1
|
Singh A, Wu M, Ye TT, Brown AC, Wittenberg NJ. Engineering Planar Gram-Negative Outer Membrane Mimics Using Bacterial Outer Membrane Vesicles. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:23289-23300. [PMID: 39453730 PMCID: PMC11542184 DOI: 10.1021/acs.langmuir.4c02632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 10/02/2024] [Accepted: 10/15/2024] [Indexed: 10/27/2024]
Abstract
Antibiotic resistance is a major challenge in modern medicine. The unique double membrane structure of Gram-negative bacteria limits the efficacy of many existing antibiotics and adds complexity to antibiotic development by limiting transport of antibiotics to the bacterial cytosol. New methods to mimic this barrier would enable high-throughput studies for antibiotic development. In this study, we introduce an innovative approach to modify outer membrane vesicles (OMVs) from Aggregatibacter actinomycetemcomitans, to generate planar supported lipid bilayer membranes. Our method first involves the incorporation of synthetic lipids into OMVs using a rapid freeze-thaw technique to form outer membrane hybrid vesicles (OM-Hybrids). Subsequently, these OM-Hybrids can spontaneously rupture when in contact with SiO2 surfaces to form a planar outer membrane supported bilayer (OM-SB). We assessed the formation of OM-Hybrids using dynamic light scattering and a fluorescence quenching assay. To analyze the formation of OM-SBs from OM-Hybrids we used quartz crystal microbalance with dissipation monitoring (QCM-D) and fluorescence recovery after photobleaching (FRAP). Additionally, we conducted assays to detect surface-associated DNA and proteins on OM-SBs. The interaction of an antimicrobial peptide, polymyxin B, with the OM-SBs was also assessed. These findings emphasize the capability of our platform to produce planar surfaces of bacterial outer membranes, which in turn, could function as a valuable tool for streamlining the development of antibiotics.
Collapse
Affiliation(s)
- Aarshi
N. Singh
- Department
of Chemistry, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Meishan Wu
- Department
of Chemical and Biomolecular Engineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Tiffany T. Ye
- Department
of Chemistry, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Angela C. Brown
- Department
of Chemical and Biomolecular Engineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Nathan J. Wittenberg
- Department
of Chemistry, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| |
Collapse
|
2
|
Singh AN, Wu M, Ye TT, Brown AC, Wittenberg NJ. Engineering Planar Gram-Negative Outer Membrane Mimics Using Bacterial Outer Membrane Vesicles. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.11.570829. [PMID: 39229024 PMCID: PMC11370475 DOI: 10.1101/2023.12.11.570829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Antibiotic resistance is a major challenge in modern medicine. The unique double membrane structure of gram-negative bacteria limits the efficacy of many existing antibiotics and adds complexity to antibiotic development by limiting transport of antibiotics to the bacterial cytosol. New methods to mimic this barrier would enable high-throughput studies for antibiotic development. In this study, we introduce an innovative approach to modify outer membrane vesicles (OMVs) from Aggregatibacter actinomycetemcomitans, to generate planar supported lipid bilayer membranes. Our method first involves the incorporation of synthetic lipids into OMVs using a rapid freeze-thaw technique to form outer membrane hybrid vesicles (OM-Hybrids). Subsequently, these OM-Hybrids can spontaneously rupture when in contact with SiO2 surfaces to form a planar outer membrane supported bilayer (OM-SB). We assessed the formation of OM-Hybrids using dynamic light scattering and a fluorescence quenching assay. To analyze the formation of OM-SBs from OM-Hybrids we used quartz crystal microbalance with dissipation monitoring (QCM-D) and fluorescence recovery after photobleaching (FRAP). Additionally, we conducted assays to detect surface-associated DNA and proteins on OM-SBs. The interaction of an antimicrobial peptide, polymyxin B, with the OM-SBs was also assessed. These findings emphasize the capability of our platform to produce planar surfaces of bacterial outer membranes, which in turn, could function as a valuable tool for streamlining the development of antibiotics.
Collapse
Affiliation(s)
- Aarshi N. Singh
- Department of Chemistry, Lehigh University, Bethlehem, PA, USA
| | - Meishan Wu
- Department of Chemical and Biomolecular Engineering, Lehigh University, Bethlehem, PA, USA
| | - Tiffany T. Ye
- Department of Chemistry, Lehigh University, Bethlehem, PA, USA
| | - Angela C. Brown
- Department of Chemical and Biomolecular Engineering, Lehigh University, Bethlehem, PA, USA
| | | |
Collapse
|
3
|
Buchholz KR, Reichelt M, Johnson MC, Robinson SJ, Smith PA, Rutherford ST, Quinn JG. Potent activity of polymyxin B is associated with long-lived super-stoichiometric accumulation mediated by weak-affinity binding to lipid A. Nat Commun 2024; 15:4733. [PMID: 38830951 PMCID: PMC11148078 DOI: 10.1038/s41467-024-49200-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 05/23/2024] [Indexed: 06/05/2024] Open
Abstract
Polymyxins are gram-negative antibiotics that target lipid A, the conserved membrane anchor of lipopolysaccharide in the outer membrane. Despite their clinical importance, the molecular mechanisms underpinning polymyxin activity remain unresolved. Here, we use surface plasmon resonance to kinetically interrogate interactions between polymyxins and lipid A and derive a phenomenological model. Our analyses suggest a lipid A-catalyzed, three-state mechanism for polymyxins: transient binding, membrane insertion, and super-stoichiometric cluster accumulation with a long residence time. Accumulation also occurs for brevicidine, another lipid A-targeting antibacterial molecule. Lipid A modifications that impart polymyxin resistance and a non-bactericidal polymyxin derivative exhibit binding that does not evolve into long-lived species. We propose that transient binding to lipid A permeabilizes the outer membrane and cluster accumulation enables the bactericidal activity of polymyxins. These findings could establish a blueprint for discovery of lipid A-targeting antibiotics and provide a generalizable approach to study interactions with the gram-negative outer membrane.
Collapse
Affiliation(s)
- Kerry R Buchholz
- Department of Infectious Diseases, Genentech, Inc., South San Francisco, CA, USA.
| | - Mike Reichelt
- Department of Pathology, Genentech, Inc., South San Francisco, CA, USA
| | - Matthew C Johnson
- Department of Structural Biology, Genentech, Inc., South San Francisco, CA, USA
| | - Sarah J Robinson
- Department of Discovery Chemistry, Genentech, Inc., South San Francisco, CA, USA
| | - Peter A Smith
- Department of Infectious Diseases, Genentech, Inc., South San Francisco, CA, USA
- Revagenix, Inc., San Mateo, CA, USA
| | - Steven T Rutherford
- Department of Infectious Diseases, Genentech, Inc., South San Francisco, CA, USA.
| | - John G Quinn
- Department of Biochemical and Cellular Pharmacology, Genentech, Inc., South San Francisco, CA, USA.
| |
Collapse
|
4
|
Bint-E-Naser SF, Mohamed ZJ, Chao Z, Bali K, Owens RM, Daniel S. Gram-Positive Bacterial Membrane-Based Biosensor for Multimodal Investigation of Membrane-Antibiotic Interactions. BIOSENSORS 2024; 14:45. [PMID: 38248423 PMCID: PMC10813107 DOI: 10.3390/bios14010045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/09/2024] [Accepted: 01/09/2024] [Indexed: 01/23/2024]
Abstract
As membrane-mediated antibiotic resistance continues to evolve in Gram-positive bacteria, the development of new approaches to elucidate the membrane properties involved in antibiotic resistance has become critical. Membrane vesicles (MVs) secreted by the cytoplasmic membrane of Gram-positive bacteria contain native components, preserving lipid and protein diversity, nucleic acids, and sometimes virulence factors. Thus, MV-derived membrane platforms present a great model for Gram-positive bacterial membranes. In this work, we report the development of a planar bacterial cytoplasmic membrane-based biosensor using MVs isolated from the Bacillus subtilis WT strain that can be coated on multiple surface types such as glass, quartz crystals, and polymeric electrodes, fostering the multimodal assessment of drug-membrane interactions. Retention of native membrane components such as lipoteichoic acids, lipids, and proteins is verified. This biosensor replicates known interaction patterns of the antimicrobial compound, daptomycin, with the Gram-positive bacterial membrane, establishing the applicability of this platform for carrying out biophysical characterization of the interactions of membrane-acting antibiotic compounds with the bacterial cytoplasmic membrane. We report changes in membrane viscoelasticity and permeability that correspond to partial membrane disruption when calcium ions are present with daptomycin but not when these ions are chelated. This biomembrane biosensing platform enables an assessment of membrane biophysical characteristics during exposure to antibiotic drug candidates to aid in identifying compounds that target membrane disruption as a mechanism of action.
Collapse
Affiliation(s)
- Samavi Farnush Bint-E-Naser
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA; (S.F.B.-E.-N.); (Z.C.)
| | | | - Zhongmou Chao
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA; (S.F.B.-E.-N.); (Z.C.)
| | - Karan Bali
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge CB3 0AS, UK; (K.B.); (R.M.O.)
| | - Róisín M. Owens
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge CB3 0AS, UK; (K.B.); (R.M.O.)
| | - Susan Daniel
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA; (S.F.B.-E.-N.); (Z.C.)
| |
Collapse
|
5
|
Bali K, McCoy R, Lu Z, Treiber J, Savva A, Kaminski CF, Salmond G, Salleo A, Mela I, Monson R, Owens RM. Multiparametric Sensing of Outer Membrane Vesicle-Derived Supported Lipid Bilayers Demonstrates the Specificity of Bacteriophage Interactions. ACS Biomater Sci Eng 2023. [PMID: 37137156 DOI: 10.1021/acsbiomaterials.3c00021] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
The use of bacteriophages, viruses that specifically infect bacteria, as antibiotics has become an area of great interest in recent years as the effectiveness of conventional antibiotics recedes. The detection of phage interactions with specific bacteria in a rapid and quantitative way is key for identifying phages of interest for novel antimicrobials. Outer membrane vesicles (OMVs) derived from Gram-negative bacteria can be used to make supported lipid bilayers (SLBs) and therefore in vitro membrane models that contain naturally occurring components of the bacterial outer membrane. In this study, we employed Escherichia coli OMV derived SLBs and use both fluorescent imaging and mechanical sensing techniques to show their interactions with T4 phage. We also integrate these bilayers with microelectrode arrays (MEAs) functionalized with the conducting polymer PEDOT:PSS and show that the pore forming interactions of the phages with the SLBs can be monitored using electrical impedance spectroscopy. To highlight our ability to detect specific phage interactions, we also generate SLBs using OMVs derived from Citrobacter rodentium, which is resistant to T4 phage infection, and identify their lack of interaction with the phage. The work presented here shows how interactions occurring between the phages and these complex SLB systems can be monitored using a range of experimental techniques. We believe this approach can be used to identify phages that work against bacterial strains of interest, as well as more generally to monitor any pore forming structure (such as defensins) interacting with bacterial outer membranes, and thus aid in the development of next generation antimicrobials.
Collapse
Affiliation(s)
- Karan Bali
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, United Kingdom
| | - Reece McCoy
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, United Kingdom
| | - Zixuan Lu
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, United Kingdom
| | - Jeremy Treiber
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, United States
| | - Achilleas Savva
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, United Kingdom
| | - Clemens F Kaminski
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, United Kingdom
| | - George Salmond
- Department of Biochemistry, University of Cambridge, Hopkins Building, Downing Site, Tennis Court Road, Cambridge CB2 1QW, United Kingdom
| | - Alberto Salleo
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, United States
| | - Ioanna Mela
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1PD, United Kingdom
| | - Rita Monson
- Department of Biochemistry, University of Cambridge, Hopkins Building, Downing Site, Tennis Court Road, Cambridge CB2 1QW, United Kingdom
| | - Róisín M Owens
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, United Kingdom
| |
Collapse
|
6
|
Arya SS, Morsy NK, Islayem DK, Alkhatib SA, Pitsalidis C, Pappa AM. Bacterial Membrane Mimetics: From Biosensing to Disease Prevention and Treatment. BIOSENSORS 2023; 13:bios13020189. [PMID: 36831955 PMCID: PMC9953710 DOI: 10.3390/bios13020189] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/19/2023] [Accepted: 01/24/2023] [Indexed: 05/31/2023]
Abstract
Plasma membrane mimetics can potentially play a vital role in drug discovery and immunotherapy owing to the versatility to assemble facilely cellular membranes on surfaces and/or nanoparticles, allowing for direct assessment of drug/membrane interactions. Recently, bacterial membranes (BMs) have found widespread applications in biomedical research as antibiotic resistance is on the rise, and bacteria-associated infections have become one of the major causes of death worldwide. Over the last decade, BM research has greatly benefited from parallel advancements in nanotechnology and bioelectronics, resulting in multifaceted systems for a variety of sensing and drug discovery applications. As such, BMs coated on electroactive surfaces are a particularly promising label-free platform to investigate interfacial phenomena, as well as interactions with drugs at the first point of contact: the bacterial membrane. Another common approach suggests the use of lipid-coated nanoparticles as a drug carrier system for therapies for infectious diseases and cancer. Herein, we discuss emerging platforms that make use of BMs for biosensing, bioimaging, drug delivery/discovery, and immunotherapy, focusing on bacterial infections and cancer. Further, we detail the synthesis and characteristics of BMs, followed by various models for utilizing them in biomedical applications. The key research areas required to augment the characteristics of bacterial membranes to facilitate wider applicability are also touched upon. Overall, this review provides an interdisciplinary approach to exploit the potential of BMs and current emerging technologies to generate novel solutions to unmet clinical needs.
Collapse
Affiliation(s)
- Sagar S. Arya
- Department of Biomedical Engineering, Khalifa University of Science and Technology, Abu Dhabi P.O. Box 127788, United Arab Emirates
| | - Nada K. Morsy
- Department of Biomedical Engineering, Khalifa University of Science and Technology, Abu Dhabi P.O. Box 127788, United Arab Emirates
| | - Deema K. Islayem
- Department of Biomedical Engineering, Khalifa University of Science and Technology, Abu Dhabi P.O. Box 127788, United Arab Emirates
| | - Sarah A. Alkhatib
- Department of Biomedical Engineering, Khalifa University of Science and Technology, Abu Dhabi P.O. Box 127788, United Arab Emirates
| | - Charalampos Pitsalidis
- Department of Physics Khalifa University of Science and Technology, Abu Dhabi P.O. Box 127788, United Arab Emirates
- Healthcare Engineering Innovation Center (HEIC), Khalifa University of Science and Technology, Abu Dhabi P.O. Box 127788, United Arab Emirates
- Department of Chemical Engineering and Biotechnology, Cambridge University, Philippa Fawcett Drive, Cambridge CB30AS, UK
| | - Anna-Maria Pappa
- Department of Biomedical Engineering, Khalifa University of Science and Technology, Abu Dhabi P.O. Box 127788, United Arab Emirates
- Healthcare Engineering Innovation Center (HEIC), Khalifa University of Science and Technology, Abu Dhabi P.O. Box 127788, United Arab Emirates
- Department of Chemical Engineering and Biotechnology, Cambridge University, Philippa Fawcett Drive, Cambridge CB30AS, UK
| |
Collapse
|
7
|
Li S, Ren R, Lyu L, Song J, Wang Y, Lin TW, Brun AL, Hsu HY, Shen HH. Solid and Liquid Surface-Supported Bacterial Membrane Mimetics as a Platform for the Functional and Structural Studies of Antimicrobials. MEMBRANES 2022; 12:membranes12100906. [PMID: 36295664 PMCID: PMC9609327 DOI: 10.3390/membranes12100906] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/05/2022] [Accepted: 09/13/2022] [Indexed: 06/02/2023]
Abstract
Increasing antibiotic resistance has provoked the urgent need to investigate the interactions of antimicrobials with bacterial membranes. The reasons for emerging antibiotic resistance and innovations in novel therapeutic approaches are highly relevant to the mechanistic interactions between antibiotics and membranes. Due to the dynamic nature, complex compositions, and small sizes of native bacterial membranes, bacterial membrane mimetics have been developed to allow for the in vitro examination of structures, properties, dynamics, and interactions. In this review, three types of model membranes are discussed: monolayers, supported lipid bilayers, and supported asymmetric bilayers; this review highlights their advantages and constraints. From monolayers to asymmetric bilayers, biomimetic bacterial membranes replicate various properties of real bacterial membranes. The typical synthetic methods for fabricating each model membrane are introduced. Depending on the properties of lipids and their biological relevance, various lipid compositions have been used to mimic bacterial membranes. For example, mixtures of phosphatidylethanolamines (PE), phosphatidylglycerols (PG), and cardiolipins (CL) at various molar ratios have been used, approaching actual lipid compositions of Gram-positive bacterial membranes and inner membranes of Gram-negative bacteria. Asymmetric lipid bilayers can be fabricated on solid supports to emulate Gram-negative bacterial outer membranes. To probe the properties of the model bacterial membranes and interactions with antimicrobials, three common characterization techniques, including quartz crystal microbalance with dissipation (QCM-D), surface plasmon resonance (SPR), and neutron reflectometry (NR) are detailed in this review article. Finally, we provide examples showing that the combination of bacterial membrane models and characterization techniques is capable of providing crucial information in the design of new antimicrobials that combat bacterial resistance.
Collapse
Affiliation(s)
- Shiqi Li
- Department of Materials Science and Engineering, Faculty of Engineering, Monash University, Clayton, VIC 3800, Australia
| | - Ruohua Ren
- Department of Materials Science and Engineering, Faculty of Engineering, Monash University, Clayton, VIC 3800, Australia
| | - Letian Lyu
- Department of Materials Science and Engineering, Faculty of Engineering, Monash University, Clayton, VIC 3800, Australia
| | - Jiangning Song
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Yajun Wang
- College of Chemistry & Materials Engineering, Wenzhou University, Wenzhou 325035, China
| | - Tsung-Wu Lin
- Department of Chemistry, Tunghai University, No. 1727, Sec. 4, Taiwan Boulevard, Xitun District, Taichung 40704, Taiwan
| | - Anton Le Brun
- Australian Centre for Neutron Scattering, Australian Nuclear Science and Technology Organisation, Locked Bag 2001, Kirrawee DC, NSW 2232, Australia
| | - Hsien-Yi Hsu
- Department of Materials Science and Engineering, School of Energy and Environment, City University of Hong Kong, Kowloon Tong, Hong Kong, China
| | - Hsin-Hui Shen
- Department of Materials Science and Engineering, Faculty of Engineering, Monash University, Clayton, VIC 3800, Australia
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| |
Collapse
|
8
|
Bali K, Mohamed Z, Scheeder A, Pappa AM, Daniel S, Kaminski CF, Owens RM, Mela I. Nanoscale Features of Tunable Bacterial Outer Membrane Models Revealed by Correlative Microscopy. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2022; 38:8773-8782. [PMID: 35748045 PMCID: PMC9330759 DOI: 10.1021/acs.langmuir.2c00628] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
The rise of antibiotic resistance is a growing worldwide human health issue, with major socioeconomic implications. An understanding of the interactions occurring at the bacterial membrane is crucial for the generation of new antibiotics. Supported lipid bilayers (SLBs) made from reconstituted lipid vesicles have been used to mimic these membranes, but their utility has been restricted by the simplistic nature of these systems. A breakthrough in the field has come with the use of outer membrane vesicles derived from Gram-negative bacteria to form SLBs, thus providing a more physiologically relevant system. These complex bilayer systems hold promise but have not yet been fully characterized in terms of their composition, ratio of natural to synthetic components, and membrane protein content. Here, we use correlative atomic force microscopy (AFM) with structured illumination microscopy (SIM) for the accurate mapping of complex lipid bilayers that consist of a synthetic fraction and a fraction of lipids derived from Escherichia coli outer membrane vesicles (OMVs). We exploit the high resolution and molecular specificity that SIM can offer to identify areas of interest in these bilayers and the enhanced resolution that AFM provides to create detailed topography maps of the bilayers. We are thus able to understand the way in which the two different lipid fractions (natural and synthetic) mix within the bilayers, and we can quantify the amount of bacterial membrane incorporated into the bilayer. We prove the system's tunability by generating bilayers made using OMVs engineered to contain a green fluorescent protein (GFP) binding nanobody fused with the porin OmpA. We are able to directly visualize protein-protein interactions between GFP and the nanobody complex. Our work sets the foundation for accurately understanding the composition and properties of OMV-derived SLBs to generate a high-resolution platform for investigating bacterial membrane interactions for the development of next-generation antibiotics.
Collapse
Affiliation(s)
- Karan Bali
- Department
of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge CB3 0AS, U.K.
| | - Zeinab Mohamed
- School
of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Anna Scheeder
- Department
of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge CB3 0AS, U.K.
| | - Anna-Maria Pappa
- Department
of Biomedical Engineering, Khalifa University
of Science and Technology, Abu
Dhabi 127788, United Arab
Emirates
| | - Susan Daniel
- School
of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States
- School
of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Clemens F. Kaminski
- Department
of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge CB3 0AS, U.K.
| | - Róisín M. Owens
- Department
of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge CB3 0AS, U.K.
| | - Ioanna Mela
- Department
of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge CB3 0AS, U.K.
| |
Collapse
|
9
|
Suri M, Mohamed Z, Bint E Naser SF, Mao X, Chen P, Daniel S, Hanrath T. Bioelectronic Platform to Investigate Charge Transfer between Photoexcited Quantum Dots and Microbial Outer Membranes. ACS APPLIED MATERIALS & INTERFACES 2022; 14:15799-15810. [PMID: 35344337 DOI: 10.1021/acsami.1c25032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Photosynthetic semiconductor biohybrids (PSBs) convert light energy to chemical energy through photo-driven charge transfer from nanocrystals to microorganisms that perform bioreactions of interest. Initial proof-of-concept PSB studies with an emphasis on enhanced CO2 conversion have been encouraging; however, bringing the broad prospects of PSBs to fruition is contingent on establishing a firm fundamental understanding of underlying interfacial charge transfer processes. We introduce a bioelectronic platform that reduces the complexity of PSBs by focusing explicitly on interactions between colloidal quantum dots (QDs), microbial outer membranes, and native, small-molecule redox mediators. Our model platform employs a standard three-electrode electrochemical cell with supported outer membranes of Pseudomonas aeruginosa, pyocyanin redox mediators, and semiconducting CdSe QDs dispersed in an aqueous electrolyte. We present a comprehensive electrochemical analysis of this platform via electrochemical impedance spectroscopy (EIS), cyclic voltammetry (CV), and chronoamperometry (CA). EIS reveals the formation and electronic properties of supported outer membrane films. CV reveals the electrochemically active surface area of P. aeruginosa outer membranes and that pyocyanin is the sole species that performs redox with these outer membranes under sweeping applied potential. CA demonstrates that photoexcited charge transfer in this system is driven by the reduction of pyocyanin at the QD surface followed by diffusion of reduced pyocyanin through the outer membrane. The broad applicability of this platform across many bacterial species, QD architectures, and controlled environmental conditions affords the possibility to define design principles for future PSB systems to synergistically integrate concurrent advances in genetically engineered organisms and inorganic nanomaterials.
Collapse
Affiliation(s)
- Mokshin Suri
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14853, United States
- Department of Materials Science and Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Zeinab Mohamed
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Samavi Farnush Bint E Naser
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Xianwen Mao
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
| | - Peng Chen
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
| | - Susan Daniel
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14853, United States
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Tobias Hanrath
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14853, United States
| |
Collapse
|
10
|
Lee S, Bayley H. Reconstruction of the Gram-Negative Bacterial Outer-Membrane Bilayer. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2200007. [PMID: 35289495 DOI: 10.1002/smll.202200007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 02/27/2022] [Indexed: 06/14/2023]
Abstract
The outer membrane (OM) of gram-negative bacteria is highly asymmetric. The outer leaflet comprises lipopolysaccharides (LPS) and the inner leaflet phospholipids. Here, it is shown that the outer membrane lipid bilayer (OMLB) of Escherichia coli can be reconstructed as a droplet interface bilayer (DIB), which separates two aqueous droplets in oil. The trimeric porin OmpF is inserted into the model OMLB and the translocation of the bacteriocin colicin E9 (colE9) through it is monitored. By contrast with LPS-free bilayers, it is found that colE9 made multiple failed attempts to engage with OmpF in an OMLB before successful translocation occurred. In addition, the observed rate for the second step of colE9 translocation is 3-times smaller than that in LPS-free bilayers, and further, the colE9 dissociates when the membrane potential is reversed. The findings demonstrate the utility of the DIB approach for constructing model OMLBs from physiologically realistic lipids and that the properties of the model OMLBs differ from those of a simple lipid bilayer. The model OMLB offers a credible platform for screening the properties of antibiotics.
Collapse
Affiliation(s)
- Sejeong Lee
- Chemistry Research Laboratory, University of Oxford, 12 Mansfield Road, Oxford, OX1 3TA, UK
| | - Hagan Bayley
- Chemistry Research Laboratory, University of Oxford, 12 Mansfield Road, Oxford, OX1 3TA, UK
| |
Collapse
|
11
|
Richter R, Kamal MAM, Koch M, Niebuur B, Huber A, Goes A, Volz C, Vergalli J, Kraus T, Müller R, Schneider‐Daum N, Fuhrmann G, Pagès J, Lehr C. An Outer Membrane Vesicle-Based Permeation Assay (OMPA) for Assessing Bacterial Bioavailability. Adv Healthc Mater 2022; 11:e2101180. [PMID: 34614289 PMCID: PMC11468809 DOI: 10.1002/adhm.202101180] [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: 06/15/2021] [Revised: 09/14/2021] [Indexed: 11/11/2022]
Abstract
When searching for new antibiotics against Gram-negative bacterial infections, a better understanding of the permeability across the cell envelope and tools to discriminate high from low bacterial bioavailability compounds are urgently needed. Inspired by the phospholipid vesicle-based permeation assay (PVPA), which is designed to predict non-facilitated permeation across phospholipid membranes, outer membrane vesicles (OMVs) of Escherichia coli either enriched or deficient of porins are employed to coat filter supports for predicting drug uptake across the complex cell envelope. OMVs and the obtained in vitro model are structurally and functionally characterized using cryo-TEM, SEM, CLSM, SAXS, and light scattering techniques. In vitro permeability, obtained from the membrane model for a set of nine antibiotics, correlates with reported in bacterio accumulation data and allows to discriminate high from low accumulating antibiotics. In contrast, the correlation of the same data set generated by liposome-based comparator membranes is poor. This better correlation of the OMV-derived membranes points to the importance of hydrophilic membrane components, such as lipopolysaccharides and porins, since those features are lacking in liposomal comparator membranes. This approach can offer in the future a high throughput screening tool with high predictive capacity or can help to identify compound- and bacteria-specific passive uptake pathways.
Collapse
Affiliation(s)
- Robert Richter
- Helmholtz Centre for Infection ResearchHelmholtz Institute for Pharmaceutical Research SaarlandCampus E8.1Saarbrücken66123Germany
| | - Mohamed A. M. Kamal
- Helmholtz Centre for Infection ResearchHelmholtz Institute for Pharmaceutical Research SaarlandCampus E8.1Saarbrücken66123Germany
- Saarland UniversityDepartment of PharmacyCampus E8.1Saarbrücken66123Germany
| | - Marcus Koch
- INM – Leibniz Institute for New MaterialsCampus D2.2Saarbrücken66123Germany
| | - Bart‐Jan Niebuur
- INM – Leibniz Institute for New MaterialsCampus D2.2Saarbrücken66123Germany
| | - Anna‐Lena Huber
- Helmholtz Centre for Infection ResearchHelmholtz Institute for Pharmaceutical Research SaarlandCampus E8.1Saarbrücken66123Germany
- Saarland UniversityDepartment of PharmacyCampus E8.1Saarbrücken66123Germany
| | - Adriely Goes
- Helmholtz Centre for Infection ResearchHelmholtz Institute for Pharmaceutical Research SaarlandCampus E8.1Saarbrücken66123Germany
- Saarland UniversityDepartment of PharmacyCampus E8.1Saarbrücken66123Germany
| | - Carsten Volz
- Helmholtz Centre for Infection ResearchHelmholtz Institute for Pharmaceutical Research SaarlandCampus E8.1Saarbrücken66123Germany
| | - Julia Vergalli
- UMR_MD1U‐1261Aix‐Marseille UniversitéINSERMIRBAMCTFaculté de Pharmacie27 Boulevard Jean MoulinMarseille13005France
| | - Tobias Kraus
- INM – Leibniz Institute for New MaterialsCampus D2.2Saarbrücken66123Germany
- Colloid and Interface ChemistrySaarland UniversityCampus D2.2Saarbrücken66123Germany
| | - Rolf Müller
- Helmholtz Centre for Infection ResearchHelmholtz Institute for Pharmaceutical Research SaarlandCampus E8.1Saarbrücken66123Germany
- Saarland UniversityDepartment of PharmacyCampus E8.1Saarbrücken66123Germany
| | - Nicole Schneider‐Daum
- Helmholtz Centre for Infection ResearchHelmholtz Institute for Pharmaceutical Research SaarlandCampus E8.1Saarbrücken66123Germany
| | - Gregor Fuhrmann
- Helmholtz Centre for Infection ResearchHelmholtz Institute for Pharmaceutical Research SaarlandCampus E8.1Saarbrücken66123Germany
- Saarland UniversityDepartment of PharmacyCampus E8.1Saarbrücken66123Germany
| | - Jean‐Marie Pagès
- UMR_MD1U‐1261Aix‐Marseille UniversitéINSERMIRBAMCTFaculté de Pharmacie27 Boulevard Jean MoulinMarseille13005France
| | - Claus‐Michael Lehr
- Helmholtz Centre for Infection ResearchHelmholtz Institute for Pharmaceutical Research SaarlandCampus E8.1Saarbrücken66123Germany
- Saarland UniversityDepartment of PharmacyCampus E8.1Saarbrücken66123Germany
| |
Collapse
|
12
|
Paracini N, Schneck E, Imberty A, Micciulla S. Lipopolysaccharides at Solid and Liquid Interfaces: Models for Biophysical Studies of the Gram-negative Bacterial Outer Membrane. Adv Colloid Interface Sci 2022; 301:102603. [PMID: 35093846 DOI: 10.1016/j.cis.2022.102603] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 01/14/2022] [Accepted: 01/15/2022] [Indexed: 11/18/2022]
Abstract
Lipopolysaccharides (LPSs) are a constitutive element of the cell envelope of Gram-negative bacteria, representing the main lipid in the external leaflet of their outer membrane (OM) lipid bilayer. These unique surface-exposed glycolipids play a central role in the interactions of Gram-negative organisms with their surrounding environment and represent a key element for protection against antimicrobials and the development of antibiotic resistance. The biophysical investigation of a wide range of different types of in vitro model membranes containing reconstituted LPS has revealed functional and structural properties of these peculiar membrane lipids, providing molecular-level details of their interaction with antimicrobial compounds. LPS assemblies reconstituted at interfaces represent a versatile tool to study the properties of the Gram-negative OM by exploiting several surface-sensitive techniques, in particular X-ray and neutron scattering, which can probe the structure of thin films with sub-nanometer resolution. This review provides an overview of different approaches employed to investigate structural and biophysical properties of LPS, focusing on studies on Langmuir monolayers of LPS at the air/liquid interface and a range of supported LPS-containing model membranes reconstituted at solid/liquid interfaces.
Collapse
Affiliation(s)
| | - Emanuel Schneck
- Physics Departent, Technische Universität Darmstadt, Darmstadt, Germany
| | - Anne Imberty
- Université Grenoble Alpes, CNRS, CERMAV, Grenoble, France
| | | |
Collapse
|
13
|
Carey AB, Ashenden A, Köper I. Model architectures for bacterial membranes. Biophys Rev 2022; 14:111-143. [PMID: 35340604 PMCID: PMC8921416 DOI: 10.1007/s12551-021-00913-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 11/14/2021] [Indexed: 02/06/2023] Open
Abstract
The complex composition of bacterial membranes has a significant impact on the understanding of pathogen function and their development towards antibiotic resistance. In addition to the inherent complexity and biosafety risks of studying biological pathogen membranes, the continual rise of antibiotic resistance and its significant economical and clinical consequences has motivated the development of numerous in vitro model membrane systems with tuneable compositions, geometries, and sizes. Approaches discussed in this review include liposomes, solid-supported bilayers, and computational simulations which have been used to explore various processes including drug-membrane interactions, lipid-protein interactions, host-pathogen interactions, and structure-induced bacterial pathogenesis. The advantages, limitations, and applicable analytical tools of all architectures are summarised with a perspective for future research efforts in architectural improvement and elucidation of resistance development strategies and membrane-targeting antibiotic mechanisms. Supplementary Information The online version contains supplementary material available at 10.1007/s12551-021-00913-7.
Collapse
Affiliation(s)
- Ashley B. Carey
- Institute for Nanoscale Science and Technology, College for Science and Engineering, Flinders University, Adelaide, SA 5042 Australia
| | - Alex Ashenden
- Institute for Nanoscale Science and Technology, College for Science and Engineering, Flinders University, Adelaide, SA 5042 Australia
| | - Ingo Köper
- Institute for Nanoscale Science and Technology, College for Science and Engineering, Flinders University, Adelaide, SA 5042 Australia
| |
Collapse
|
14
|
Tang T, Savva A, Traberg WC, Xu C, Thiburce Q, Liu HY, Pappa AM, Martinelli E, Withers A, Cornelius M, Salleo A, Owens RM, Daniel S. Functional Infectious Nanoparticle Detector: Finding Viruses by Detecting Their Host Entry Functions Using Organic Bioelectronic Devices. ACS NANO 2021; 15:18142-18152. [PMID: 34694775 DOI: 10.1021/acsnano.1c06813] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Emerging viruses will continue to be a threat to human health and wellbeing into the foreseeable future. The COVID-19 pandemic revealed the necessity for rapid viral sensing and inhibitor screening in mitigating viral spread and impact. Here, we present a platform that uses a label-free electronic readout as well as a dual capability of optical (fluorescence) readout to sense the ability of a virus to bind and fuse with a host cell membrane, thereby sensing viral entry. This approach introduces a hitherto unseen level of specificity by distinguishing fusion-competent viruses from fusion-incompetent viruses. The ability to discern between competent and incompetent viruses means that this device could also be used for applications beyond detection, such as screening antiviral compounds for their ability to block virus entry mechanisms. Using optical means, we first demonstrate the ability to recapitulate the entry processes of influenza virus using a biomembrane containing the viral receptor that has been functionalized on a transparent organic bioelectronic device. Next, we detect virus membrane fusion, using the same, label-free devices. Using both reconstituted and native cell membranes as materials to functionalize organic bioelectronic devices, configured as electrodes and transistors, we measure changes in membrane properties when virus fusion is triggered by a pH drop, inducing hemagglutinin to undergo a conformational change that leads to membrane fusion.
Collapse
Affiliation(s)
- Tiffany Tang
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Olin Hall, Ithaca, New York 14853, United States
| | - Achilleas Savva
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, CB30AS Cambridge, United Kingdom
| | - Walther C Traberg
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, CB30AS Cambridge, United Kingdom
| | - Cheyan Xu
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Olin Hall, Ithaca, New York 14853, United States
| | - Quentin Thiburce
- Department of Materials Science and Engineering, Stanford University, 496 Lomita Mall, Stanford, California 94305, United States
| | - Han-Yuan Liu
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Olin Hall, Ithaca, New York 14853, United States
| | - Anna-Maria Pappa
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, CB30AS Cambridge, United Kingdom
| | - Eleonora Martinelli
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, CB30AS Cambridge, United Kingdom
| | - Aimee Withers
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, CB30AS Cambridge, United Kingdom
| | - Mercedes Cornelius
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, CB30AS Cambridge, United Kingdom
| | - Alberto Salleo
- Department of Materials Science and Engineering, Stanford University, 496 Lomita Mall, Stanford, California 94305, United States
| | - Róisín M Owens
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, CB30AS Cambridge, United Kingdom
| | - Susan Daniel
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Olin Hall, Ithaca, New York 14853, United States
| |
Collapse
|
15
|
Mohamed Z, Shin JH, Ghosh S, Sharma AK, Pinnock F, Bint E Naser Farnush S, Dörr T, Daniel S. Clinically Relevant Bacterial Outer Membrane Models for Antibiotic Screening Applications. ACS Infect Dis 2021; 7:2707-2722. [PMID: 34227387 DOI: 10.1021/acsinfecdis.1c00217] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Antibiotic resistance is a growing global health concern that has been increasing in prevalence over the past few decades. In Gram-negative bacteria, the outer membrane is an additional barrier through which antibiotics must traverse to kill the bacterium. In addition, outer membrane features and properties, like membrane surface charge, lipopolysaccharide (LPS) length, and membrane porins, can be altered in response to antibiotics and therefore, further mediate resistance. Model membranes have been used to mimic bacterial membranes to study antibiotic-induced membrane changes but often lack the compositional complexity of the actual outer membrane. Here, we developed a surface-supported membrane platform using outer membrane vesicles (OMVs) from clinically relevant Gram-negative bacteria and use it to characterize membrane biophysical properties and investigate its interaction with antibacterial compounds. We demonstrate that this platform maintains critical features of outer membranes, like fluidity, while retaining complex membrane components, like OMPs and LPS, which are central to membrane-mediated antibiotic resistance. This platform offers a non-pathogenic, cell-free surface to study such phenomena that is compatible with advanced microscopy and surface characterization tools like quartz crystal microbalance. We confirm these OMV bilayers recapitulate membrane interactions (or lack thereof) with the antibiotic compounds polymyxin B, bacitracin, and vancomycin, validating their use as representative models for the bacterial surface. By forming OMV bilayers from different strains, we envision that this platform could be used to investigate underlying biophysical differences in outer membranes leading to resistance, to screen and identify membrane-active antibiotics, or for the development of phage technologies targeting a particular membrane surface component.
Collapse
Affiliation(s)
- Zeinab Mohamed
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York United States
| | - Jung-Ho Shin
- Weill Institute for Cell and Molecular Biology and Department of Microbiology, Cornell University, Ithaca, New York United States
| | - Surajit Ghosh
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York United States
| | - Abhishek K. Sharma
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York United States
| | - Ferra Pinnock
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York United States
| | - Samavi Bint E Naser Farnush
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York United States
| | - Tobias Dörr
- Weill Institute for Cell and Molecular Biology and Department of Microbiology, Cornell University, Ithaca, New York United States
| | - Susan Daniel
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York United States
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York United States
| |
Collapse
|
16
|
Tan JYB, Yoon BK, Cho NJ, Lovrić J, Jug M, Jackman JA. Lipid Nanoparticle Technology for Delivering Biologically Active Fatty Acids and Monoglycerides. Int J Mol Sci 2021; 22:9664. [PMID: 34575831 PMCID: PMC8465605 DOI: 10.3390/ijms22189664] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 09/03/2021] [Accepted: 09/05/2021] [Indexed: 12/12/2022] Open
Abstract
There is enormous interest in utilizing biologically active fatty acids and monoglycerides to treat phospholipid membrane-related medical diseases, especially with the global health importance of membrane-enveloped viruses and bacteria. However, it is difficult to practically deliver lipophilic fatty acids and monoglycerides for therapeutic applications, which has led to the emergence of lipid nanoparticle platforms that support molecular encapsulation and functional presentation. Herein, we introduce various classes of lipid nanoparticle technology and critically examine the latest progress in utilizing lipid nanoparticles to deliver fatty acids and monoglycerides in order to treat medical diseases related to infectious pathogens, cancer, and inflammation. Particular emphasis is placed on understanding how nanoparticle structure is related to biological function in terms of mechanism, potency, selectivity, and targeting. We also discuss translational opportunities and regulatory needs for utilizing lipid nanoparticles to deliver fatty acids and monoglycerides, including unmet clinical opportunities.
Collapse
Affiliation(s)
- Jia Ying Brenda Tan
- School of Chemical Engineering and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea; (J.Y.B.T.); (B.K.Y.)
- School of Materials Science and Engineering, Nanyang Technological University, Singapore 637553, Singapore;
| | - Bo Kyeong Yoon
- School of Chemical Engineering and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea; (J.Y.B.T.); (B.K.Y.)
- School of Healthcare and Biomedical Engineering, Chonnam National University, Yeosu 59626, Korea
| | - Nam-Joon Cho
- School of Materials Science and Engineering, Nanyang Technological University, Singapore 637553, Singapore;
| | - Jasmina Lovrić
- Department of Pharmaceutical Technology, Faculty of Pharmacy and Biochemistry, University of Zagreb, 10000 Zagreb, Croatia; (J.L.); (M.J.)
| | - Mario Jug
- Department of Pharmaceutical Technology, Faculty of Pharmacy and Biochemistry, University of Zagreb, 10000 Zagreb, Croatia; (J.L.); (M.J.)
| | - Joshua A. Jackman
- School of Chemical Engineering and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea; (J.Y.B.T.); (B.K.Y.)
| |
Collapse
|
17
|
Calvaresi V, Redsted A, Norais N, Rand KD. Hydrogen-Deuterium Exchange Mass Spectrometry with Integrated Size-Exclusion Chromatography for Analysis of Complex Protein Samples. Anal Chem 2021; 93:11406-11414. [PMID: 34387074 DOI: 10.1021/acs.analchem.1c01171] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The growing use of hydrogen-deuterium exchange mass spectrometry (HDX-MS) for studying membrane proteins, large protein assemblies, and highly disulfide-bonded species is often challenged by the presence in the sample of large amounts of lipids, protein ligands, and/or highly ionizable reducing agents. Here, we describe how a short size-exclusion chromatography (SEC) column can be integrated with a conventional temperature-controlled HDX-MS setup to achieve fast and online removal of unwanted species from the HDX sample prior to chromatographic separation and MS analysis. Dual-mode valves permit labeled proteins eluting after SEC to be directed to the proteolytic and chromatographic columns, while unwanted sample components are led to waste. The SEC-coupled HDX-MS method allows analyses to be completed with lower or similar back-exchange compared to conventional experiments. We demonstrate the suitability of the method for the analysis of challenging protein samples, achieving efficient online removal of lipid components from protein-lipid systems, depletion of an antibody from an antigen during epitope mapping, and elimination of MS interfering compounds such as tris(2-carboxyethyl)phosphine (TCEP) during HDX-MS analysis of a disulfide-bonded protein. The implementation of the short SEC column to the conventional HDX-MS setup is straightforward and could be of significant general utility during the HDX-MS analysis of complex protein states.
Collapse
Affiliation(s)
- Valeria Calvaresi
- Protein Analysis Group, Department of Pharmacy, University of Copenhagen, Copenhagen 2100, Denmark.,GSK, Siena 53100, Italy
| | - Andreas Redsted
- Protein Analysis Group, Department of Pharmacy, University of Copenhagen, Copenhagen 2100, Denmark.,GSK, Siena 53100, Italy
| | | | - Kasper D Rand
- Protein Analysis Group, Department of Pharmacy, University of Copenhagen, Copenhagen 2100, Denmark
| |
Collapse
|
18
|
Studying the surfaces of bacteria using neutron scattering: finding new openings for antibiotics. Biochem Soc Trans 2021; 48:2139-2149. [PMID: 33005925 PMCID: PMC7609035 DOI: 10.1042/bst20200320] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/26/2020] [Accepted: 09/01/2020] [Indexed: 12/29/2022]
Abstract
The use of neutrons as a scattering probe to investigate biological membranes has steadily grown in the past three decades, shedding light on the structure and behaviour of this ubiquitous and fundamental biological barrier. Meanwhile, the rise of antibiotic resistance has catalysed a renewed interest in understanding the mechanisms underlying the dynamics of antibiotics interaction with the bacterial cell envelope. It is widely recognised that the key reason behind the remarkable success of Gram-negative pathogens in developing antibiotic resistance lies in the effectiveness of their outer membrane (OM) in defending the cell from antibacterial compounds. Critical to its function, the highly asymmetric lipid distribution between the inner and outer bilayer leaflets of the OM, adds an extra level of complexity to the study of this crucial defence barrier. Here we review the opportunities offered by neutron scattering techniques, in particular reflectometry, to provide structural information on the interactions of antimicrobials with in vitro models of the OM. The differential sensitivity of neutrons towards hydrogen and deuterium makes them a unique probe to study the structure and behaviour of asymmetric membranes. Molecular-level understanding of the interactions between antimicrobials and the Gram-negative OM provides valuable insights that can aid drug development and broaden our knowledge of this critically important biological barrier.
Collapse
|
19
|
Richter R, Lehr CM. Extracellular vesicles as novel assay tools to study cellular interactions of anti-infective compounds - A perspective. Adv Drug Deliv Rev 2021; 173:492-503. [PMID: 33857554 DOI: 10.1016/j.addr.2021.04.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/21/2021] [Accepted: 04/08/2021] [Indexed: 12/13/2022]
Abstract
Sudden outbreaks of novel infectious diseases and the persistent evolution of antimicrobial resistant pathogens make it necessary to develop specific tools to quickly understand pathogen-cell interactions and to study appropriate drug delivery strategies. Extracellular vesicles (EVs) are cell-specific biogenic transport systems, which are gaining more and more popularity as either diagnostic markers or drug delivery systems. Apart from that, there are emerging possibilities for EVs as tools to study drug penetration, drug-membrane interactions as well as pathogen-membrane interactions. However, it appears that the potential of EVs for such applications has not been fully exploited yet. Considering the vast variety of cells that can be involved in an infection, vesicle-based analytical methods are just emerging and the number of reported applications is still relatively small. Aim of this review is to discuss the current state of the art of EV-based assays, especially in the context of antimicrobial research and therapy, and to present some new perspectives for a more exhaustive and creative exploration in the future.
Collapse
Affiliation(s)
- Robert Richter
- Department of Drug Delivery Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS) Helmholtz-Centre for Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany
| | - Claus-Michael Lehr
- Department of Drug Delivery Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS) Helmholtz-Centre for Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany; Department of Pharmacy, Saarland University, Campus E8.1, 66123 Saarbrücken, Germany.
| |
Collapse
|
20
|
Guntnur RT, Muzzio N, Morales M, Romero G. Phase transition characterization of poly(oligo(ethylene glycol)methyl ether methacrylate) brushes using the quartz crystal microbalance with dissipation. SOFT MATTER 2021; 17:2530-2538. [PMID: 33508060 PMCID: PMC7954969 DOI: 10.1039/d0sm02169e] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Heterogeneous non-linear poly(ethylene glycol) analogs, like poly(oligo(ethylene glycol)methyl ether methacrylate) (POEGMA), are of particular interest in the fabrication of smart biocompatible coatings as they undergo a reversible macromolecular rearrangement in response to external heat stimuli. The phase transition dynamics of POEGMA coatings in response to external temperature stimuli have been poorly investigated. The quartz crystal microbalance with dissipation (QCM-D) can be used to investigate the phase transition of these functional coatings as polymer brushes in a dynamic and noninvasive in situ measurement. POEGMA brushes with different thickness are synthesized from the surface of a QCM-D sensor following a living radical polymerization technique by varying the monomer molecular weight. Investigations on the thermoresponsive collapse and swelling of POEGMA brushes grafted from the surface of a QCM-D sensor reveal the reversible phase transition nature of these coatings. Furthermore, the potential of these smart coatings in the field of biotechnology was explored by investigating the absorption and desorption of a model drug. A pulsatile drug release profile triggered by an increase in temperature is observed from POEGMA brushes. POEGMA brushes have the potential to be utilized as polymer coatings for controlled and programable drug release.
Collapse
Affiliation(s)
- Rohini Thevi Guntnur
- Department of Biomedical Engineering and Chemical Engineering, University of Texas at San Antonio One UTSA Circle, San Antonio, Texas 78249, USA.
| | - Nicolas Muzzio
- Department of Biomedical Engineering and Chemical Engineering, University of Texas at San Antonio One UTSA Circle, San Antonio, Texas 78249, USA.
| | - Madison Morales
- Department of Biomedical Engineering and Chemical Engineering, University of Texas at San Antonio One UTSA Circle, San Antonio, Texas 78249, USA.
| | - Gabriela Romero
- Department of Biomedical Engineering and Chemical Engineering, University of Texas at San Antonio One UTSA Circle, San Antonio, Texas 78249, USA.
| |
Collapse
|
21
|
Cawley JL, Jordan LR, Wittenberg NJ. Detection and Characterization of Vesicular Gangliosides Binding to Myelin-Associated Glycoprotein on Supported Lipid Bilayers. Anal Chem 2021; 93:1185-1192. [PMID: 33296186 DOI: 10.1021/acs.analchem.0c04412] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In the nervous system, a myelin sheath that originates from oligodendrocytes or Schwann cells wraps around axons to facilitate electrical signal transduction. The interface between an axon and myelin is maintained by a number of biomolecular interactions. Among the interactions are those between GD1a and GT1b gangliosides on the axon and myelin-associated glycoprotein (MAG) on myelin. Interestingly, these interactions can also inhibit neuronal outgrowth. Ganglioside-MAG interactions are often studied in cellular or animal models where their relative concentrations are not easily controlled or in assays where the gangliosides and MAG are not presented as part of fluid lipid bilayers. Here, we present an approach to characterize MAG-ganglioside interactions in real time, where MAG, GD1a, and GT1b contents are controlled and they are in their in vivo orientation within fluid lipid bilayers. Using a quartz crystal microbalance with dissipation monitoring (QCM-D) biosensor functionalized with a supported lipid bilayer (SLB) and MAG, we detect vesicular GD1a and GT1b binding and determine the interaction kinetics as a function of vesicular ganglioside content. MAG-bound vesicles are deformed similarly, regardless of the ganglioside or its mole fraction. We further demonstrate how MAG-ganglioside interactions can be disrupted by antiganglioside antibodies that override MAG-based neuron growth inhibition.
Collapse
Affiliation(s)
- Jennie L Cawley
- Department of Chemistry, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Luke R Jordan
- Department of Chemistry, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Nathan J Wittenberg
- Department of Chemistry, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| |
Collapse
|
22
|
Pappa AM, Liu HY, Traberg-Christensen W, Thiburce Q, Savva A, Pavia A, Salleo A, Daniel S, Owens RM. Optical and Electronic Ion Channel Monitoring from Native Human Membranes. ACS NANO 2020; 14:12538-12545. [PMID: 32469490 DOI: 10.1021/acsnano.0c01330] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Transmembrane proteins represent a major target for modulating cell activity, both in terms of therapeutics drugs and for pathogen interactions. Work on screening such therapeutics or identifying toxins has been severely limited by the lack of available methods that would give high content information on functionality (ideally multimodal) and that are suitable for high-throughput. Here, we have demonstrated a platform that is capable of multimodal (optical and electronic) screening of ligand gated ion-channel activity in human-derived membranes. The TREK-1 ion-channel was expressed within supported lipid bilayers, formed via vesicle fusion of blebs obtained from the HEK cell line overexpressing TREK-1. The resulting reconstituted native membranes were confirmed via fluorescence recovery after photobleaching to form mobile bilayers on top of films of the polymeric electroactive transducer poly(3,4-ethylenedioxythiophene) polystyrenesulfonate (PEDOT:PSS). PEDOT:PSS electrodes were then used for quantitative electrochemical impedance spectroscopy measurements of ligand-mediated TREK-1 interactions with two compounds, spadin and arachidonic acid, known to suppress and activate TREK-1 channels, respectively. PEDOT:PSS-based organic electrochemical transistors were then used for combined optical and electronic measurements of TREK-1 functionality. The technology demonstrated here is highly promising for future high-throughput screening of transmembrane protein modulators owing to the robust nature of the membrane integrated device and the highly quantitative electrical signals obtained. This is in contrast with live-cell-based electrophysiology assays (e.g., patch clamp) which compare poorly in terms of cost, usability, and compatibility with optical transduction.
Collapse
Affiliation(s)
- Anna-Maria Pappa
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, CB30AS Cambridge, United Kingdom
| | - Han-Yuan Liu
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Olin Hall, Ithaca, New York 14853, United States
| | - Walther Traberg-Christensen
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, CB30AS Cambridge, United Kingdom
| | - Quentin Thiburce
- Department of Materials Science and Engineering, Stanford University, 496 Lomita Mall, Stanford, California 94305, United States
| | - Achilleas Savva
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, CB30AS Cambridge, United Kingdom
| | - Aimie Pavia
- Department of Flexible Electronics, Ecole Nationale Supérieure des Mines, CMP-EMSE, 13541 Gardanne, France
- Panaxium SAS, 13100 Aix-en-Provence, France
| | - Alberto Salleo
- Department of Materials Science and Engineering, Stanford University, 496 Lomita Mall, Stanford, California 94305, United States
| | - Susan Daniel
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Olin Hall, Ithaca, New York 14853, United States
| | - Róisín M Owens
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, CB30AS Cambridge, United Kingdom
| |
Collapse
|
23
|
Stephan MS, Broeker NK, Saragliadis A, Roos N, Linke D, Barbirz S. In vitro Analysis of O-Antigen-Specific Bacteriophage P22 Inactivation by Salmonella Outer Membrane Vesicles. Front Microbiol 2020; 11:510638. [PMID: 33072001 PMCID: PMC7541932 DOI: 10.3389/fmicb.2020.510638] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 08/26/2020] [Indexed: 11/23/2022] Open
Abstract
Bacteriophages use a large number of different bacterial cell envelope structures as receptors for surface attachment. As a consequence, bacterial surfaces represent a major control point for the defense against phage attack. One strategy for phage population control is the production of outer membrane vesicles (OMVs). In Gram-negative host bacteria, O-antigen-specific bacteriophages address lipopolysaccharide (LPS) to initiate infection, thus relying on an essential outer membrane glycan building block as receptor that is constantly present also in OMVs. In this work, we have analyzed interactions of Salmonella (S.) bacteriophage P22 with OMVs. For this, we isolated OMVs that were formed in large amounts during mechanical cell lysis of the P22 S. Typhimurium host. In vitro, these OMVs could efficiently reduce the number of infective phage particles. Fluorescence spectroscopy showed that upon interaction with OMVs, bacteriophage P22 released its DNA into the vesicle lumen. However, only about one third of the phage P22 particles actively ejected their genome. For the larger part, no genome release was observed, albeit the majority of phages in the system had lost infectivity towards their host. With OMVs, P22 ejected its DNA more rapidly and could release more DNA against elevated osmotic pressures compared to DNA release triggered with protein-free LPS aggregates. This emphasizes that OMV composition is a key feature for the regulation of infective bacteriophage particles in the system.
Collapse
Affiliation(s)
- Mareike S Stephan
- Physical Biochemistry, Department for Biochemistry and Biology, University of Potsdam, Potsdam, Germany
| | - Nina K Broeker
- Physical Biochemistry, Department for Biochemistry and Biology, University of Potsdam, Potsdam, Germany
| | | | - Norbert Roos
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Dirk Linke
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Stefanie Barbirz
- Physical Biochemistry, Department for Biochemistry and Biology, University of Potsdam, Potsdam, Germany
| |
Collapse
|
24
|
Structure, Formation, and Biological Interactions of Supported Lipid Bilayers (SLB) Incorporating Lipopolysaccharide. COATINGS 2020. [DOI: 10.3390/coatings10100981] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Biomimetic membrane systems play a crucial role in the field of biosensor engineering. Over the years, significant progress has been achieved creating artificial membranes by various strategies from vesicle fusion to Langmuir transfer approaches to meet an ever-growing demand for supported lipid bilayers on various substrates such as glass, mica, gold, polymer cushions, and many more. This paper reviews the diversity seen in the preparation of biologically relevant model lipid membranes which includes monolayers and bilayers of phospholipid and other crucial components such as proteins, characterization techniques, changes in the physical properties of the membranes during molecular interactions and the dynamics of the lipid membrane with biologically active molecules with special emphasis on lipopolysaccharides (LPS).
Collapse
|
25
|
Uribe J, Liu HY, Mohamed Z, Chiou AE, Fischbach C, Daniel S. Supported Membrane Platform to Assess Surface Interactions between Extracellular Vesicles and Stromal Cells. ACS Biomater Sci Eng 2020; 6:3945-3956. [PMID: 33463350 DOI: 10.1021/acsbiomaterials.0c00133] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Extracellular vesicles (EVs) are membrane-encapsulated particles secreted by eukaryotic cells that stimulate cell communication and horizontal cargo exchange. EV interactions with stromal cells can result in molecular changes in the recipient cell and, in some cases, lead to disease progression. However, mechanisms leading to these changes are poorly understood. A few model systems are available for studying the outcomes of surface interactions between EV membranes with stromal cells. Here, we created a hybrid supported bilayer incorporating EVs membrane material, called an extracellular vesicle supported bilayer, EVSB. Using EVSBs, we investigated the surface interactions between breast cancer EVs and adipose-derived stem cells (ADSCs) by culturing ADSCs on EVSBs and analyzing cell adhesion, spreading, viability, vascular endothelial growth factor (VEGF) secretion, and myofibroblast differentiation. Results show that cell viability, adhesion, spreading, and proangiogenic activity were enhanced, conditions that promote oncogenic activity, but cell differentiation was not. This model system could be used to develop therapeutic strategies to limit EV-ADSC interactions and proangiogenic conditions. Finally, this model system is not limited to the study of cancer but can be used to study surface interactions between EVs from any origin and any target cell to investigate EV mechanisms leading to cellular changes in other diseases.
Collapse
Affiliation(s)
- Johana Uribe
- Meinig School of Biomedical Engineering, Cornell University, 101 Weill Hall, 273 Tower Road, Ithaca, New York 14853, United States
| | - Han-Yuan Liu
- School of Chemical and Biomolecular Engineering, Cornell University, 120 Olin Hall, 6-44 Ho Plaza, Ithaca, New York 14853, United States
| | - Zeinab Mohamed
- Meinig School of Biomedical Engineering, Cornell University, 101 Weill Hall, 273 Tower Road, Ithaca, New York 14853, United States
| | - Aaron E Chiou
- Meinig School of Biomedical Engineering, Cornell University, 101 Weill Hall, 273 Tower Road, Ithaca, New York 14853, United States
| | - Claudia Fischbach
- Meinig School of Biomedical Engineering, Cornell University, 101 Weill Hall, 273 Tower Road, Ithaca, New York 14853, United States.,School of Chemical and Biomolecular Engineering, Cornell University, 120 Olin Hall, 6-44 Ho Plaza, Ithaca, New York 14853, United States
| | - Susan Daniel
- Meinig School of Biomedical Engineering, Cornell University, 101 Weill Hall, 273 Tower Road, Ithaca, New York 14853, United States.,School of Chemical and Biomolecular Engineering, Cornell University, 120 Olin Hall, 6-44 Ho Plaza, Ithaca, New York 14853, United States
| |
Collapse
|
26
|
Liu HY, Pappa AM, Pavia A, Pitsalidis C, Thiburce Q, Salleo A, Owens RM, Daniel S. Self-Assembly of Mammalian-Cell Membranes on Bioelectronic Devices with Functional Transmembrane Proteins. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2020; 36:7325-7331. [PMID: 32388991 DOI: 10.1021/acs.langmuir.0c00804] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Transmembrane proteins (TMPs) regulate processes occurring at the cell surface and are essential gatekeepers of information flow across the membrane. TMPs are difficult to study, given the complex environment of the membrane and its influence on protein conformation, mobility, biomolecule interaction, and activity. For the first time, we create mammalian biomembranes supported on a transparent, electrically conducting polymer surface, which enables dual electrical and optical monitoring of TMP function in its native membrane environment. Mammalian plasma membrane vesicles containing ATP-gated P2X2 ion channels self-assemble on a biocompatible polymer cushion that transduces the changes in ion flux during ATP exposure. This platform maintains the complexity of the native plasma membrane, the fluidity of its constituents, and protein orientation critical to ion channel function. We demonstrate the dual-modality readout using microscopy to characterize protein mobility by single-particle tracking and sensing of ATP gating of P2X2 using electrical impedance spectroscopy. This measurement of TMP activity important for pain sensing, neurological activity, and sensory activity raises new possibilities for drug screening and biosensing applications.
Collapse
Affiliation(s)
- Han-Yuan Liu
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Olin Hall, Ithaca, New York 14853, United States
| | - Anna-Maria Pappa
- Department of Chemical Engineering and BiotechnologyUniversity of Cambridge, Philippa Fawcett Drive, CB30AS Cambridge, UK
| | - Aimie Pavia
- Department of Flexible Electronics, Ecole Nationale Supérieure des Mines, CMP-EMSE, 13541 Gardanne, France
- Panaxium SAS, 13100 Aix-en-Provence, France
| | - Charalampos Pitsalidis
- Department of Chemical Engineering and BiotechnologyUniversity of Cambridge, Philippa Fawcett Drive, CB30AS Cambridge, UK
| | - Quentin Thiburce
- Department of Materials Science and Engineering, Stanford University, 496 Lomita Mall, Stanford, California 94305, United States
| | - Alberto Salleo
- Department of Materials Science and Engineering, Stanford University, 496 Lomita Mall, Stanford, California 94305, United States
| | - Róisín M Owens
- Department of Chemical Engineering and BiotechnologyUniversity of Cambridge, Philippa Fawcett Drive, CB30AS Cambridge, UK
| | - Susan Daniel
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Olin Hall, Ithaca, New York 14853, United States
| |
Collapse
|
27
|
Lakey JH. Recent advances in neutron reflectivity studies of biological membranes. Curr Opin Colloid Interface Sci 2019. [DOI: 10.1016/j.cocis.2019.02.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
28
|
Solvent-assisted preparation of supported lipid bilayers. Nat Protoc 2019; 14:2091-2118. [DOI: 10.1038/s41596-019-0174-2] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 04/02/2019] [Indexed: 11/08/2022]
|
29
|
Nomura K, Yamaguchi T, Mori S, Fujikawa K, Nishiyama KI, Shimanouchi T, Tanimoto Y, Morigaki K, Shimamoto K. Alteration of Membrane Physicochemical Properties by Two Factors for Membrane Protein Integration. Biophys J 2019; 117:99-110. [PMID: 31164197 PMCID: PMC6626835 DOI: 10.1016/j.bpj.2019.05.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 05/08/2019] [Accepted: 05/14/2019] [Indexed: 12/01/2022] Open
Abstract
After a nascent chain of a membrane protein emerges from the ribosomal tunnel, the protein is integrated into the cell membrane. This process is controlled by a series of proteinaceous molecular devices, such as signal recognition particles and Sec translocons. In addition to these proteins, we discovered two endogenous components regulating membrane protein integration in the inner membrane of Escherichia coli. The integration is blocked by diacylglycerol (DAG), whereas the blocking is relieved by a glycolipid named membrane protein integrase (MPIase). Here, we investigated the influence of these integration-blocking and integration-promoting factors on the physicochemical properties of membrane lipids via solid-state NMR and fluorescence measurements. These factors did not have destructive effects on membrane morphology because the membrane maintained its lamellar structure and did not fuse in the presence of DAG and/or MPIase at their effective concentrations. We next focused on membrane flexibility. DAG did not affect the mobility of the membrane surface, whereas the sugar chain in MPIase was highly mobile and enhanced the flexibility of membrane lipid headgroups. Comparison with a synthetic MPIase analog revealed the effects of the long sugar chain on membrane properties. The acyl chain order inside the membrane was increased by DAG, whereas the increase was cancelled by the addition of MPIase. MPIase also loosened the membrane lipid packing. Focusing on the transbilayer movement, MPIase reduced the rapid flip-flop motion of DAG. On the other hand, MPIase could not compensate for the diminished lateral diffusion by DAG. These results suggest that by manipulating the membrane lipids dynamics, DAG inhibits the protein from contacting the inner membrane, whereas the flexible long sugar chain of MPIase increases the opportunity for interaction between the membrane and the protein, leading to membrane integration of the newly formed protein.
Collapse
Affiliation(s)
- Kaoru Nomura
- Bioorganic Research Institute, Suntory Foundation for Life Sciences, Kyoto, Japan.
| | - Toshiyuki Yamaguchi
- Bioorganic Research Institute, Suntory Foundation for Life Sciences, Kyoto, Japan
| | - Shoko Mori
- Bioorganic Research Institute, Suntory Foundation for Life Sciences, Kyoto, Japan
| | - Kohki Fujikawa
- Bioorganic Research Institute, Suntory Foundation for Life Sciences, Kyoto, Japan
| | - Ken-Ichi Nishiyama
- Department of Biological Chemistry and Food Sciences, Faculty of Agriculture, Iwate University, Morioka, Iwate, Japan
| | | | - Yasushi Tanimoto
- Graduate School of Agricultural Science, Kobe University, Kobe, Japan
| | | | - Keiko Shimamoto
- Bioorganic Research Institute, Suntory Foundation for Life Sciences, Kyoto, Japan.
| |
Collapse
|
30
|
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
|
31
|
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
|
32
|
Tumurbaatar B, Kim MJ, Park CH, Kim CS. A portable and computer-simulation analysis for the real-time measurement of the QCMD systems for the biomedical application. SENSING AND BIO-SENSING RESEARCH 2018. [DOI: 10.1016/j.sbsr.2018.08.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
|
33
|
Pace HP, Hannestad JK, Armonious A, Adamo M, Agnarsson B, Gunnarsson A, Micciulla S, Sjövall P, Gerelli Y, Höök F. Structure and Composition of Native Membrane Derived Polymer-Supported Lipid Bilayers. Anal Chem 2018; 90:13065-13072. [PMID: 30350611 DOI: 10.1021/acs.analchem.8b04110] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Over the last two decades, supported lipid bilayers (SLBs) have been extensively used as model systems to study cell membrane structure and function. While SLBs have been traditionally produced from simple lipid mixtures, there has been a recent surge in compositional complexity to better mimic cellular membranes and thereby bridge the gap between classic biophysical approaches and cell experiments. To this end, native cellular membrane derived SLBs (nSLBs) have emerged as a new category of SLBs. As a new type of biomimetic material, an analytical workflow must be designed to characterize its molecular composition and structure. Herein, we demonstrate how a combination of fluorescence microscopy, neutron reflectometry, and secondary ion mass spectrometry offers new insights on structure, composition, and quality of nSLB systems formed using so-called hybrid vesicles, which are a mixture of native membrane material and synthetic lipids. With this approach, we demonstrate that the nSLB formed a continuous structure with complete mixing of the synthetic and native membrane components and a molecular stoichiometry that essentially mirrors that of the hybrid vesicles. Furthermore, structural investigation of the nSLB revealed that PEGylated lipids do not significantly thicken the hydration layer between the bilayer and substrate when on silicon substrates; however, nSLBs do have more topology than their simpler, purely synthetic counterparts. Beyond new insights regarding the structure and composition of nSLB systems, this work also serves to guide future researchers in producing and characterizing nSLBs from their cellular membrane of choice.
Collapse
Affiliation(s)
- Hudson P Pace
- Department of Physics , Chalmers University of Technology , SE-412 96 Göteborg , Sweden
| | - Jonas K Hannestad
- Department of Physics , Chalmers University of Technology , SE-412 96 Göteborg , Sweden.,Biosciences and Materials , Research Institutes of Sweden , SE-501 15 Borås , Sweden
| | - Antonious Armonious
- Department of Physics , Chalmers University of Technology , SE-412 96 Göteborg , Sweden
| | - Marco Adamo
- Institute Laue-Langevin , 38000 Grenoble , France.,Department of Chemical Engineering , Imperial College London , London SW7 2AZ , United Kingdom
| | - Bjorn Agnarsson
- Department of Physics , Chalmers University of Technology , SE-412 96 Göteborg , Sweden
| | - Anders Gunnarsson
- Discovery Sciences, IMED Biotech Unit , AstraZeneca , Gothenburg , Sweden
| | - Samantha Micciulla
- Institute Laue-Langevin , 38000 Grenoble , France.,Max Planck Institute of Colloids and Interfaces , 14476 Potsdam , Germany
| | - Peter Sjövall
- Department of Physics , Chalmers University of Technology , SE-412 96 Göteborg , Sweden.,Biosciences and Materials , Research Institutes of Sweden , SE-501 15 Borås , Sweden
| | - Yuri Gerelli
- Institute Laue-Langevin , 38000 Grenoble , France
| | - Fredrik Höök
- Department of Physics , Chalmers University of Technology , SE-412 96 Göteborg , Sweden
| |
Collapse
|
34
|
Vala M, Jordan LR, Warrington AE, Maher LJ, Rodriguez M, Wittenberg NJ, Oh SH. Surface Plasmon Resonance Sensing on Naturally Derived Membranes: A Remyelination-Promoting Human Antibody Binds Myelin with Extraordinary Affinity. Anal Chem 2018; 90:12567-12573. [DOI: 10.1021/acs.analchem.8b02664] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Milan Vala
- Department of Electrical and Computer Engineering, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Luke R. Jordan
- Department of Electrical and Computer Engineering, University of Minnesota, Minneapolis, Minnesota 55455, United States
- Department of Chemistry, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Arthur E. Warrington
- Departments of Neurology and Immunology, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - L. James Maher
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - Moses Rodriguez
- Departments of Neurology and Immunology, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - Nathan J. Wittenberg
- Department of Electrical and Computer Engineering, University of Minnesota, Minneapolis, Minnesota 55455, United States
- Department of Chemistry, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Sang-Hyun Oh
- Department of Electrical and Computer Engineering, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
35
|
Pitsalidis C, Pappa AM, Porel M, Artim CM, Faria GC, Duong DD, Alabi CA, Daniel S, Salleo A, Owens RM. Biomimetic Electronic Devices for Measuring Bacterial Membrane Disruption. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1803130. [PMID: 30117203 DOI: 10.1002/adma.201803130] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 06/28/2018] [Indexed: 06/08/2023]
Abstract
Antibiotic discovery has experienced a severe slowdown in terms of discovery of new candidates. In vitro screening methods using phospholipids to model the bacterial membrane provide a route to identify molecules that specifically disrupt bacterial membranes causing cell death. Thanks to the electrically insulating properties of the major component of the cell membrane, phospholipids, electronic devices are highly suitable transducers of membrane disruption. The organic electrochemical transistor (OECT) is a highly sensitive ion-to-electron converter. Here, the OECT is used as a transducer of the permeability of a lipid monolayer (ML) at a liquid:liquid interface, designed to read out changes in ion flux caused by compounds that interact with, and disrupt, lipid assembly. This concept is illustrated using the well-documented antibiotic Polymixin B and the highly sensitive quantitation of permeability of the lipid ML induced by two novel recently described antibacterial amine-based oligothioetheramides is shown, highlighting molecular scale differences in their disruption capabilities. It is anticipated that this platform has the potential to play a role in front-line antimicrobial compound design and characterization thanks to the compatibility of semiconductor microfabrication technology with high-throughput readouts.
Collapse
Affiliation(s)
- Charalampos Pitsalidis
- Department of Chemical Engineering and Biotechnology, Philippa Fawcett Drive, CB30AS, Cambridge, UK
| | - Anna-Maria Pappa
- Department of Chemical Engineering and Biotechnology, Philippa Fawcett Drive, CB30AS, Cambridge, UK
| | - Mintu Porel
- Department of Chemical and Biomolecular Engineering, Olin hall, Ithaca, NY, 14850, USA
| | - Christine M Artim
- Department of Chemical and Biomolecular Engineering, Olin hall, Ithaca, NY, 14850, USA
| | - Gregorio C Faria
- Department of Materials Science and Engineering, Stanford University, 496 Lomita Mall, Stanford, CA, 94305, USA
| | - Duc D Duong
- Department of Materials Science and Engineering, Stanford University, 496 Lomita Mall, Stanford, CA, 94305, USA
| | - Christopher A Alabi
- Department of Chemical and Biomolecular Engineering, Olin hall, Ithaca, NY, 14850, USA
| | - Susan Daniel
- Department of Chemical and Biomolecular Engineering, Olin hall, Ithaca, NY, 14850, USA
| | - Alberto Salleo
- Department of Materials Science and Engineering, Stanford University, 496 Lomita Mall, Stanford, CA, 94305, USA
| | - Róisín M Owens
- Department of Chemical Engineering and Biotechnology, Philippa Fawcett Drive, CB30AS, Cambridge, UK
| |
Collapse
|
36
|
Kakimoto Y, Tachihara Y, Okamoto Y, Miyazawa K, Fukuma T, Tero R. Morphology and Physical Properties of Hydrophilic-Polymer-Modified Lipids in Supported Lipid Bilayers. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2018; 34:7201-7209. [PMID: 29788718 DOI: 10.1021/acs.langmuir.8b00870] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Lipid molecules such as glycolipids that are modified with hydrophilic biopolymers participate in the biochemical reactions occurring on cell membranes. Their functions and efficiency are determined by the formation of microdomains and their physical properties. We investigated the morphology and properties of domains induced by the hydrophilic-polymer-modified lipid applying a polyethylene glycol (PEG)-modified lipid as a model modified lipid. We formed supported lipid bilayers (SLBs) using a 0-10 mol % range of PEG-modified lipid concentration ( CPEG). We studied their morphology and fluidity by fluorescence microscopy, the fluorescence recovery after photobleaching method, and atomic force microscopy (AFM). Fluorescence images showed that domains rich in the PEG-modified lipid appeared and SLB fluidity decreased when CPEG ≥ 5%. AFM topographies showed that clusters of the PEG-modified lipid appeared prior to domain formation and the PEG-lipid-rich domains were observed as depressions. Frequency-modulation AFM revealed a force-dependent appearance of the PEG-lipid-rich domain.
Collapse
Affiliation(s)
- Yasuhiro Kakimoto
- Department of Environmental and Life Sciences , Toyohashi University of Technology , Toyohashi , Aichi 441-8580 , Japan
| | - Yoshihiro Tachihara
- Department of Environmental and Life Sciences , Toyohashi University of Technology , Toyohashi , Aichi 441-8580 , Japan
| | - Yoshiaki Okamoto
- Department of Environmental and Life Sciences , Toyohashi University of Technology , Toyohashi , Aichi 441-8580 , Japan
| | - Keisuke Miyazawa
- Division of Electrical Engineering and Computer Science , Kanazawa University , Kakuma-machi, Kanazawa 920-1192 , Japan
| | - Takeshi Fukuma
- Division of Electrical Engineering and Computer Science , Kanazawa University , Kakuma-machi, Kanazawa 920-1192 , Japan
- Nano Life Science Institute (WPI-NanoLSI) , Kakuma-machi, Kanazawa 920-1192 , Japan
| | - Ryugo Tero
- Department of Environmental and Life Sciences , Toyohashi University of Technology , Toyohashi , Aichi 441-8580 , Japan
| |
Collapse
|
37
|
Peerboom N, Schmidt E, Trybala E, Block S, Bergström T, Pace HP, Bally M. Cell Membrane Derived Platform To Study Virus Binding Kinetics and Diffusion with Single Particle Sensitivity. ACS Infect Dis 2018; 4:944-953. [PMID: 29688001 DOI: 10.1021/acsinfecdis.7b00270] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Discovery and development of new antiviral therapies essentially rely on two key factors: an in-depth understanding of the mechanisms involved in viral infection and the development of fast and versatile drug screening platforms. To meet those demands, we present a biosensing platform to probe virus-cell membrane interactions on a single particle level. Our method is based on the formation of supported lipid bilayers from cell membrane material. Using total internal reflection fluorescence microscopy, we report the contribution of viral and cellular components to the interaction kinetics of herpes simplex virus type 1 with the cell membrane. Deletion of glycoprotein C (gC), the main viral attachment glycoprotein, or deletion of heparan sulfate, an attachment factor on the cell membrane, leads to an overall decrease in association of virions to the membrane and faster dissociation from the membrane. In addition to this, we perform binding inhibition studies using the antiviral compound heparin to estimate its IC50 value. Finally, single particle tracking is used to characterize the diffusive behavior of the virus particles on the supported lipid bilayers. Altogether, our results promote this platform as a complement to existing bioanalytical assays, being at the interface between simplified artificial membrane models and live cell experiments.
Collapse
Affiliation(s)
- Nadia Peerboom
- Department of Physics, Chalmers University of Technology, Fysikgränd 3, 412 96 Göteborg, Sweden
| | - Eneas Schmidt
- Department of Physics, Chalmers University of Technology, Fysikgränd 3, 412 96 Göteborg, Sweden
| | - Edward Trybala
- Department of Infectious Diseases, Section for Clinical Virology, Institute of Biomedicine, University of Gothenburg, Guldhedsgatan 10B, 413 46 Göteborg, Sweden
| | - Stephan Block
- Department of Chemistry and Biochemistry, Freie Universität Berlin, Takustraße 3, 141 95 Berlin, Germany
| | - Tomas Bergström
- Department of Infectious Diseases, Section for Clinical Virology, Institute of Biomedicine, University of Gothenburg, Guldhedsgatan 10B, 413 46 Göteborg, Sweden
| | - Hudson P. Pace
- Department of Physics, Chalmers University of Technology, Fysikgränd 3, 412 96 Göteborg, Sweden
| | - Marta Bally
- Department of Physics, Chalmers University of Technology, Fysikgränd 3, 412 96 Göteborg, Sweden
- Wallenberg Centre for Molecular Medicine and Department of Clinical Microbiology, Umeå University, NUS Målpunkt R, 901 85 Umeå, Sweden
| |
Collapse
|
38
|
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: 51] [Impact Index Per Article: 7.3] [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
|
39
|
Liu HY, Chen WL, Ober CK, Daniel S. Biologically Complex Planar Cell Plasma Membranes Supported on Polyelectrolyte Cushions Enhance Transmembrane Protein Mobility and Retain Native Orientation. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2018; 34:1061-1072. [PMID: 29020444 DOI: 10.1021/acs.langmuir.7b02945] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Reconstituted supported lipid bilayers (SLB) are widely used as in vitro cell-surface models because they are compatible with a variety of surface-based analytical techniques. However, one of the challenges of using SLBs as a model of the cell surface is the limited complexity in membrane composition, including the incorporation of transmembrane proteins and lipid diversity that may impact the activity of those proteins. Additionally, it is challenging to preserve the transmembrane protein native orientation, function, and mobility in SLBs. Here, we leverage the interaction between cell plasma membrane vesicles and polyelectrolyte brushes to create planar bilayers from cell plasma membrane vesicles that have budded from the cell surface. This approach promotes the direct incorporation of membrane proteins and other species into the planar bilayer without using detergent or reconstitution and preserves membrane constituents. Furthermore, the structure of the polyelectrolyte brush serves as a cushion between the planar bilayer and rigid supporting surface, limiting the interaction of the cytosolic domains of membrane proteins with this surface. Single particle tracking was used to analyze the motion of GPI-linked yellow fluorescent proteins (GPI-YFP) and neon-green fused transmembrane P2X2 receptors (P2X2-neon) and shows that this platform retains over 75% mobility of multipass transmembrane proteins in its native membrane environment. An enzyme accessibility assay confirmed that the protein orientation is preserved and results in the extracellular domain facing toward the bulk phase and the cytosolic side facing the support. Because the platform presented here retains the complexity of the cell plasma membrane and preserves protein orientation and mobility, it is a better representative mimic of native cell surfaces, which may find many applications in biological assays aimed at understanding cell membrane phenomena.
Collapse
Affiliation(s)
- Han-Yuan Liu
- Robert F. Smith School of Chemical and Biomolecular Engineering, ‡Department of Material Science and Engineering, Cornell University , Ithaca, New York 14853, United States
| | - Wei-Liang Chen
- Robert F. Smith School of Chemical and Biomolecular Engineering, ‡Department of Material Science and Engineering, Cornell University , Ithaca, New York 14853, United States
| | - Christopher K Ober
- Robert F. Smith School of Chemical and Biomolecular Engineering, ‡Department of Material Science and Engineering, Cornell University , Ithaca, New York 14853, United States
| | - Susan Daniel
- Robert F. Smith School of Chemical and Biomolecular Engineering, ‡Department of Material Science and Engineering, Cornell University , Ithaca, New York 14853, United States
| |
Collapse
|
40
|
Andersson J, Fuller MA, Wood K, Holt SA, Köper I. A tethered bilayer lipid membrane that mimics microbial membranes. Phys Chem Chem Phys 2018; 20:12958-12969. [DOI: 10.1039/c8cp01346b] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
This work presents a self-assembled lipid bilayer architecture mimicking the outer membrane of Gram negative bacteria.
Collapse
Affiliation(s)
- Jakob Andersson
- Flinders Centre for Nanoscale Science and Technology and College of Science and Engineering
- Flinders University
- Adelaide
- Australia
| | - Melanie A. Fuller
- Flinders Centre for Nanoscale Science and Technology and College of Science and Engineering
- Flinders University
- Adelaide
- Australia
| | - Kathleen Wood
- Australian Centre for Neutron Scattering
- Australian Nuclear Science and Technology Organisation
- Kirrawee DC
- Australia
| | - Stephen A. Holt
- Australian Centre for Neutron Scattering
- Australian Nuclear Science and Technology Organisation
- Kirrawee DC
- Australia
| | - Ingo Köper
- Flinders Centre for Nanoscale Science and Technology and College of Science and Engineering
- Flinders University
- Adelaide
- Australia
| |
Collapse
|
41
|
Tero R, Fukumoto K, Motegi T, Yoshida M, Niwano M, Hirano-Iwata A. Formation of Cell Membrane Component Domains in Artificial Lipid Bilayer. Sci Rep 2017; 7:17905. [PMID: 29263355 PMCID: PMC5738377 DOI: 10.1038/s41598-017-18242-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 12/07/2017] [Indexed: 12/31/2022] Open
Abstract
The lipid bilayer environment around membrane proteins strongly affects their structure and functions. Here, we aimed to study the fusion of proteoliposomes (PLs) derived from cultured cells with an artificial lipid bilayer membrane and the distribution of the PL components after the fusion. PLs, which were extracted as a crude membrane fraction from Chinese hamster ovary (CHO) cells, formed isolated domains in a supported lipid bilayer (SLB), comprising phosphatidylcholine (PC), phosphatidylethanolamine (PE), and cholesterol (Chol), after the fusion. Observation with a fluorescence microscope and an atomic force microscope showed that the membrane fusion occurred selectively at microdomains in the PC + PE + Chol-SLB, and that almost all the components of the PL were retained in the domain. PLs derived from human embryonic kidney 293 (HEK) cells also formed isolated domains in the PC + PE + Chol-SLB, but their fusion kinetics was different from that of the CHO-PLs. We attempted to explain the mechanism of the PL-SLB fusion and the difference between CHO- and HEK-PLs, based on a kinetic model. The domains that contained the whole cell membrane components provided environments similar to that of natural cell membranes, and were thus effective for studying membrane proteins using artificial lipid bilayer membranes.
Collapse
Affiliation(s)
- Ryugo Tero
- Department of Environmental and Life Sciences, Toyohashi University of Technology, Toyohashi, Aichi, 441-8580, Japan. .,Electronics-Inspired Interdisciplinary Research Institute, Toyohashi University of Technology, Toyohashi, Aichi, 441-8580, Japan.
| | - Kohei Fukumoto
- Department of Environmental and Life Sciences, Toyohashi University of Technology, Toyohashi, Aichi, 441-8580, Japan
| | - Toshinori Motegi
- Electronics-Inspired Interdisciplinary Research Institute, Toyohashi University of Technology, Toyohashi, Aichi, 441-8580, Japan.,Division of Molecular Science, Faculty of Science and Technology, Gunma University, Kiryu, Gunma, 376-8515, Japan
| | - Miyu Yoshida
- Laboratory for Nanoelectronics and Spintronics, Research Institute of Electrical Communication, Tohoku University, Sendai, Miyagi, 980-8577, Japan
| | - Michio Niwano
- Laboratory for Nanoelectronics and Spintronics, Research Institute of Electrical Communication, Tohoku University, Sendai, Miyagi, 980-8577, Japan.,Kansei Fukushi Research Institute, Tohoku Fukushi University, Sendai, Miyagi, 989-3201, Japan
| | - Ayumi Hirano-Iwata
- Laboratory for Nanoelectronics and Spintronics, Research Institute of Electrical Communication, Tohoku University, Sendai, Miyagi, 980-8577, Japan.,Advanced Institute for Materials Research, Tohoku University, Sendai, Miyagi, 980-8577, Japan
| |
Collapse
|
42
|
Liu HY, Grant H, Hsu HL, Sorkin R, Bošković F, Wuite G, Daniel S. Supported Planar Mammalian Membranes as Models of in Vivo Cell Surface Architectures. ACS APPLIED MATERIALS & INTERFACES 2017; 9:35526-35538. [PMID: 28930438 DOI: 10.1021/acsami.7b07500] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Emerging technologies use cell plasma membrane vesicles or "blebs" as an intermediate to form molecularly complete, planar cell surface mimetics that are compatible with a variety of characterization tools and microscopy methods. This approach enables direct incorporation of membrane proteins into supported lipid bilayers without using detergents and reconstitution and preserves native lipids and membrane species. Such a system can be advantageous as in vitro models of in vivo cell surfaces for study of the roles of membrane proteins as drug targets in drug delivery, host-pathogen interactions, tissue engineering, and many other bioanalytical and sensing applications. However, the impact of methods used to induce cell blebbing (vesiculation) on protein and membrane properties is still unknown. This study focuses on characterization of cell blebs created under various bleb-inducing conditions and the result on protein properties (orientation, mobility, activity, etc.) and lipid scrambling in this platform. The orientation of proteins in the cell blebs and planar bilayers is revealed using a protease cleavage assay. Lipid scrambling in both cell blebs and planar bilayers is indicated through an annexin V binding assay. To quantify protein confinement, immobility, etc., incorporation of GPI-linked yellow fluorescent protein (GPI-YFP) was used in conjunction with single-molecule tracking (SMT) microscopy. Finally, to investigate the impact of the bleb induction method on protein activity and expression level, cell blebs expressing human aminopeptidase N (hAPN) were analyzed by an enzyme activity assay and immunoblotting. This work enriches our understanding of cell plasma membrane bleb bilayers as a biomimetic platform, reveals conditions under which specific properties are met, and represents one of the few ways to make molecularly complete supported bilayers directly from cell plasma membranes.
Collapse
Affiliation(s)
- Han-Yuan Liu
- School of Chemical and Biomolecular Engineering, Cornell University , Ithaca, New York 14853, United States
| | - Hannah Grant
- School of Chemical and Biomolecular Engineering, Cornell University , Ithaca, New York 14853, United States
| | - Hung-Lun Hsu
- School of Chemical and Biomolecular Engineering, Cornell University , Ithaca, New York 14853, United States
| | - Raya Sorkin
- Department of Physics and Astronomy and Laser Lab, Vrije Universiteit Amsterdam , Amsterdam 1081 HV, The Netherlands
| | - Filip Bošković
- Department of Physics and Astronomy and Laser Lab, Vrije Universiteit Amsterdam , Amsterdam 1081 HV, The Netherlands
| | - Gijs Wuite
- Department of Physics and Astronomy and Laser Lab, Vrije Universiteit Amsterdam , Amsterdam 1081 HV, The Netherlands
| | - Susan Daniel
- School of Chemical and Biomolecular Engineering, Cornell University , Ithaca, New York 14853, United States
| |
Collapse
|