University, Clayton, Victoria, Australia
| | - Dean Walsh
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
| | - Ioanna T. Nakou
- Department of Chemistry, University of Warwick, Coventry, United Kingdom
| | - Madhu Shankar
- Department of Biochemistry and Molecular Biology and the Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Centre to Impact AMR, Monash University, Clayton, Victoria, Australia
| | - Wai Chung Tse
- School of Medicine, Monash University, Clayton, Victoria, Australia
| | - Yu Zhang
- Department of Chemistry, University of Warwick, Coventry, United Kingdom
| | - Rebecca Inman
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
| | - Roger J. Mulder
- CSIRO Manufacturing, Research Way, Clayton, Victoria, Australia
| | - Freya Harrison
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
| | - Marie-Isabel Aguilar
- Department of Biochemistry and Molecular Biology and the Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Gregory L. Challis
- Department of Biochemistry and Molecular Biology and the Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Department of Chemistry, University of Warwick, Coventry, United Kingdom
- ARC Centre of Excellence for Innovations in Peptide and Protein Science, Monash University, Clayton, Victoria, Australia
| | - Ana Traven
- Department of Biochemistry and Molecular Biology and the Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Centre to Impact AMR, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
5
|
Gamage YI, Pan J. Nanoscale Perturbations of Lipid Bilayers Induced by Magainin 2: Insights from AFM Imaging and Force Spectroscopy. Chem Phys Lipids 2024; 263:105421. [PMID: 39067642 DOI: 10.1016/j.chemphyslip.2024.105421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/13/2024] [Accepted: 07/25/2024] [Indexed: 07/30/2024]
Abstract
This study explores the impact of the antimicrobial peptide magainin 2 (Mag2) on lipid bilayers with varying compositions. We employed high-resolution atomic force microscopy (AFM) to reveal a dynamic spectrum of structural changes induced by Mag2. Our AFM imaging unveiled distinct structural alterations in zwitterionic POPC bilayers upon Mag2 exposure, notably the formation of nanoscale depressions within the bilayer surface, which we term as "surface pores" to differentiate them from transmembrane pores. These surface pores are characterized by a limited depth that does not appear to fully traverse the bilayer and reach the opposing leaflet. Additionally, our AFM-based force spectroscopy investigation on POPC bilayers revealed a reduction in bilayer puncture force (FP) and Young's modulus (E) upon Mag2 interaction, indicating a weakening of bilayer stability and increased flexibility, which may facilitate peptide insertion. The inclusion of anionic POPG into POPC bilayers elucidated its modulatory effects on Mag2 activity, highlighting the role of lipid composition in peptide-bilayer interactions. In contrast to surface pores, Mag2 treatment of E. coli total lipid extract bilayers resulted in increased surface roughness, which we describe as a fluctuation-like morphology. We speculate that the weaker cohesive interactions between heterogeneous lipids in E. coli bilayers may render them more susceptible to Mag2-induced perturbations. This could lead to widespread disruptions manifested as surface fluctuations throughout the bilayer, rather than the formation of well-defined pores. Together, our findings of nanoscale bilayer perturbations provide useful insights into the molecular mechanisms governing Mag2-membrane interactions.
Collapse
Affiliation(s)
| | - Jianjun Pan
- Department of Physics, University of South Florida, Tampa, FL 33620, United States.
| |
Collapse
|
6
|
Lee TH, Charchar P, Separovic F, Reid GE, Yarovsky I, Aguilar MI. The intricate link between membrane lipid structure and composition and membrane structural properties in bacterial membranes. Chem Sci 2024; 15:3408-3427. [PMID: 38455013 PMCID: PMC10915831 DOI: 10.1039/d3sc04523d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 01/26/2024] [Indexed: 03/09/2024] Open
Abstract
It is now evident that the cell manipulates lipid composition to regulate different processes such as membrane protein insertion, assembly and function. Moreover, changes in membrane structure and properties, lipid homeostasis during growth and differentiation with associated changes in cell size and shape, and responses to external stress have been related to drug resistance across mammalian species and a range of microorganisms. While it is well known that the biomembrane is a fluid self-assembled nanostructure, the link between the lipid components and the structural properties of the lipid bilayer are not well understood. This perspective aims to address this topic with a view to a more detailed understanding of the factors that regulate bilayer structure and flexibility. We describe a selection of recent studies that address the dynamic nature of bacterial lipid diversity and membrane properties in response to stress conditions. This emerging area has important implications for a broad range of cellular processes and may open new avenues of drug design for selective cell targeting.
Collapse
Affiliation(s)
- Tzong-Hsien Lee
- Department of Biochemistry and Molecular Biology, Monash University Clayton VIC 3800 Australia
| | - Patrick Charchar
- School of Engineering, RMIT University Melbourne Victoria 3001 Australia
| | - Frances Separovic
- School of Chemistry, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne VIC 3010 Australia
| | - Gavin E Reid
- School of Chemistry, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne VIC 3010 Australia
- Department of Biochemistry and Pharmacology, University of Melbourne Parkville VIC 3010 Australia
| | - Irene Yarovsky
- School of Engineering, RMIT University Melbourne Victoria 3001 Australia
| | - Marie-Isabel Aguilar
- Department of Biochemistry and Molecular Biology, Monash University Clayton VIC 3800 Australia
| |
Collapse
|
7
|
Liu F, Greenwood AI, Xiong Y, Miceli RT, Fu R, Anderson KW, McCallum SA, Mihailescu M, Gross R, Cotten ML. Host Defense Peptide Piscidin and Yeast-Derived Glycolipid Exhibit Synergistic Antimicrobial Action through Concerted Interactions with Membranes. JACS AU 2023; 3:3345-3365. [PMID: 38155643 PMCID: PMC10751773 DOI: 10.1021/jacsau.3c00506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 09/25/2023] [Accepted: 09/29/2023] [Indexed: 12/30/2023]
Abstract
Developing new antimicrobials as alternatives to conventional antibiotics has become an urgent race to eradicate drug-resistant bacteria and to save human lives. Conventionally, antimicrobial molecules are studied independently even though they can be cosecreted in vivo. In this research, we investigate two classes of naturally derived antimicrobials: sophorolipid (SL) esters as modified yeast-derived glycolipid biosurfactants that feature high biocompatibility and low production cost; piscidins, which are host defense peptides (HDPs) from fish. While HDPs such as piscidins target the membrane of pathogens, and thus result in low incidence of resistance, SLs are not well understood on a mechanistic level. Here, we demonstrate that combining SL-hexyl ester (SL-HE) with subinhibitory concentration of piscidins 1 (P1) and 3 (P3) stimulates strong antimicrobial synergy, potentiating a promising therapeutic window. Permeabilization assays and biophysical studies employing circular dichroism, NMR, mass spectrometry, and X-ray diffraction are performed to investigate the mechanism underlying this powerful synergy. We reveal four key mechanistic features underlying the synergistic action: (1) P1/3 binds to SL-HE aggregates, becoming α-helical; (2) piscidin-glycolipid assemblies synergistically accumulate on membranes; (3) SL-HE used alone or bound to P1/3 associates with phospholipid bilayers where it induces defects; (4) piscidin-glycolipid complexes disrupt the bilayer structure more dramatically and differently than either compound alone, with phase separation occurring when both agents are present. Overall, dramatic enhancement in antimicrobial activity is associated with the use of two membrane-active agents, with the glycolipid playing the roles of prefolding the peptide, coordinating the delivery of both agents to bacterial surfaces, recruiting the peptide to the pathogenic membranes, and supporting membrane disruption by the peptide. Given that SLs are ubiquitously and safely used in consumer products, the SL/peptide formulation engineered and mechanistically characterized in this study could represent fertile ground to develop novel synergistic agents against drug-resistant bacteria.
Collapse
Affiliation(s)
- Fei Liu
- Department
of Chemistry, Rensselaer Polytechnic Institute, Troy, New York 12180, United States
| | - Alexander I. Greenwood
- Department
of Applied Science, William & Mary, Williamsburg, Virginia 23185, United States
| | - Yawei Xiong
- Department
of Applied Science, William & Mary, Williamsburg, Virginia 23185, United States
| | - Rebecca T. Miceli
- Department
of Chemistry, Rensselaer Polytechnic Institute, Troy, New York 12180, United States
- Center
for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York 12180, United States
| | - Riqiang Fu
- Center
of Interdisciplinary Magnetic Resonance, National High Magnetic Field Laboratory, Tallahassee, Florida 32310, United States
| | - Kyle W. Anderson
- National
Institute of Standards and Technology, Rockville, Maryland 20850, United States
| | - Scott A. McCallum
- Center
for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York 12180, United States
| | - Mihaela Mihailescu
- Institute
for Bioscience and Biotechnology Research, University of Maryland, Rockville, Maryland 20850, United States
| | - Richard Gross
- Department
of Chemistry, Rensselaer Polytechnic Institute, Troy, New York 12180, United States
- Center
for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York 12180, United States
| | - Myriam L. Cotten
- Department
of Applied Science, William & Mary, Williamsburg, Virginia 23185, United States
| |
Collapse
|
8
|
Mescola A, Ragazzini G, Facci P, Alessandrini A. The potential of AFM in studying the role of the nanoscale amphipathic nature of (lipo)-peptides interacting with lipid bilayers. NANOTECHNOLOGY 2022; 33:432001. [PMID: 35830770 DOI: 10.1088/1361-6528/ac80c9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 07/13/2022] [Indexed: 06/15/2023]
Abstract
Antimicrobial peptides (AMPs) and lipopeptides (LPs) represent very promising molecules to fight resistant bacterial infections due to their broad-spectrum of activity, their first target, i.e. the bacterial membrane, and the rapid bactericidal action. For both types of molecules, the action mechanism starts from the membrane of the pathogen agents, producing a disorganization of their phase structure or the formation of pores of different size altering their permeability. This mechanism of action is based on physical interactions more than on a lock-and-key recognition event and it is difficult for the pathogens to rapidly develop an effective resistance. Very small differences in the sequence of both AMPs and LPs might lead to very different effects on the target membrane. Therefore, a correct understanding of their mechanism of action is required with the aim of developing new synthetic peptides, analogues of the natural ones, with specific and more powerful bactericidal activity. Atomic force microscopy (AFM), with its high resolution and the associated force spectroscopy resource, provides a valuable technique to investigate the reorganization of lipid bilayers exposed to antimicrobial or lipopeptides. Here, we present AFM results obtained by ours and other groups on the action of AMPs and LPs on supported lipid bilayers (SLBs) of different composition. We also consider data obtained by fluorescence microscopy to compare the AFM data with another technique which can be used on different lipid bilayer model systems such as SLBs and giant unilamellar vesicles. The outcomes here presented highlight the powerful of AFM-based techniques in detecting nanoscale peptide-membrane interactions and strengthen their use as an exceptional complementary tool toin vivoinvestigations. Indeed, the combination of these approaches can help decipher the mechanisms of action of different antimicrobials and lipopeptides at both the micro and nanoscale levels, and to design new and more efficient antimicrobial compounds.
Collapse
Affiliation(s)
- Andrea Mescola
- CNR-Nanoscience Institute-S3, Via Campi 213/A, I-41125, Modena, Italy
| | - Gregorio Ragazzini
- Department of Physics, Informatics and Mathematics, University of Modena and Reggio Emilia, Via Campi 213/A, I-41125, Modena, Italy
| | - Paolo Facci
- CNR-Ibf, Via De Marini 6, I-16149, Genova, Italy
| | - Andrea Alessandrini
- CNR-Nanoscience Institute-S3, Via Campi 213/A, I-41125, Modena, Italy
- Department of Physics, Informatics and Mathematics, University of Modena and Reggio Emilia, Via Campi 213/A, I-41125, Modena, Italy
| |
Collapse
|
9
|
Insights into molecular mechanism of action of citrus flavonoids hesperidin and naringin on lipid bilayers using spectroscopic, calorimetric, microscopic and theoretical studies. J Mol Liq 2022. [DOI: 10.1016/j.molliq.2021.118411] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
10
|
Juhl DW, Glattard E, Aisenbrey C, Bechinger B. Antimicrobial peptides: mechanism of action and lipid-mediated synergistic interactions within membranes. Faraday Discuss 2021; 232:419-434. [PMID: 34533138 DOI: 10.1039/d0fd00041h] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Biophysical and structural studies of peptide-lipid interactions, peptide topology and dynamics have changed our view of how antimicrobial peptides insert and interact with membranes. Clearly, both peptides and lipids are highly dynamic, and change and mutually adapt their conformation, membrane penetration and detailed morphology on a local and a global level. As a consequence, peptides and lipids can form a wide variety of supramolecular assemblies in which the more hydrophobic sequences preferentially, but not exclusively, adopt transmembrane alignments and have the potential to form oligomeric structures similar to those suggested by the transmembrane helical bundle model. In contrast, charged amphipathic sequences tend to stay intercalated at the membrane interface. Although the membranes are soft and can adapt, at increasing peptide density they cause pronounced disruptions of the phospholipid fatty acyl packing. At even higher local or global concentrations the peptides cause transient membrane openings, rupture and ultimately lysis. Interestingly, mixtures of peptides such as magainin 2 and PGLa, which are stored and secreted naturally as a cocktail, exhibit considerably enhanced antimicrobial activities when investigated together in antimicrobial assays and also in pore forming experiments applied to biophysical model systems. Our most recent investigations reveal that these peptides do not form stable complexes but act by specific lipid-mediated interactions and the nanoscale properties of phospholipid bilayers.
Collapse
Affiliation(s)
- Dennis W Juhl
- Université de Strasbourg/CNRS, UMR7177, Institut de Chimie, 4, rue Blaise Pascal, 67070 Strasbourg, France.
| | - Elise Glattard
- Université de Strasbourg/CNRS, UMR7177, Institut de Chimie, 4, rue Blaise Pascal, 67070 Strasbourg, France.
| | - Christopher Aisenbrey
- Université de Strasbourg/CNRS, UMR7177, Institut de Chimie, 4, rue Blaise Pascal, 67070 Strasbourg, France.
| | - Burkhard Bechinger
- Université de Strasbourg/CNRS, UMR7177, Institut de Chimie, 4, rue Blaise Pascal, 67070 Strasbourg, France. .,Institut Universitaire de France, France
| |
Collapse
|
11
|
Timmons PB, Hewage CM. Conformation and membrane interaction studies of the potent antimicrobial and anticancer peptide palustrin-Ca. Sci Rep 2021; 11:22468. [PMID: 34789753 PMCID: PMC8599514 DOI: 10.1038/s41598-021-01769-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 11/03/2021] [Indexed: 01/13/2023] Open
Abstract
Palustrin-Ca (GFLDIIKDTGKEFAVKILNNLKCKLAGGCPP) is a host defence peptide with potent antimicrobial and anticancer activities, first isolated from the skin of the American bullfrog Lithobates catesbeianus. The peptide is 31 amino acid residues long, cationic and amphipathic. Two-dimensional NMR spectroscopy was employed to characterise its three-dimensional structure in a 50/50% water/2,2,2-trifluoroethanol-\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$d_{3}$$\end{document}d3 mixture. The structure is defined by an \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$\alpha$$\end{document}α-helix that spans between Ile\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$^{6}$$\end{document}6-Ala\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$^{26}$$\end{document}26, and a cyclic disulfide-bridged domain at the C-terminal end of the peptide sequence, between residues 23 and 29. A molecular dynamics simulation was employed to model the peptide’s interactions with sodium dodecyl sulfate micelles, a widely used bacterial membrane-mimicking environment. Throughout the simulation, the peptide was found to maintain its \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$\alpha$$\end{document}α-helical conformation between residues Ile\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$^{6}$$\end{document}6-Ala\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$^{26}$$\end{document}26, while adopting a position parallel to the surface to micelle, which is energetically-favourable due to many hydrophobic and electrostatic contacts with the micelle.
Collapse
Affiliation(s)
- Patrick B Timmons
- UCD School of Biomolecular and Biomedical Science, UCD Centre for Synthesis and Chemical Biology, UCD Conway Institute, University College Dublin, Dublin 4, Ireland.
| | - Chandralal M Hewage
- UCD School of Biomolecular and Biomedical Science, UCD Centre for Synthesis and Chemical Biology, UCD Conway Institute, University College Dublin, Dublin 4, Ireland
| |
Collapse
|
12
|
Birchenough HL, Jowitt TA. Quartz Crystal Microbalance with Dissipation Monitoring (QCM-D): Preparing Functionalized Lipid Layers for the Study of Complex Protein-Ligand Interactions. Methods Mol Biol 2021; 2263:183-197. [PMID: 33877598 DOI: 10.1007/978-1-0716-1197-5_7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Quartz crystal microbalance with dissipation monitoring (QCM-D) is one of the most widely used techniques for the deposition of lipid layers and provides a useful tool for protein-ligand analysis. By using functionalized lipids, for example, with nitrilotriacetic acid (NTA) or biotin, one can couple a molecule to the surface to investigate ligand interactions. Using lipid layers in this way allows for the analysis of complex binding events such as conformational changes, fibrillation, and hierarchical clustering on the surface, which is difficult to interpret with conventional surface sensor techniques. Deposition of lipids and subsequent molecular interactions are easily monitored using both the frequency and the dissipation, which have distinct features in bilayer formation and make QCM-D the ideal technique to use. Here we describe the formation of biotinylated lipid bilayers using two different types of lipids and the subsequent addition of avidin, which can then be used as a basis for linking biotinylated molecules to the surface. These protocols can be adapted to use other lipid moieties and linking chemistries.
Collapse
Affiliation(s)
- Holly L Birchenough
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Wellcome Trust Centre for Cell Matrix Research, University of Manchester, Manchester, UK
| | - Thomas A Jowitt
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
- Wellcome Trust Centre for Cell Matrix Research, University of Manchester, Manchester, UK.
| |
Collapse
|
13
|
Lee TH, Hofferek V, Sani MA, Separovic F, Reid GE, Aguilar MI. The impact of antibacterial peptides on bacterial lipid membranes depends on stage of growth. Faraday Discuss 2021; 232:399-418. [DOI: 10.1039/d0fd00052c] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Impact of maculatin 1.1 on supported lipid bilayers (SLBs) derived from early growth phase (EGP) or stationary growth phase (SGP) E. coli lipid extracts, monitored by atomic force microscopy which images bilayer morphology in real time.
Collapse
Affiliation(s)
- Tzong-Hsien Lee
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia
| | - Vinzenz Hofferek
- School of Chemistry, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, VIC 3010, Australia
| | - Marc-Antoine Sani
- School of Chemistry, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, VIC 3010, Australia
| | - Frances Separovic
- School of Chemistry, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, VIC 3010, Australia
| | - Gavin E. Reid
- School of Chemistry, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, VIC 3010, Australia
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Marie-Isabel Aguilar
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia
| |
Collapse
|
14
|
Nielsen JE, Prévost SF, Jenssen H, Lund R. Impact of antimicrobial peptides on E. coli-mimicking lipid model membranes: correlating structural and dynamic effects using scattering methods. Faraday Discuss 2021; 232:203-217. [DOI: 10.1039/d0fd00046a] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Using X-rays and neutrons we address the effect of AMPs on structure and dynamics of lipids in bacterial model membranes.
Collapse
Affiliation(s)
| | | | - Håvard Jenssen
- Department of Science and Environment, Roskilde University, 4000 Roskilde, Denmark
| | - Reidar Lund
- Department of Chemistry, University of Oslo, 0315 Oslo, Norway
| |
Collapse
|
15
|
Bechinger B, Juhl DW, Glattard E, Aisenbrey C. Revealing the Mechanisms of Synergistic Action of Two Magainin Antimicrobial Peptides. FRONTIERS IN MEDICAL TECHNOLOGY 2020; 2:615494. [PMID: 35047895 PMCID: PMC8757784 DOI: 10.3389/fmedt.2020.615494] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 11/30/2020] [Indexed: 12/18/2022] Open
Abstract
The study of peptide-lipid and peptide-peptide interactions as well as their topology and dynamics using biophysical and structural approaches have changed our view how antimicrobial peptides work and function. It has become obvious that both the peptides and the lipids arrange in soft supramolecular arrangements which are highly dynamic and able to change and mutually adapt their conformation, membrane penetration, and detailed morphology. This can occur on a local and a global level. This review focuses on cationic amphipathic peptides of the magainin family which were studied extensively by biophysical approaches. They are found intercalated at the membrane interface where they cause membrane thinning and ultimately lysis. Interestingly, mixtures of two of those peptides namely magainin 2 and PGLa which occur naturally as a cocktail in the frog skin exhibit synergistic enhancement of antimicrobial activities when investigated together in antimicrobial assays but also in biophysical experiments with model membranes. Detailed dose-response curves, presented here for the first time, show a cooperative behavior for the individual peptides which is much increased when PGLa and magainin are added as equimolar mixture. This has important consequences for their bacterial killing activities and resistance development. In membranes that carry unsaturations both peptides align parallel to the membrane surface where they have been shown to arrange into mesophases involving the peptides and the lipids. This supramolecular structuration comes along with much-increased membrane affinities for the peptide mixture. Because this synergism is most pronounced in membranes representing the bacterial lipid composition it can potentially be used to increase the therapeutic window of pharmaceutical formulations.
Collapse
Affiliation(s)
- Burkhard Bechinger
- University of Strasbourg/CNRS, UMR7177, Institut de Chimie de Strasbourg, Strasbourg, France
- Institut Universitaire de France (IUF), Paris, France
| | - Dennis Wilkens Juhl
- University of Strasbourg/CNRS, UMR7177, Institut de Chimie de Strasbourg, Strasbourg, France
| | - Elise Glattard
- University of Strasbourg/CNRS, UMR7177, Institut de Chimie de Strasbourg, Strasbourg, France
| | - Christopher Aisenbrey
- University of Strasbourg/CNRS, UMR7177, Institut de Chimie de Strasbourg, Strasbourg, France
| |
Collapse
|
16
|
Hammond K, Ryadnov MG, Hoogenboom BW. Atomic force microscopy to elucidate how peptides disrupt membranes. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1863:183447. [PMID: 32835656 DOI: 10.1016/j.bbamem.2020.183447] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/30/2020] [Accepted: 08/13/2020] [Indexed: 12/24/2022]
Abstract
Atomic force microscopy is an increasingly attractive tool to study how peptides disrupt membranes. Often performed on reconstituted lipid bilayers, it provides access to time and length scales that allow dynamic investigations with nanometre resolution. Over the last decade, AFM studies have enabled visualisation of membrane disruption mechanisms by antimicrobial or host defence peptides, including peptides that target malignant cells and biofilms. Moreover, the emergence of high-speed modalities of the technique broadens the scope of investigations to antimicrobial kinetics as well as the imaging of peptide action on live cells in real time. This review describes how methodological advances in AFM facilitate new insights into membrane disruption mechanisms.
Collapse
Affiliation(s)
- Katharine Hammond
- National Physical Laboratory, Hampton Road, Teddington TW11 0LW, UK; London Centre for Nanotechnology, University College London, London WC1H 0AH, UK; Department of Physics & Astronomy, University College London, London WC1E 6BT, UK.
| | - Maxim G Ryadnov
- National Physical Laboratory, Hampton Road, Teddington TW11 0LW, UK; Department of Physics, King's College London, Strand Lane, London WC2R 2LS, UK.
| | - Bart W Hoogenboom
- London Centre for Nanotechnology, University College London, London WC1H 0AH, UK; Department of Physics & Astronomy, University College London, London WC1E 6BT, UK.
| |
Collapse
|
17
|
Houri AJ, Mechler A. Mechanism of Action of the Antimicrobial Peptide Caerin1.1. ChemistrySelect 2020. [DOI: 10.1002/slct.202000851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Aamd J. Houri
- La Trobe Institute for Molecular Science La Trobe University Melbourne VIC 3086 Australia
| | - Adam Mechler
- La Trobe Institute for Molecular Science La Trobe University Melbourne VIC 3086 Australia
| |
Collapse
|
18
|
Hoyo J, Ivanova K, Torrent-Burgues J, Tzanov T. Interaction of Silver-Lignin Nanoparticles With Mammalian Mimetic Membranes. Front Bioeng Biotechnol 2020; 8:439. [PMID: 32457895 PMCID: PMC7225684 DOI: 10.3389/fbioe.2020.00439] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 04/16/2020] [Indexed: 11/13/2022] Open
Abstract
Silver nanoparticles (AgNPs) have broad spectrum antibacterial activity, but their toxicity to human cells has raised concerns related to their use as disinfectants or coatings of medically relevant surfaces. To address this issue, NPs comprising intrinsically bactericidal and biocompatible biopolymer and Ag with high antibacterial efficacy against common pathogens and compatibility to human cells have been engineered. However, the reason for their lower toxicity compared to AgNPs has not yet been elucidated. This work studies the in vitro interaction of AgLNPs with model mammalian membranes through two approaches: (i) Langmuir films and (ii) supported planar bilayers studied by quartz crystal microbalance and atomic force spectroscopy. These approaches elucidate the interactions of AgLNPs with the model membranes indicating a prominent effect of the bioresourced lignin to facilitate the binding of AgLNPs to the mammalian membrane, without penetrating through it. This study opens a new avenue for engineering of hybrid antimicrobial biopolymer – Ag or other metal NPs with improved bactericidal effect whereas maintaining good biocompatibility.
Collapse
Affiliation(s)
- Javier Hoyo
- Grup de Biotecnologia Molecular i Industrial, Department of Chemical Engineering, Universitat Politècnica de Catalunya, Terrasa, Spain
| | - Kristina Ivanova
- Grup de Biotecnologia Molecular i Industrial, Department of Chemical Engineering, Universitat Politècnica de Catalunya, Terrasa, Spain
| | - Juan Torrent-Burgues
- Grup de Biotecnologia Molecular i Industrial, Department of Chemical Engineering, Universitat Politècnica de Catalunya, Terrasa, Spain
| | - Tzanko Tzanov
- Grup de Biotecnologia Molecular i Industrial, Department of Chemical Engineering, Universitat Politècnica de Catalunya, Terrasa, Spain
| |
Collapse
|
19
|
Birchenough HL, Swann MJ, Zindy E, Day AJ, Jowitt TA. Enhanced avidin binding to lipid bilayers using PDP-PE lipids with PEG-biotin linkers. NANOSCALE ADVANCES 2020; 2:1625-1633. [PMID: 36132312 PMCID: PMC9417969 DOI: 10.1039/d0na00060d] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 03/07/2020] [Indexed: 06/15/2023]
Abstract
Two of the most important aspects of lipid bilayers that have increased their popularity in the field of nanotechnology and biosensors are their fluid nature, which is highly beneficial in ensuring the spatial organization of attached molecules, and the relative ease in which they can be manipulated to change the surface chemistry. Here we have used two different types of functionalized lipids to study the interaction of avidin, which is a common approach to attach further ligands for study. We have tested the commonly used Biotinyl-Cap-PE lipids at different molar percentages and reveal that avidin is not evenly distributed, but forms what looks like clusters even at low percentage occupancy which hampers the level of avidin that can be associated with the surface. We have then successfully employed the novel strategy of using PDP-PE lipids which contain a reducible disulphide to which we added maleamide-PEG-biotin spacers of different lengths. There is a more even distribution of avidin on these layers and thereby increasing the amount and efficiency of avidin association. The reduced levels of avidin that was being associated with the Biotinyl-Cap-PE layers as compared to the PDP-PE lipids could be analysed with QCM-D and interferometry approaches, but it was only with SEEC microscopy that the reason for the reduced occupancy was resolved.
Collapse
Affiliation(s)
| | - Marcus J Swann
- Swann Scientific Consulting Ltd 110 Sandy Lane Lymm WA13 9HR UK
| | - Egor Zindy
- Wellcome Trust Centre for Cell-Matrix Research UK
| | | | | |
Collapse
|
20
|
Overton K, Greer HM, Ferguson MA, Spain EM, Elmore DE, Núñez ME, Volle CB. Qualitative and Quantitative Changes to Escherichia coli during Treatment with Magainin 2 Observed in Native Conditions by Atomic Force Microscopy. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2020; 36:650-659. [PMID: 31876422 PMCID: PMC7430157 DOI: 10.1021/acs.langmuir.9b02726] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
The bacterial membrane has been suggested as a good target for future antibiotics, so it is important to understand how naturally occurring antibiotics like antimicrobial peptides (AMPs) disrupt those membranes. The interaction of the AMP magainin 2 (MAG2) with the bacterial cell membrane has been well characterized using supported lipid substrates, unilamellar vesicles, and spheroplasts created from bacterial cells. However, to fully understand how MAG2 kills bacteria, we must consider its effect on the outer membrane found in Gram-negative bacteria. Here, we use atomic force microscopy (AFM) to directly investigate MAG2 interaction with the outer membrane of Escherichia coli and characterize the biophysical consequences of MAG2 treatment under native conditions. While propidium iodide penetration indicates that MAG2 permeabilizes cells within seconds, a corresponding decrease in cellular turgor pressure is not observed until minutes after MAG2 application, suggesting that cellular homeostasis machinery may be responsible for helping the cell maintain turgor pressure despite a loss of membrane integrity. AFM imaging and force measurement modes applied in tandem reveal that the outer membrane becomes pitted, more flexible, and more adhesive after MAG2 treatment. MAG2 appears to have a highly disruptive effect on the outer membrane, extending the known mechanism of MAG2 to the Gram-negative outer membrane.
Collapse
Affiliation(s)
- Kanesha Overton
- Department of Biology , Cottey College , 1000 West Austin Boulevard , Nevada , Missouri 64772 , United States
| | - Helen M Greer
- Department of Biology , Cottey College , 1000 West Austin Boulevard , Nevada , Missouri 64772 , United States
| | - Megan A Ferguson
- Department of Chemistry , State University of New York , 1 Hawk Drive , New Paltz , New York 12561 , United States
| | - Eileen M Spain
- Department of Chemistry , Occidental College , 1600 Campus Road , Los Angeles , California 90041 , United States
| | - Donald E Elmore
- Department of Chemistry and Program in Biochemistry , Wellesley College , 106 Central Street , Wellesley , Massachusetts 02481 , United States
| | - Megan E Núñez
- Department of Chemistry and Program in Biochemistry , Wellesley College , 106 Central Street , Wellesley , Massachusetts 02481 , United States
| | - Catherine B Volle
- Department of Biology , Cottey College , 1000 West Austin Boulevard , Nevada , Missouri 64772 , United States
| |
Collapse
|
21
|
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
|
22
|
Calvelo M, Granja JR, Garcia-Fandino R. Competitive double-switched self-assembled cyclic peptide nanotubes: a dual internal and external control. Phys Chem Chem Phys 2019; 21:20750-20756. [PMID: 31513191 DOI: 10.1039/c9cp02327e] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
"Intelligent" materials based on synthetic small molecules that become functional only under specific conditions provide new opportunities for developing regulated systems aimed at a large number of applications. For instance, biologically active supramolecular entities that are sensitive to environmental conditions, such as the presence of bacterial membranes, are extremely interesting in biomedicine. In this work, we have designed and investigated, using molecular dynamics simulations, a doubly modulable nanotube formed by the self-assembly of cyclic peptides sensitive to both the presence of a lipid membrane and the pH of the aqueous media. The cyclic peptides were designed to self-assemble into peptide nanotubes in the presence of a lipid bilayer and at low pH values. Under these conditions, the residual side chains point outside the cyclic peptides, being exposed to the lipid bilayer, and the inner groups (carboxylic acids) are protonated, thus allowing the permeation of water and preventing that of ions. Higher pH values are expected to create carboxylate groups at the lumen of the peptides, leading to the disassembly of the nanotube, the attraction and translocation of ions towards the hydrophobic core of the bilayer, and eventually killing the target malignant cells. Our results suggest that by introducing a second switch in a membrane sensitive system, it is possible to modulate its interaction with the lipid bilayer. This opens the door to new strategies for the preparation of antimicrobial peptides that interact at the membrane level.
Collapse
Affiliation(s)
- Martín Calvelo
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Química Orgánica, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain.
| | - Juan R Granja
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Química Orgánica, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain.
| | - Rebeca Garcia-Fandino
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Química Orgánica, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain.
| |
Collapse
|
23
|
Juretić D, Simunić J. Design of α-helical antimicrobial peptides with a high selectivity index. Expert Opin Drug Discov 2019; 14:1053-1063. [DOI: 10.1080/17460441.2019.1642322] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
- Davor Juretić
- Mediterranean Institute for Life Sciences, Split, Croatia
- Department of Physics, Faculty of Science, University of Split, Split, Croatia
| | - Juraj Simunić
- Division of molecular biology, Ruđer Bošković Institute, Zagreb, Croatia
| |
Collapse
|
24
|
Aguilar MI. A comment by Prof. Mibel Aguilar-2018 recipient of the Australian Society for Biophysics' McAulay-Hope Prize for Original Biophysics. Biophys Rev 2019; 11:271-272. [PMID: 31041667 DOI: 10.1007/s12551-019-00520-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Accepted: 04/12/2019] [Indexed: 11/30/2022] Open
Affiliation(s)
- Marie-Isabel Aguilar
- Department of Biochemistry and Molecular Biology, Monash University, Wellington Rd, Clayton, Vic, 3800, Australia.
| |
Collapse
|
25
|
Serna N, Sánchez JM, Unzueta U, Sánchez-García L, Sánchez-Chardi A, Mangues R, Vázquez E, Villaverde A. Recruiting potent membrane penetrability in tumor cell-targeted protein-only nanoparticles. NANOTECHNOLOGY 2019; 30:115101. [PMID: 30561375 DOI: 10.1088/1361-6528/aaf959] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The membrane pore-forming activities of the antimicrobial peptide GWH1 have been evaluated in combination with the CXCR4-binding properties of the peptide T22, in self-assembling protein nanoparticles with high clinical potential. The resulting materials, of 25 nm in size and with regular morphologies, show a dramatically improved cell penetrability into CXCR4+ cells (more than 10-fold) and enhanced endosomal escape (the lysosomal degradation dropping from 90% to 50%), when compared with equivalent protein nanoparticles lacking GWH1. These data reveal that GWH1 retains its potent membrane activity in form of nanostructured protein complexes. On the other hand, the specificity of T22 in the CXCR4 receptor binding is subsequently minimized but, unexpectedly, not abolished by the presence of the antimicrobial peptide. The functional combination T22-GWH1 results in 30% of the nanoparticles entering cells via CXCR4 while also exploiting pore-based uptake. Such functional materials are capable to selectively deliver highly potent cytotoxic drugs upon chemical conjugation, promoting CXCR4-dependent cell death. These data support the further development of GWH1-empowered cell-targeted proteins as nanoscale drug carriers for precision medicines. This is a very promising approach to overcome lysosomal degradation of protein nanostructured materials with therapeutic value.
Collapse
Affiliation(s)
- Naroa Serna
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, E-08193 Barcelona, Spain. Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, E-08193 Barcelona, Spain. CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Bellaterra, E-08193 Barcelona, Spain
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Barbosa SC, Nobre TM, Volpati D, Cilli EM, Correa DS, Oliveira ON. The cyclic peptide labaditin does not alter the outer membrane integrity of Salmonella enterica serovar Typhimurium. Sci Rep 2019; 9:1993. [PMID: 30760803 PMCID: PMC6374527 DOI: 10.1038/s41598-019-38551-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 12/27/2018] [Indexed: 12/21/2022] Open
Abstract
Antimicrobial peptides are a promising class of new antibiotics with the ability to kill bacteria by disrupting their cell membrane, which is especially difficult for Gram-negative bacteria whose cell wall contains an outer layer of lipopolysaccharides (LPS). Here we show that the cyclic decapeptide Labaditin (Lo), with proven activity against the Gram-positive Staphylococcus aureus and Streptococcus mutans, is not able to kill the Gram-negative Salmonella enterica serovar Typhimurium (S.e.s. Typhimurium). We found that Lo induced significant changes in the surface pressure isotherms of Langmuir monolayers representing the Salmonella enterica serovar Typhimurium inner membrane (S.e.s. Typhimurium IM), and caused leakage in large unilamellar vesicles made with this IM lipid composition. On the basis of these results one should expect bactericidal activity against S.e.s. Typhimurium. However, Lo could not interact with a monolayer of LPS, causing no significant changes in either the surface pressure isotherms or in the polarization-modulated infrared reflection absorption spectra (PM-IRRAS). Therefore, the failure of Lo to kill S.e.s. Typhimurium is associated with the lack of interaction with LPS from the outer bacteria membrane. Our approach with distinct monolayer compositions and combined techniques to investigate molecular-level interactions is useful for drug design to fight antibiotic-resistant bacteria.
Collapse
Affiliation(s)
- Simone C Barbosa
- São Carlos Institute of Physics, University of São Paulo, CP 369, 13560-970, São Carlos-SP, Brazil
| | - Thatyane M Nobre
- São Carlos Institute of Physics, University of São Paulo, CP 369, 13560-970, São Carlos-SP, Brazil
| | | | - Eduardo M Cilli
- Universidade Estadual Paulista (UNESP), Institute of Chemistry, 14800-060, Araraquara-SP, Brazil
| | - Daniel S Correa
- Nanotechnology National Laboratory for Agriculture (LNNA), Embrapa Instrumentação, 13560-970, São Carlos, SP, Brazil
| | - Osvaldo N Oliveira
- São Carlos Institute of Physics, University of São Paulo, CP 369, 13560-970, São Carlos-SP, Brazil.
| |
Collapse
|
27
|
Artim CM, Brown JS, Alabi CA. Biophysical Characterization of Cationic Antibacterial Oligothioetheramides. Anal Chem 2019; 91:3118-3124. [PMID: 30675774 DOI: 10.1021/acs.analchem.8b05721] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Biophysical analysis into the mechanism of action of membrane-disrupting antibiotics such as antimicrobial peptides (AMPs) and AMP mimetics is necessary to improve our understanding of this promising but relatively untapped class of antibiotics. We evaluate the impact of cationic nature, specifically the presence of guanidine versus amine functional groups using sequence-defined oligothioetheramides (oligoTEAs). Relative to amines, guanidine groups demonstrated improved antibacterial activity against methicillin-resistant Staphylococcus aureus (MRSA). To understand the mechanism of action, we evaluated membrane interactions by performing a propidium iodide assay and fluorescence microscopy of supported MRSA mimetic bilayers treated with oligoTEAs. Both studies demonstrated membrane disruption, while fluorescence microscopy showed the formation of lipid aggregates. We further analyzed the mechanism using surface plasmon resonance with a recently developed two-state binding model with loss. Our biophysical analysis points to the importance of lipid aggregation for antibacterial activity and suggests that guanidine groups improve antibacterial activity by increasing the extent of lipid aggregation. Altogether, these results verify and rationalize the importance of guanidines for enhanced antibacterial activity of oligoTEAs, and present biophysical phenomena for the design and analysis of additional membrane-active antibiotics.
Collapse
Affiliation(s)
- Christine M Artim
- Robert Frederick Smith School of Chemical and Biomolecular Engineering , Ithaca , New York 14853 , United States
| | - Joseph S Brown
- Robert Frederick Smith School of Chemical and Biomolecular Engineering , Ithaca , New York 14853 , United States
| | - Christopher A Alabi
- Robert Frederick Smith School of Chemical and Biomolecular Engineering , Ithaca , New York 14853 , United States
| |
Collapse
|
28
|
Aisenbrey C, Marquette A, Bechinger B. The Mechanisms of Action of Cationic Antimicrobial Peptides Refined by Novel Concepts from Biophysical Investigations. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1117:33-64. [PMID: 30980352 DOI: 10.1007/978-981-13-3588-4_4] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Even 30 years after the discovery of magainins, biophysical and structural investigations on how these peptides interact with membranes can still bear surprises and add new interesting detail to how these peptides exert their antimicrobial action. Early on, using oriented solid-state NMR spectroscopy, it was found that the amphipathic helices formed by magainins are active when being oriented parallel to the membrane surface. More recent investigations indicate that this in-planar alignment is also found when PGLa and magainin in combination exert synergistic pore-forming activities, where studies on the mechanism of synergistic interaction are ongoing. In a related manner, the investigation of dimeric antimicrobial peptide sequences has become an interesting topic of research which bears promise to refine our views how antimicrobial action occurs. The molecular shape concept has been introduced to explain the effects of lipids and peptides on membrane morphology, locally and globally, and in particular of cationic amphipathic helices that partition into the membrane interface. This concept has been extended in this review to include more recent ideas on soft membranes that can adapt to external stimuli including membrane-disruptive molecules. In this manner, the lipids can change their shape in the presence of low peptide concentrations, thereby maintaining the bilayer properties. At higher peptide concentrations, phase transitions occur which lead to the formation of pores and membrane lytic processes. In the context of the molecular shape concept, the properties of lipopeptides, including surfactins, are shortly presented, and comparisons with the hydrophobic alamethicin sequence are made.
Collapse
Affiliation(s)
| | - Arnaud Marquette
- Université de Strasbourg/CNRS, UMR7177, Institut de Chimie, Strasbourg, France
| | - Burkhard Bechinger
- Université de Strasbourg/CNRS, UMR7177, Institut de Chimie, Strasbourg, France. .,Faculté de chimie, Institut le Bel, Strasbourg, France.
| |
Collapse
|
29
|
Brown JS, Mohamed ZJ, Artim CM, Thornlow DN, Hassler JF, Rigoglioso VP, Daniel S, Alabi CA. Antibacterial isoamphipathic oligomers highlight the importance of multimeric lipid aggregation for antibacterial potency. Commun Biol 2018; 1:220. [PMID: 30534612 PMCID: PMC6286309 DOI: 10.1038/s42003-018-0230-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 11/13/2018] [Indexed: 12/02/2022] Open
Abstract
Cationic charge and hydrophobicity have long been understood to drive the potency and selectivity of antimicrobial peptides (AMPs). However, these properties alone struggle to guide broad success in vivo, where AMPs must differentiate bacterial and mammalian cells, while avoiding complex barriers. New parameters describing the biophysical processes of membrane disruption could provide new opportunities for antimicrobial optimization. In this work, we utilize oligothioetheramides (oligoTEAs) to explore the membrane-targeting mechanism of oligomers, which have the same cationic charge and hydrophobicity, yet show a unique ~ 10-fold difference in antibacterial potency. Solution-phase characterization reveals little difference in structure and dynamics. However, fluorescence microscopy of oligomer-treated Staphylococcus aureus mimetic membranes shows multimeric lipid aggregation that correlates with biological activity and helps establish a framework for the kinetic mechanism of action. Surface plasmon resonance supports the kinetic framework and supports lipid aggregation as a driver of antimicrobial function. Joseph Brown et al. use oligothioetheramides (oligo TEAs) to show that multimeric lipid aggregation in Staphylococcus aureus mimetic membranes correlates with the biological activity of oligoTEAs. These results may explain why antimicrobial peptides with identical cationic charge and hydrophobicity show different biological activity.
Collapse
Affiliation(s)
- Joseph S Brown
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, 120 Olin Hall, Cornell University, Ithaca, NY 14853 USA
| | - Zeinab J Mohamed
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, 120 Olin Hall, Cornell University, Ithaca, NY 14853 USA
| | - Christine M Artim
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, 120 Olin Hall, Cornell University, Ithaca, NY 14853 USA
| | - Dana N Thornlow
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, 120 Olin Hall, Cornell University, Ithaca, NY 14853 USA
| | - Joseph F Hassler
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, 120 Olin Hall, Cornell University, Ithaca, NY 14853 USA
| | - Vincent P Rigoglioso
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, 120 Olin Hall, Cornell University, Ithaca, NY 14853 USA
| | - Susan Daniel
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, 120 Olin Hall, Cornell University, Ithaca, NY 14853 USA
| | - Christopher A Alabi
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, 120 Olin Hall, Cornell University, Ithaca, NY 14853 USA
| |
Collapse
|
30
|
Jafari M, Doustdar F, Mehrnejad F. Molecular Self-Assembly Strategy for Encapsulation of an Amphipathic α-Helical Antimicrobial Peptide into the Different Polymeric and Copolymeric Nanoparticles. J Chem Inf Model 2018; 59:550-563. [PMID: 30475620 DOI: 10.1021/acs.jcim.8b00641] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Encapsulation of peptide and protein-based drugs in polymeric nanoparticles is one of the fundamental fields in controlled-release drug delivery systems. The molecular mechanisms of absorption of peptides to the polymeric nanoparticles are still unknown, and there is no precise molecular data on the encapsulation process of peptide and protein-based drugs. Herein, the self-assembly of different polymers and block copolymers with combinations of the various molecular weight of blocks and the effects of resultant polymer and copolymer nanomicelles on the stability of magainin2, an α-helical antimicrobial peptide, were investigated by means of all-atom molecular dynamics (MD) simulation. The micelle forming, morphology of micellar aggregations and changes in the first hydration shell of the micelles during micelles formation were explored as well. The results showed that the peptide binds to the polymer and copolymer micelles and never detaches during the MD simulation time. In general, all polymers and copolymers simultaneously encapsulated the peptide during micelles formation and had the ability to maintain the helical structure of the peptide, whereas the first hydration shell of the peptide remained unchanged. Among the micelles, the polyethylene glycol (PEG) micelles completely encapsulated magainin2 and, surprisingly, the NMR structure of the peptide was perfectly kept during the encapsulation process. The MD results also indicated that the aromatic and basic residues of the peptide strongly interact with polymers/copolymers and play important roles in the encapsulation mechanism. This research will provide a good opportunity in the design of polymer surfaces for drug delivery applications such as controlled-release peptide delivery systems.
Collapse
Affiliation(s)
- Majid Jafari
- Infectious Diseases and Tropical Medicine Research Center , Shahid Beheshti University of Medical Sciences , P.O. Box 1985717443, Tehran , Iran.,Department of Life Science Engineering, Faculty of New Sciences and Technologies , University of Tehran , P.O. Box 14395-1561, Tehran , Iran
| | - Farahnoosh Doustdar
- Infectious Diseases and Tropical Medicine Research Center , Shahid Beheshti University of Medical Sciences , P.O. Box 1985717443, Tehran , Iran.,Department of Microbiology, Faculty of Medicine , Shahid Beheshti University of Medical Sciences , P.O. Box 19839-63113 Tehran , Iran
| | - Faramarz Mehrnejad
- Department of Life Science Engineering, Faculty of New Sciences and Technologies , University of Tehran , P.O. Box 14395-1561, Tehran , Iran
| |
Collapse
|
31
|
Heath GR, Harrison PL, Strong PN, Evans SD, Miller K. Visualization of diffusion limited antimicrobial peptide attack on supported lipid membranes. SOFT MATTER 2018; 14:6146-6154. [PMID: 29999090 DOI: 10.1039/c8sm00707a] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Understanding the mechanism of action of antimicrobial peptides (AMP) is fundamental to the development and design of peptide based antimicrobials. Utilizing fast-scan atomic force microscopy (AFM) we detail the attack of an AMP on both prototypical prokaryotic (DOPC:DOPG) and eukaryotic (DOPC:DOPE) model lipid membranes on the nanoscale and in real time. Previously shown to have a favourable therapeutic index, we study Smp43, an AMP with a helical-hinge-helical topology isolated from the venom of the North African scorpion Scorpio maurus palmatus. We observe the dynamic formation of highly branched defects being supported by 2D diffusion models and further experimental data from liposome leakage assays and quartz crystal microbalance-dissipation (QCM-D) analysis, we propose that Smp43 disrupts these membranes via a common mechanism, which we have termed 'diffusion limited disruption' that encompasses elements of both the carpet model and the expanding pore mechanism.
Collapse
Affiliation(s)
- George R Heath
- Department of Physics and Astronomy, Leeds University, Leeds, LS2 9JT, UK.
| | | | | | | | | |
Collapse
|
32
|
Lee TH, Hirst DJ, Kulkarni K, Del Borgo MP, Aguilar MI. Exploring Molecular-Biomembrane Interactions with Surface Plasmon Resonance and Dual Polarization Interferometry Technology: Expanding the Spotlight onto Biomembrane Structure. Chem Rev 2018; 118:5392-5487. [PMID: 29793341 DOI: 10.1021/acs.chemrev.7b00729] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The molecular analysis of biomolecular-membrane interactions is central to understanding most cellular systems but has emerged as a complex technical challenge given the complexities of membrane structure and composition across all living cells. We present a review of the application of surface plasmon resonance and dual polarization interferometry-based biosensors to the study of biomembrane-based systems using both planar mono- or bilayers or liposomes. We first describe the optical principals and instrumentation of surface plasmon resonance, including both linear and extraordinary transmission modes and dual polarization interferometry. We then describe the wide range of model membrane systems that have been developed for deposition on the chips surfaces that include planar, polymer cushioned, tethered bilayers, and liposomes. This is followed by a description of the different chemical immobilization or physisorption techniques. The application of this broad range of engineered membrane surfaces to biomolecular-membrane interactions is then overviewed and how the information obtained using these techniques enhance our molecular understanding of membrane-mediated peptide and protein function. We first discuss experiments where SPR alone has been used to characterize membrane binding and describe how these studies yielded novel insight into the molecular events associated with membrane interactions and how they provided a significant impetus to more recent studies that focus on coincident membrane structure changes during binding of peptides and proteins. We then discuss the emerging limitations of not monitoring the effects on membrane structure and how SPR data can be combined with DPI to provide significant new information on how a membrane responds to the binding of peptides and proteins.
Collapse
Affiliation(s)
- Tzong-Hsien Lee
- Department of Biochemistry and Molecular Biology and Biomedicine Discovery Institute , Monash University , Clayton , VIC 3800 , Australia
| | - Daniel J Hirst
- Department of Biochemistry and Molecular Biology and Biomedicine Discovery Institute , Monash University , Clayton , VIC 3800 , Australia
| | - Ketav Kulkarni
- Department of Biochemistry and Molecular Biology and Biomedicine Discovery Institute , Monash University , Clayton , VIC 3800 , Australia
| | - Mark P Del Borgo
- Department of Biochemistry and Molecular Biology and Biomedicine Discovery Institute , Monash University , Clayton , VIC 3800 , Australia
| | - Marie-Isabel Aguilar
- Department of Biochemistry and Molecular Biology and Biomedicine Discovery Institute , Monash University , Clayton , VIC 3800 , Australia
| |
Collapse
|
33
|
Marquette A, Bechinger B. Biophysical Investigations Elucidating the Mechanisms of Action of Antimicrobial Peptides and Their Synergism. Biomolecules 2018; 8:E18. [PMID: 29670065 PMCID: PMC6023007 DOI: 10.3390/biom8020018] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 04/13/2018] [Accepted: 04/16/2018] [Indexed: 01/30/2023] Open
Abstract
Biophysical and structural investigations are presented with a focus on the membrane lipid interactions of cationic linear antibiotic peptides such as magainin, PGLa, LL37, and melittin. Observations made with these peptides are distinct as seen from data obtained with the hydrophobic peptide alamethicin. The cationic amphipathic peptides predominantly adopt membrane alignments parallel to the bilayer surface; thus the distribution of polar and non-polar side chains of the amphipathic helices mirror the environmental changes at the membrane interface. Such a membrane partitioning of an amphipathic helix has been shown to cause considerable disruptions in the lipid packing arrangements, transient openings at low peptide concentration, and membrane disintegration at higher peptide-to-lipid ratios. The manifold supramolecular arrangements adopted by lipids and peptides are represented by the 'soft membranes adapt and respond, also transiently' (SMART) model. Whereas molecular dynamics simulations provide atomistic views on lipid membranes in the presence of antimicrobial peptides, the biophysical investigations reveal interesting details on a molecular and supramolecular level, and recent microscopic imaging experiments delineate interesting sequences of events when bacterial cells are exposed to such peptides. Finally, biophysical studies that aim to reveal the mechanisms of synergistic interactions of magainin 2 and PGLa are presented, including unpublished isothermal titration calorimetry (ITC), circular dichroism (CD) and dynamic light scattering (DLS) measurements that suggest that the peptides are involved in liposome agglutination by mediating intermembrane interactions. A number of structural events are presented in schematic models that relate to the antimicrobial and synergistic mechanism of amphipathic peptides when they are aligned parallel to the membrane surface.
Collapse
Affiliation(s)
- Arnaud Marquette
- Université de Strasbourg/CNRS, UMR7177, Institut de Chimie, 4, rue Blaise Pascal, 67070 Strasbourg, France.
| | - Burkhard Bechinger
- Université de Strasbourg/CNRS, UMR7177, Institut de Chimie, 4, rue Blaise Pascal, 67070 Strasbourg, France.
| |
Collapse
|
34
|
Antimicrobial peptides: biochemical determinants of activity and biophysical techniques of elucidating their functionality. World J Microbiol Biotechnol 2018; 34:62. [PMID: 29651655 DOI: 10.1007/s11274-018-2444-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 04/05/2018] [Indexed: 10/17/2022]
Abstract
Antimicrobial peptides (AMPs) have been established over millennia as powerful components of the innate immune system of many organisms. Due to their broad spectrum of activity and the development of host resistance against them being unlikely, AMPs are strong candidates for controlling drug-resistant pathogenic microbial pathogens. AMPs cause cell death through several independent or cooperative mechanisms involving membrane lysis, non-lytic activity, and/or intracellular mechanisms. Biochemical determinants such as peptide length, primary sequence, charge, secondary structure, hydrophobicity, amphipathicity and host cell membrane composition together influence the biological activities of peptides. A number of biophysical techniques have been used in recent years to study the mechanisms of action of AMPs. This work appraises the molecular parameters that determine the biocidal activity of AMPs and overviews their mechanisms of actions and the diverse biochemical, biophysical and microscopy techniques utilised to elucidate these.
Collapse
|
35
|
Lee TH, Sani MA, Overall S, Separovic F, Aguilar MI. Effect of phosphatidylcholine bilayer thickness and molecular order on the binding of the antimicrobial peptide maculatin 1.1. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2018; 1860:300-309. [DOI: 10.1016/j.bbamem.2017.10.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 09/18/2017] [Accepted: 10/08/2017] [Indexed: 01/01/2023]
|
36
|
Enhanced transdermal delivery with less irritation by magainin pore-forming peptide with a N-lauroylsarcosine and sorbitan monolaurate mixture. Drug Deliv Transl Res 2017; 8:54-63. [DOI: 10.1007/s13346-017-0433-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
37
|
Phenylalanine residues act as membrane anchors in the antimicrobial action of Aurein 1.2. Biointerphases 2017; 12:05G605. [PMID: 29078702 DOI: 10.1116/1.4995674] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Aurein 1.2 is a small cationic antimicrobial peptide, one of the shortest peptides that can exert antimicrobial activity at low micromolar concentrations. Aurein 1.2 is a surface acting peptide, following the "carpet" mechanism of thresholded membrane disruption. It is generally assumed that the activity of such cationic α-helical membrane disrupting peptides is charge driven. Here, the authors show that instead of charge interactions, aromatic phenylalanine residues of the Aurein 1.2 sequence facilitate the membrane binding. The activity of the wild type peptide was compared to mutants in which the Phe residues were substituted, singly and in tandem, with alanine. Measurements by quartz crystal microbalance, impedance spectroscopy, and dye leakage experiments demonstrated that single residue mutants retain a much-reduced activity whereas the deletion of both Phe residues prevents membrane disruption entirely. The single residue mutants exhibited an altered mechanism of action, permeabilizing but not dissolving the target membranes. These results offer a new design rule for membrane disrupting peptides with potential pharmacological applications.
Collapse
|
38
|
Booth V, Warschawski DE, Santisteban NP, Laadhari M, Marcotte I. Recent progress on the application of 2H solid-state NMR to probe the interaction of antimicrobial peptides with intact bacteria. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2017; 1865:1500-1511. [PMID: 28844739 DOI: 10.1016/j.bbapap.2017.07.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2017] [Revised: 07/13/2017] [Accepted: 07/25/2017] [Indexed: 10/19/2022]
Abstract
Discoveries relating to innate immunity and antimicrobial peptides (AMPs) granted Bruce Beutler and Jules Hoffmann a Nobel prize in medicine in 2011, and opened up new avenues for the development of therapies against infections, and even cancers. The mechanisms by which AMPs interact with, and ultimately disrupt, bacterial cell membranes is still, to a large extent, incompletely understood. Up until recently, this mechanism was studied using model lipid membranes that failed to reproduce the complexity of molecular interactions present in real cells comprising lipids but also membrane proteins, a cell wall containing peptidoglycan or lipopolysaccharides, and other molecules. In this review, we focus on recent attempts to study, at the molecular level, the interaction between cationic AMPs and intact bacteria, by 2H solid-state NMR. Specifically-labeled lipids allow us to focus on the interaction of AMPs with the heart of the bacterial membrane, and measure the lipid order and its variation upon interaction with various peptides. We will review the important parameters to consider in such a study, and summarize the results obtained in the past 5years on various peptides, in particular aurein 1.2, caerin 1.1, MSI-78 and CA(1-8)M(1-10). This article is part of a Special Issue entitled: Biophysics in Canada, edited by Lewis Kay, John Baenziger, Albert Berghuis and Peter Tieleman.
Collapse
Affiliation(s)
- Valerie Booth
- Department of Biochemistry, Memorial University of Newfoundland, St. John's, NL A1B 3X9, Canada; Department of Physics and Physical Oceanography, Memorial University of Newfoundland, St. John's, NL A1B 3X7, Canada
| | - Dror E Warschawski
- UMR 7099, CNRS - Université Paris Diderot, IBPC, 13 rue Pierre et Marie Curie, F-75005 Paris, France; Department of Chemistry, Université du Québec à Montréal, P.O. Box 8888, Downtown Station, Montréal H3C 3P8, Canada
| | - Nury P Santisteban
- Department of Physics and Physical Oceanography, Memorial University of Newfoundland, St. John's, NL A1B 3X7, Canada
| | - Marwa Laadhari
- Department of Chemistry, Université du Québec à Montréal, P.O. Box 8888, Downtown Station, Montréal H3C 3P8, Canada
| | - Isabelle Marcotte
- Department of Chemistry, Université du Québec à Montréal, P.O. Box 8888, Downtown Station, Montréal H3C 3P8, Canada.
| |
Collapse
|
39
|
Dekan Z, Headey SJ, Scanlon M, Baldo BA, Lee T, Aguilar M, Deuis JR, Vetter I, Elliott AG, Amado M, Cooper MA, Alewood D, Alewood PF. Δ‐Myrtoxin‐Mp1a is a Helical Heterodimer from the Venom of the Jack Jumper Ant that has Antimicrobial, Membrane‐Disrupting, and Nociceptive Activities. Angew Chem Int Ed Engl 2017; 56:8495-8499. [DOI: 10.1002/anie.201703360] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Indexed: 11/07/2022]
Affiliation(s)
- Zoltan Dekan
- Institute for Molecular Bioscience The University of Queensland St Lucia QLD 4072 Australia
| | - Stephen J. Headey
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences Monash University 381 Royal Parade Parkville VIC 3052 Australia
| | - Martin Scanlon
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences Monash University 381 Royal Parade Parkville VIC 3052 Australia
| | - Brian A. Baldo
- Kolling Institute of Medical Research Royal North Shore Hospital of Sydney St. Leonards NSW 2065 Australia
| | - Tzong‐Hsien Lee
- Department of Biochemistry and Molecular Biology Monash University Wellington Rd Clayton Vic 3800 Australia
| | - Marie‐Isabel Aguilar
- Department of Biochemistry and Molecular Biology Monash University Wellington Rd Clayton Vic 3800 Australia
| | - Jennifer R. Deuis
- Institute for Molecular Bioscience The University of Queensland St Lucia QLD 4072 Australia
| | - Irina Vetter
- Institute for Molecular Bioscience The University of Queensland St Lucia QLD 4072 Australia
| | - Alysha G. Elliott
- Institute for Molecular Bioscience The University of Queensland St Lucia QLD 4072 Australia
| | - Maite Amado
- Institute for Molecular Bioscience The University of Queensland St Lucia QLD 4072 Australia
| | - Matthew A. Cooper
- Institute for Molecular Bioscience The University of Queensland St Lucia QLD 4072 Australia
| | - Dianne Alewood
- Institute for Molecular Bioscience The University of Queensland St Lucia QLD 4072 Australia
| | - Paul F. Alewood
- Institute for Molecular Bioscience The University of Queensland St Lucia QLD 4072 Australia
| |
Collapse
|
40
|
Dekan Z, Headey SJ, Scanlon M, Baldo BA, Lee T, Aguilar M, Deuis JR, Vetter I, Elliott AG, Amado M, Cooper MA, Alewood D, Alewood PF. Δ‐Myrtoxin‐Mp1a is a Helical Heterodimer from the Venom of the Jack Jumper Ant that has Antimicrobial, Membrane‐Disrupting, and Nociceptive Activities. Angew Chem Int Ed Engl 2017. [DOI: 10.1002/ange.201703360] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Zoltan Dekan
- Institute for Molecular Bioscience The University of Queensland St Lucia QLD 4072 Australia
| | - Stephen J. Headey
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences Monash University 381 Royal Parade Parkville VIC 3052 Australia
| | - Martin Scanlon
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences Monash University 381 Royal Parade Parkville VIC 3052 Australia
| | - Brian A. Baldo
- Kolling Institute of Medical Research Royal North Shore Hospital of Sydney St. Leonards NSW 2065 Australia
| | - Tzong‐Hsien Lee
- Department of Biochemistry and Molecular Biology Monash University Wellington Rd Clayton Vic 3800 Australia
| | - Marie‐Isabel Aguilar
- Department of Biochemistry and Molecular Biology Monash University Wellington Rd Clayton Vic 3800 Australia
| | - Jennifer R. Deuis
- Institute for Molecular Bioscience The University of Queensland St Lucia QLD 4072 Australia
| | - Irina Vetter
- Institute for Molecular Bioscience The University of Queensland St Lucia QLD 4072 Australia
| | - Alysha G. Elliott
- Institute for Molecular Bioscience The University of Queensland St Lucia QLD 4072 Australia
| | - Maite Amado
- Institute for Molecular Bioscience The University of Queensland St Lucia QLD 4072 Australia
| | - Matthew A. Cooper
- Institute for Molecular Bioscience The University of Queensland St Lucia QLD 4072 Australia
| | - Dianne Alewood
- Institute for Molecular Bioscience The University of Queensland St Lucia QLD 4072 Australia
| | - Paul F. Alewood
- Institute for Molecular Bioscience The University of Queensland St Lucia QLD 4072 Australia
| |
Collapse
|
41
|
Sani MA, Carne S, Overall SA, Poulhazan A, Separovic F. One pathogen two stones: are Australian tree frog antimicrobial peptides synergistic against human pathogens? EUROPEAN BIOPHYSICS JOURNAL: EBJ 2017; 46:639-646. [PMID: 28478484 DOI: 10.1007/s00249-017-1215-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Revised: 02/28/2017] [Accepted: 04/24/2017] [Indexed: 01/10/2023]
Abstract
Antimicrobial peptides (AMPs) may act by targeting the lipid membranes and disrupting the bilayer structure. In this study, three AMPs from the skin of Australian tree frogs, aurein 1.2, maculatin 1.1 and caerin 1.1, were investigated against Gram-negative Escherichia coli, Gram-positive Staphylococcus aureus, and vesicles that mimic their lipid compositions. Furthermore, equimolar mixtures of the peptides were tested to identify any synergistic interactions in antimicrobial activity. Minimum inhibition concentration and minimum bactericidal concentration assays showed significant activity against S. aureus but not against E. coli. Aurein was the least active while maculatin was the most active peptide and some synergistic effects were observed against S. aureus. Circular dichroism experiments showed that, in the presence of phospholipid vesicles, the peptides transitioned from an unstructured to a predominantly helical conformation (>50%), with greater helicity for POPG/TOCL compared to POPE/POPG vesicles. The helical content, however, was less in the presence of live E. coli and S. aureus, 25 and 5%, respectively. Equimolar concentrations of the peptides did not appear to form greater supramolecular structures. Dye release assays showed that aurein required greater concentration than caerin and maculatin to disrupt the lipid bilayers, and mixtures of the peptides did not cooperate to enhance their lytic activity. Overall, aurein, maculatin, and caerin showed moderate synergy in antimicrobial activity against S. aureus without becoming more structured or enhancement of their membrane-disrupting activity in phospholipid vesicles.
Collapse
Affiliation(s)
- Marc-Antoine Sani
- School of Chemistry, Bio21 Institute, University of Melbourne, Melbourne, VIC, 3010, Australia.
| | - Siobhan Carne
- School of Chemistry, Bio21 Institute, University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Sarah A Overall
- School of Chemistry, Bio21 Institute, University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Alexandre Poulhazan
- School of Chemistry, Bio21 Institute, University of Melbourne, Melbourne, VIC, 3010, Australia
- Universite Pierre et Marie Curie (Paris VI), 4 Place Jussieu, 75252, Paris Cedex 5, France
| | - Frances Separovic
- School of Chemistry, Bio21 Institute, University of Melbourne, Melbourne, VIC, 3010, Australia.
| |
Collapse
|
42
|
Handschuh-Wang S, Wang T, Zhou X. Recent advances in hybrid measurement methods based on atomic force microscopy and surface sensitive measurement techniques. RSC Adv 2017. [DOI: 10.1039/c7ra08515j] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
This review summaries the recent progress of the combination of optical and non-optical surface sensitive techniques with the atomic force microscopy.
Collapse
Affiliation(s)
- Stephan Handschuh-Wang
- College of Chemistry and Environmental Engineering
- Shenzhen University
- Shenzhen 518060
- P. R. China
| | - Tao Wang
- Functional Thin Films Research Center
- Shenzhen Institutes of Advanced Technology
- Chinese Academy of Sciences
- Shenzhen 518055
- P. R. China
| | - Xuechang Zhou
- College of Chemistry and Environmental Engineering
- Shenzhen University
- Shenzhen 518060
- P. R. China
| |
Collapse
|
43
|
Francois-Moutal L, Ouberai MM, Maniti O, Welland ME, Strzelecka-Kiliszek A, Wos M, Pikula S, Bandorowicz-Pikula J, Marcillat O, Granjon T. Two-Step Membrane Binding of NDPK-B Induces Membrane Fluidity Decrease and Changes in Lipid Lateral Organization and Protein Cluster Formation. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2016; 32:12923-12933. [PMID: 27934520 DOI: 10.1021/acs.langmuir.6b03789] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Nucleoside diphosphate kinases (NDPKs) are crucial elements in a wide array of cellular physiological or pathophysiological processes such as apoptosis, proliferation, or metastasis formation. Among the NDPK isoenzymes, NDPK-B, a cytoplasmic protein, was reported to be associated with several biological membranes such as plasma or endoplasmic reticulum membranes. Using several membrane models (liposomes, lipid monolayers, and supported lipid bilayers) associated with biophysical approaches, we show that lipid membrane binding occurs in a two-step process: first, initiation by a strong electrostatic adsorption process and followed by shallow penetration of the protein within the membrane. The NDPK-B binding leads to a decrease in membrane fluidity and formation of protein patches. The ability of NDPK-B to form microdomains at the membrane level may be related to protein-protein interactions triggered by its association with anionic phospholipids. Such accumulation of NDPK-B would amplify its effects in functional platform formation and protein recruitment at the membrane.
Collapse
Affiliation(s)
- Liberty Francois-Moutal
- Organisation et Dynamique des Membrane Biologiques, Institut de Chimie et Biochimie Moléculaires et Supramoléculaires, CNRS UMR 5246 ICBMS , Bâtiment Chevreul, 43 Boulevard du 11 Novembre 1918, Villeurbanne Cedex 69622, France
| | - Myriam M Ouberai
- Nanoscience Centre, University of Cambridge , 11 J.J. Thomson Avenue Cambridge, Cambridge CB3 0FF, U.K
| | - Ofelia Maniti
- Organisation et Dynamique des Membrane Biologiques, Institut de Chimie et Biochimie Moléculaires et Supramoléculaires, CNRS UMR 5246 ICBMS , Bâtiment Chevreul, 43 Boulevard du 11 Novembre 1918, Villeurbanne Cedex 69622, France
| | - Mark E Welland
- Nanoscience Centre, University of Cambridge , 11 J.J. Thomson Avenue Cambridge, Cambridge CB3 0FF, U.K
| | - Agnieszka Strzelecka-Kiliszek
- Department of Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences , 3 Pasteur Street, Warsaw 02-093, Poland
| | - Marcin Wos
- Department of Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences , 3 Pasteur Street, Warsaw 02-093, Poland
| | - Slawomir Pikula
- Department of Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences , 3 Pasteur Street, Warsaw 02-093, Poland
| | - Joanna Bandorowicz-Pikula
- Department of Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences , 3 Pasteur Street, Warsaw 02-093, Poland
| | - Olivier Marcillat
- Organisation et Dynamique des Membrane Biologiques, Institut de Chimie et Biochimie Moléculaires et Supramoléculaires, CNRS UMR 5246 ICBMS , Bâtiment Chevreul, 43 Boulevard du 11 Novembre 1918, Villeurbanne Cedex 69622, France
| | - Thierry Granjon
- Organisation et Dynamique des Membrane Biologiques, Institut de Chimie et Biochimie Moléculaires et Supramoléculaires, CNRS UMR 5246 ICBMS , Bâtiment Chevreul, 43 Boulevard du 11 Novembre 1918, Villeurbanne Cedex 69622, France
| |
Collapse
|
44
|
Shahmiri M, Enciso M, Adda CG, Smith BJ, Perugini MA, Mechler A. Membrane Core-Specific Antimicrobial Action of Cathelicidin LL-37 Peptide Switches Between Pore and Nanofibre Formation. Sci Rep 2016; 6:38184. [PMID: 27901075 PMCID: PMC5128859 DOI: 10.1038/srep38184] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 11/04/2016] [Indexed: 11/09/2022] Open
Abstract
Membrane-disrupting antimicrobial peptides provide broad-spectrum defence against localized bacterial invasion in a range of hosts including humans. The most generally held consensus is that targeting to pathogens is based on interactions with the head groups of membrane lipids. Here we show that the action of LL-37, a human antimicrobial peptide switches the mode of action based on the structure of the alkyl chains, and not the head groups of the membrane forming lipids. We demonstrate that LL-37 exhibits two distinct interaction pathways: pore formation in bilayers of unsaturated phospholipids and membrane modulation with saturated phospholipids. Uniquely, the membrane modulation yields helical-rich fibrous peptide-lipid superstructures. Our results point at alternative design strategies for peptide antimicrobials.
Collapse
Affiliation(s)
- Mahdi Shahmiri
- La Trobe Institute for Molecular Science, La Trobe University, Australia
| | - Marta Enciso
- La Trobe Institute for Molecular Science, La Trobe University, Australia
| | - Christopher G Adda
- La Trobe Institute for Molecular Science, La Trobe University, Australia
| | - Brian J Smith
- La Trobe Institute for Molecular Science, La Trobe University, Australia
| | - Matthew A Perugini
- La Trobe Institute for Molecular Science, La Trobe University, Australia
| | - Adam Mechler
- La Trobe Institute for Molecular Science, La Trobe University, Australia
| |
Collapse
|
45
|
Li W, O'Brien-Simpson NM, Tailhades J, Pantarat N, Dawson RM, Otvos L, Reynolds EC, Separovic F, Hossain MA, Wade JD. Multimerization of a Proline-Rich Antimicrobial Peptide, Chex-Arg20, Alters Its Mechanism of Interaction with the Escherichia coli Membrane. ACTA ACUST UNITED AC 2016; 22:1250-8. [PMID: 26384569 DOI: 10.1016/j.chembiol.2015.08.011] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 07/28/2015] [Accepted: 08/07/2015] [Indexed: 10/23/2022]
Abstract
A3-APO, a de novo designed branched dimeric proline-rich antimicrobial peptide (PrAMP), is highly effective against a variety of in vivo bacterial infections. We undertook a selective examination of the mechanism for the Gram-negative Escherichia coli bacterial membrane interaction of the monomer (Chex-Arg20), dimer (A3-APO), and tetramer (A3-APO disulfide-linked dimer). All three synthetic peptides were effective at killing E. coli. However, the tetramer was 30-fold more membrane disruptive than the dimer while the monomer showed no membrane activity. Using flow cytometry and high-resolution fluorescent microscopy, it was observed that dimerization and tetramerization of the Chex-Arg20 monomer led to an alteration in the mechanism of action from non-lytic/membrane hyperpolarization to membrane disruption/depolarization. Our findings show that the membrane interaction and permeability of Chex-Arg20 was altered by multimerization.
Collapse
Affiliation(s)
- Wenyi Li
- School of Chemistry, University of Melbourne, VIC 3010, Australia; The Florey Institute of Neuroscience and Mental Health, University of Melbourne, VIC 3010, Australia
| | - Neil M O'Brien-Simpson
- Oral Health CRC, Melbourne Dental School, University of Melbourne, VIC 3010, Australia; Bio21 Institute, University of Melbourne, VIC 3010, Australia
| | - Julien Tailhades
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, VIC 3010, Australia
| | - Namfon Pantarat
- Oral Health CRC, Melbourne Dental School, University of Melbourne, VIC 3010, Australia
| | - Raymond M Dawson
- Land Division, Defence Science and Technology Organization, Fishermans Bend, VIC 3207, Australia
| | - Laszlo Otvos
- Department of Biology, Temple University, Philadelphia, PA 19122, USA; Institute of Medical Microbiology, Semmelweis University, Budapest 1089, Hungary
| | - Eric C Reynolds
- Oral Health CRC, Melbourne Dental School, University of Melbourne, VIC 3010, Australia; Bio21 Institute, University of Melbourne, VIC 3010, Australia
| | - Frances Separovic
- School of Chemistry, University of Melbourne, VIC 3010, Australia; Bio21 Institute, University of Melbourne, VIC 3010, Australia
| | - Mohammed Akhter Hossain
- School of Chemistry, University of Melbourne, VIC 3010, Australia; The Florey Institute of Neuroscience and Mental Health, University of Melbourne, VIC 3010, Australia.
| | - John D Wade
- School of Chemistry, University of Melbourne, VIC 3010, Australia; The Florey Institute of Neuroscience and Mental Health, University of Melbourne, VIC 3010, Australia.
| |
Collapse
|
46
|
Deuis JR, Dekan Z, Inserra MC, Lee TH, Aguilar MI, Craik DJ, Lewis RJ, Alewood PF, Mobli M, Schroeder CI, Henriques ST, Vetter I. Development of a μO-Conotoxin Analogue with Improved Lipid Membrane Interactions and Potency for the Analgesic Sodium Channel NaV1.8. J Biol Chem 2016; 291:11829-42. [PMID: 27026701 DOI: 10.1074/jbc.m116.721662] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Indexed: 12/19/2022] Open
Abstract
The μO-conotoxins MrVIA, MrVIB, and MfVIA inhibit the voltage-gated sodium channel NaV1.8, a well described target for the treatment of pain; however, little is known about the residues or structural elements that define this activity. In this study, we determined the three-dimensional structure of MfVIA, examined its membrane binding properties, performed alanine-scanning mutagenesis, and identified residues important for its activity at human NaV1.8. A second round of mutations resulted in (E5K,E8K)MfVIA, a double mutant with greater positive surface charge and greater affinity for lipid membranes compared with MfVIA. This analogue had increased potency at NaV1.8 and was analgesic in the mouse formalin assay.
Collapse
Affiliation(s)
- Jennifer R Deuis
- From the Institute for Molecular Bioscience and School of Pharmacy, The University of Queensland, Woolloongabba, Queensland 4102, Australia, and
| | | | - Marco C Inserra
- From the Institute for Molecular Bioscience and School of Pharmacy, The University of Queensland, Woolloongabba, Queensland 4102, Australia, and
| | - Tzong-Hsien Lee
- Department of Biochemistry and Molecular Biology, Monash University, Wellington Road, Clayton, Victoria 3800, Australia
| | - Marie-Isabel Aguilar
- Department of Biochemistry and Molecular Biology, Monash University, Wellington Road, Clayton, Victoria 3800, Australia
| | | | | | | | - Mehdi Mobli
- Centre for Advanced Imaging, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | | | | | - Irina Vetter
- From the Institute for Molecular Bioscience and School of Pharmacy, The University of Queensland, Woolloongabba, Queensland 4102, Australia, and
| |
Collapse
|
47
|
Shah P, Hsiao FSH, Ho YH, Chen CS. The proteome targets of intracellular targeting antimicrobial peptides. Proteomics 2016; 16:1225-37. [PMID: 26648572 DOI: 10.1002/pmic.201500380] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 10/30/2015] [Accepted: 12/03/2015] [Indexed: 01/28/2023]
Abstract
Antimicrobial peptides have been considered well-deserving candidates to fight the battle against microorganisms due to their broad-spectrum antimicrobial activities. Several studies have suggested that membrane disruption is the basic mechanism of AMPs that leads to killing or inhibiting microorganisms. Also, AMPs have been reported to interact with macromolecules inside the microbial cells such as nucleic acids (DNA/RNA), protein synthesis, essential enzymes, membrane septum formation and cell wall synthesis. Proteins are associated with many intracellular mechanisms of cells, thus protein targets may be specifically involved in mechanisms of action of AMPs. AMPs like pyrrhocoricin, drosocin, apidecin and Bac 7 are documented to have protein targets, DnaK and GroEL. Moreover, the intracellular targeting AMPs are reported to influence more than one protein targets inside the cell, suggesting for the multiple modes of actions. This complex mechanism of intracellular targeting AMPs makes them more difficult for the development of resistance. Herein, we have summarized the current status of AMPs in terms of their mode of actions, entry to cytoplasm and inhibition of macromolecules. To reveal the mechanism of action, we have focused on AMPs with intracellular protein targets. We have also included the use of high-throughput proteome microarray to determine the unidentified AMP protein targets in this review.
Collapse
Affiliation(s)
- Pramod Shah
- Graduate Institute of Systems Biology and Bioinformatics, National Central University, Jhongli, Taiwan.,Department of Biomedical Science and Engineering, National Central University, Jhongli, Taiwan
| | - Felix Shih-Hsiang Hsiao
- Graduate Institute of Systems Biology and Bioinformatics, National Central University, Jhongli, Taiwan.,Department of Biomedical Science and Engineering, National Central University, Jhongli, Taiwan
| | - Yu-Hsuan Ho
- Graduate Institute of Systems Biology and Bioinformatics, National Central University, Jhongli, Taiwan.,Department of Biomedical Science and Engineering, National Central University, Jhongli, Taiwan
| | - Chien-Sheng Chen
- Graduate Institute of Systems Biology and Bioinformatics, National Central University, Jhongli, Taiwan.,Department of Biomedical Science and Engineering, National Central University, Jhongli, Taiwan
| |
Collapse
|
48
|
Brannan AM, Whelan WA, Cole E, Booth V. Differential scanning calorimetry of whole Escherichia coli treated with the antimicrobial peptide MSI-78 indicate a multi-hit mechanism with ribosomes as a novel target. PeerJ 2015; 3:e1516. [PMID: 26713257 PMCID: PMC4690349 DOI: 10.7717/peerj.1516] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 11/27/2015] [Indexed: 11/20/2022] Open
Abstract
Differential Scanning Calorimetry (DSC) of intact Escherichia coli (E. coli) was used to identify non-lipidic targets of the antimicrobial peptide (AMP) MSI-78. The DSC thermograms revealed that, in addition to its known lytic properties, MSI-78 also has a striking effect on ribosomes. MSI-78’s effect on DSC scans of bacteria was similar to that of kanamycin, an antibiotic drug known to target the 30S small ribosomal subunit. An in vitro transcription/translation assay helped confirm MSI-78’s targeting of ribosomes. The scrambled version of MSI-78 also affected the ribosome peak of the DSC scans, but required greater amounts of peptide to cause a similar effect to the unscrambled peptide. Furthermore, the effect of the scrambled peptide was not specific to the ribosomes; other regions of the DSC thermogram were also affected. These results suggest that MSI-78’s effects on E. coli are at least somewhat dependent on its particular structural features, rather than a sole function of its overall charge and hydrophobicity. When considered along with earlier work detailing MSI-78’s membrane lytic properties, it appears that MSI-78 operates via a multi-hit mechanism with multiple targets.
Collapse
Affiliation(s)
- Alexander M Brannan
- Department of Biochemistry, Memorial University of Newfoundland , St. John's Newfoundland and Labrador , Canada
| | - William A Whelan
- Department of Biochemistry, Memorial University of Newfoundland , St. John's Newfoundland and Labrador , Canada
| | - Emma Cole
- Department of Biochemistry, Memorial University of Newfoundland , St. John's Newfoundland and Labrador , Canada
| | - Valerie Booth
- Department of Biochemistry, Memorial University of Newfoundland , St. John's Newfoundland and Labrador , Canada ; Department of Physics and Physical Oceanography, Memorial University of Newfoundland , St. John's Newfoundland and Labrador , Canada
| |
Collapse
|
49
|
Shahmiri M, Enciso M, Mechler A. Controls and constrains of the membrane disrupting action of Aurein 1.2. Sci Rep 2015; 5:16378. [PMID: 26574052 PMCID: PMC4648102 DOI: 10.1038/srep16378] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 10/13/2015] [Indexed: 11/21/2022] Open
Abstract
Aurein 1.2 is a 13 residue antimicrobial peptide secreted by the Australian tree frog Litoria Aurea. It is a surface-acting membrane disrupting peptide that permeabilizes bacterial membranes via the carpet mechanism; the molecular details of this process are mostly unknown. Here the mechanism of action of Aurein 1.2 was investigated with an emphasis on the role of membrane charge and C-terminal amidation of the peptide. Using quartz crystal microbalance (QCM) fingerprinting it was found that the membrane charge correlates with membrane affinity of the peptide, however the binding and the membrane disrupting processes are not charge driven; increased membrane charge reduces the membrane disrupting activity. Coarse grain simulations revealed that phenylalanine residues act as membrane anchors. Accordingly Aurein 1.2 has the ability to bind to any membrane. Furthermore, bundling precludes membrane disruption in case of wild type peptides, while non C-terminal amidated peptides form random aggregates leading to detachment from the membrane. Hence C-terminal amidation is crucial for Aurein 1.2 action. Our results suggest that Aurein 1.2 acts via aggregation driven membrane penetration. The concomitant change in the tension of the outer leaflet imposes a spontaneous curvature on the membrane, leading to disintegration.
Collapse
Affiliation(s)
- Mahdi Shahmiri
- Department of Chemistry and Physics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora Vic 3086, Australia
| | - Marta Enciso
- Department of Chemistry and Physics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora Vic 3086, Australia
| | - Adam Mechler
- Department of Chemistry and Physics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora Vic 3086, Australia
| |
Collapse
|
50
|
Pissinis DE, Diaz C, Maza E, Bonini IC, Barrantes FJ, Salvarezza RC, Schilardi PL. Functional nicotinic acetylcholine receptor reconstitution in Au(111)-supported thiolipid monolayers. NANOSCALE 2015; 7:15789-15797. [PMID: 26355753 DOI: 10.1039/c5nr04109k] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
The insertion and function of the muscle-type nicotinic acetylcholine receptor (nAChR) in Au(111)-supported thiolipid self-assembled monolayers have been studied by atomic force microscopy (AFM), surface plasmon resonance (SPR), and electrochemical techniques. It was possible for the first time to resolve the supramolecular arrangement of the protein spontaneously inserted in a thiolipid monolayer in an aqueous solution. Geometric supramolecular arrays of nAChRs were observed, most commonly in a triangular form compatible with three nAChR dimers of ∼20 nm each. Addition of the full agonist carbamoylcholine activated and opened the nAChR ion channel, as revealed by the increase in capacitance relative to that of the nAChR-thiolipid system under basal conditions. Thus, the self-assembled system appears to be a viable biomimetic model to measure ionic conductance mediated by ion-gated ion channels under different experimental conditions, with potential applications in biotechnology and pharmacology.
Collapse
Affiliation(s)
- Diego E Pissinis
- Instituto de Investigaciones Fisicoquímicas Teóricas y Aplicadas (INIFTA), CONICET - Departamento de Química, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, CC16, Suc. 4, La Plata, Buenos Aires, Argentina.
| | | | | | | | | | | | | |
Collapse
|