1
|
Mardešić I, Boban Z, Raguz M. Electroformation of Giant Unilamellar Vesicles from Damp Films in Conditions Involving High Cholesterol Contents, Charged Lipids, and Saline Solutions. MEMBRANES 2024; 14:215. [PMID: 39452827 PMCID: PMC11510074 DOI: 10.3390/membranes14100215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/01/2024] [Accepted: 10/11/2024] [Indexed: 10/26/2024]
Abstract
Giant unilamellar vesicles (GUVs) are frequently used as membrane models in studies of membrane properties. They are most often produced using the electroformation method. However, there are a number of parameters that can influence the success of the procedure. Some of the most common conditions that have been shown to have a negative effect on GUV electroformation are the presence of high cholesterol (Chol) concentrations, the use of mixtures containing charged lipids, and the solutions with an elevated ionic strength. High Chol concentrations are problematic for the traditional electroformation protocol as it involves the formation of a dry lipid film by complete evaporation of the organic solvent from the lipid mixture. During drying, anhydrous Chol crystals form. They are not involved in the formation of the lipid bilayer, resulting in a lower Chol concentration in the vesicle bilayer compared to the original lipid mixture. Motivated primarily by the issue of artifactual Chol demixing, we have modified the electroformation protocol by incorporating the techniques of rapid solvent exchange (RSE), ultrasonication, plasma cleaning, and spin-coating for reproducible production of GUVs from damp lipid films. Aside from decreasing Chol demixing, we have shown that the method can also be used to produce GUVs from lipid mixtures with charged lipids and in ionic solutions used as internal solutions. A high yield of GUVs was obtained for Chol/1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC) samples with mixing ratios ranging from 0 to 2.5. We also succeeded in preparing GUVs from mixtures containing up to 60 mol% of the charged lipid 1-palmitoyl-2-oleoyl-sn-glycero-3-phospho-L-serine (POPS) and in NaCl solutions with low ionic strength (<25 mM).
Collapse
Affiliation(s)
- Ivan Mardešić
- Department of Medical Physics and Biophysics, University of Split School of Medicine, 21000 Split, Croatia; (I.M.); (Z.B.)
- Doctoral Study of Biophysics, Faculty of Science, University of Split, 21000 Split, Croatia
| | - Zvonimir Boban
- Department of Medical Physics and Biophysics, University of Split School of Medicine, 21000 Split, Croatia; (I.M.); (Z.B.)
| | - Marija Raguz
- Department of Medical Physics and Biophysics, University of Split School of Medicine, 21000 Split, Croatia; (I.M.); (Z.B.)
| |
Collapse
|
2
|
Waeterschoot J, Barniol-Xicota M, Verhelst S, Baatsen P, Koos E, Lammertyn J, Casadevall i Solvas X. Lipid vesicle formation by encapsulation of SMALPs in surfactant-stabilised droplets. Heliyon 2024; 10:e37915. [PMID: 39347415 PMCID: PMC11437848 DOI: 10.1016/j.heliyon.2024.e37915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 09/12/2024] [Accepted: 09/12/2024] [Indexed: 10/01/2024] Open
Abstract
Understanding the intricate functions of membrane proteins is pivotal in cell biology and drug discovery. The composition of the cell membrane is highly complex, with different types of membrane proteins and lipid species. Hence, studying cellular membranes in a complexity-reduced context is important to enhance our understanding of the roles of these different elements. However, reconstitution of membrane proteins in an environment that closely mimics the cell, like giant unilamellar vesicles (GUVs), remains challenging, often requiring detergents that compromise protein function. To address this challenge, we present a novel strategy to manufacture GUVs from styrene maleic acid lipid particles (SMALPs) that utilises surfactant-stabilised droplets as a template. As a first step towards the incorporation of membrane proteins, this work focusses on the conversion of pure lipid SMALPs in GUVs. To evaluate the method, we produced a new form of SMA linked to fluorescein, referred to as FSMA. We demonstrate the assembly of SMALPs at the surfactant-stabilised droplet interface, resulting in the formation of GUVs when released upon addition of a demulsifying agent. The released vesicles appear similar to electroformed vesicles imaged with confocal light microscopy, but a fluorescein leakage assay and cryo-TEM imaging reveal their porous nature, potentially as a result of residual interactions of SMA with the lipid bilayer. Our study represents a significant step towards opening new avenues for comprehensive protein research in a complexity-reduced, yet biologically relevant, setting.
Collapse
Affiliation(s)
- Jorik Waeterschoot
- Biomimetics Group, Division of Mechatronics, Biostatistics and Sensors (MeBios), Department of Biosystems, KU Leuven, Willem de Croylaan 42, 3001 Leuven, Belgium
| | - Marta Barniol-Xicota
- Department of Medicine and Life Sciences (MELIS), Barcelona Biomedical Research Park, Universitat Pompeu Fabra, Carrer Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Steven Verhelst
- Department of Cellular and Molecular Medicine, KU Leuven – University of Leuven, Herestraat 49, box 901b, 3000 Leuven, Belgium
| | - Pieter Baatsen
- Center for the Biology of Disease, VIB, Herestraat 49, Leuven, 3000, Belgium
| | - Erin Koos
- Soft Matter, Rheology and Technology (SMaRT) at KU Leuven, Celestijnenlaan 200J, 3000 Leuven, Belgium
| | - Jeroen Lammertyn
- Biosensors Group, Division of Mechatronics, Biostatistics and Sensors (MeBios), Department of Biosystems, KU Leuven, Willem de Croylaan 42, 3001 Heverlee, Belgium
| | - Xavier Casadevall i Solvas
- Biomimetics Group, Division of Mechatronics, Biostatistics and Sensors (MeBios), Department of Biosystems, KU Leuven, Willem de Croylaan 42, 3001 Leuven, Belgium
| |
Collapse
|
3
|
Karanth S, Benthin J, Wiesenfarth M, Somoza V, Koehler M. Nanodisc Technology: Direction toward Physicochemical Characterization of Chemosensory Membrane Proteins in Food Flavor Research. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:14521-14529. [PMID: 38906535 PMCID: PMC11228972 DOI: 10.1021/acs.jafc.4c01827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/31/2024] [Accepted: 06/11/2024] [Indexed: 06/23/2024]
Abstract
Chemosensory membrane proteins such as G-protein-coupled receptors (GPCRs) drive flavor perception of food formulations. To achieve this, a detailed understanding of the structure and function of these membrane proteins is needed, which is often limited by the extraction and purification methods involved. The proposed nanodisc methodology helps overcome some of these existing challenges such as protein stability and solubilization along with their reconstitution from a native cell-membrane environment. Being well-established in structural biology procedures, nanodiscs offer this elegant solution by using, e.g., a membrane scaffold protein (MSP) or styrene-maleic acid (SMA) polymer, which interacts directly with the cell membrane during protein reconstitution. Such derived proteins retain their biophysical properties without compromising the membrane architecture. Here, we seek to show that these lipidic systems can be explored for insights with a focus on chemosensory membrane protein morphology and structure, conformational dynamics of protein-ligand interactions, and binding kinetics to answer pending questions in flavor research. Additionally, the compatibility of nanodiscs across varied (labeled or label-free) techniques offers significant leverage, which has been highlighted here.
Collapse
Affiliation(s)
- Sanjai Karanth
- Leibniz
Institute for Food Systems Biology at the Technical University of
Munich, Lise-Meitner-Strasse 34, 85354 Freising, Germany
| | - Julia Benthin
- Leibniz
Institute for Food Systems Biology at the Technical University of
Munich, Lise-Meitner-Strasse 34, 85354 Freising, Germany
- TUM
Graduate School, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Alte Akademie 8, 85354 Freising, Germany
| | - Marina Wiesenfarth
- Leibniz
Institute for Food Systems Biology at the Technical University of
Munich, Lise-Meitner-Strasse 34, 85354 Freising, Germany
- TUM
Graduate School, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Alte Akademie 8, 85354 Freising, Germany
| | - Veronika Somoza
- Leibniz
Institute for Food Systems Biology at the Technical University of
Munich, Lise-Meitner-Strasse 34, 85354 Freising, Germany
- Chair
of Nutritional Systems Biology, Technical
University of Munich, 85354 Freising, Germany
- Department
of Physiological Chemistry, Faculty of Chemistry, University of Vienna, Vienna 1090, Austria
| | - Melanie Koehler
- Leibniz
Institute for Food Systems Biology at the Technical University of
Munich, Lise-Meitner-Strasse 34, 85354 Freising, Germany
- TUM
Junior Fellow at the Chair of Nutritional Systems Biology, Technical University of Munich, 85354 Freising, Germany
| |
Collapse
|
4
|
Schoenmakers LLJ, den Uijl MJ, Postma JL, van den Akker TAP, Huck WTS, Driessen AJM. SecYEG-mediated translocation in a model synthetic cell. Synth Biol (Oxf) 2024; 9:ysae007. [PMID: 38807757 PMCID: PMC11131593 DOI: 10.1093/synbio/ysae007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 04/19/2024] [Accepted: 05/07/2024] [Indexed: 05/30/2024] Open
Abstract
Giant unilamellar vesicles (GUVs) provide a powerful model compartment for synthetic cells. However, a key challenge is the incorporation of membrane proteins that allow for transport, energy transduction, compartment growth and division. Here, we have successfully incorporated the membrane protein complex SecYEG-the key bacterial translocase that is essential for the incorporation of newly synthesized membrane proteins-in GUVs. Our method consists of fusion of small unilamellar vesicles containing reconstituted SecYEG into GUVs, thereby forming SecGUVs. These are suitable for large-scale experiments while maintaining a high protein:lipid ratio. We demonstrate that incorporation of SecYEG into GUVs does not inhibit its translocation efficiency. Robust membrane protein functionalized proteo-GUVs are promising and flexible compartments for use in the formation and growth of synthetic cells.
Collapse
Affiliation(s)
- Ludo L J Schoenmakers
- Physical-Organic Chemistry, Institute for Molecules and Materials, Radboud University, Nijmegen 6525AJ, The Netherlands
| | - Max J den Uijl
- Groningen Biomolecular Sciences and Biotechnology, Molecular Biotechnology, University of Groningen, Groningen 9747 AG, The Netherlands
| | - Jelle L Postma
- General Instrumentation, Radboud University, Nijmegen 6525 AJ, The Netherlands
| | - Tim A P van den Akker
- Groningen Biomolecular Sciences and Biotechnology, Molecular Biotechnology, University of Groningen, Groningen 9747 AG, The Netherlands
| | - Wilhelm T S Huck
- Physical-Organic Chemistry, Institute for Molecules and Materials, Radboud University, Nijmegen 6525AJ, The Netherlands
| | - Arnold J M Driessen
- Groningen Biomolecular Sciences and Biotechnology, Molecular Biotechnology, University of Groningen, Groningen 9747 AG, The Netherlands
| |
Collapse
|
5
|
Mardešić I, Boban Z, Raguz M. Electroformation of Giant Unilamellar Vesicles from Damp Lipid Films with a Focus on Vesicles with High Cholesterol Content. MEMBRANES 2024; 14:79. [PMID: 38668107 PMCID: PMC11051717 DOI: 10.3390/membranes14040079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 03/20/2024] [Accepted: 03/26/2024] [Indexed: 04/28/2024]
Abstract
Giant unilamellar vesicles (GUVs) are membrane models used to study membrane properties. Electroformation is one of the methods used to produce GUVs. During electroformation protocol, dry lipid film is formed. The drying of the lipid film induces the cholesterol (Chol) demixing artifact, in which Chol forms anhydrous crystals which do not participate in the formation of vesicles. This leads to a lower Chol concentration in the vesicle bilayers compared to the Chol concentration in the initial lipid solution. To address this problem, we propose a novel electroformation protocol that includes rapid solvent exchange (RSE), plasma cleaning, and spin-coating methods to produce GUVs. We tested the protocol, focusing on vesicles with a high Chol content using different spin-coating durations and vesicle type deposition. Additionally, we compared the novel protocol using completely dry lipid film. The optimal spin-coating duration for vesicles created from the phosphatidylcholine/Chol mixture was 30 s. Multilamellar vesicles (MLVs), large unilamellar vesicles (LUVs) obtained by the extrusion of MLVs through 100 nm membrane pores and LUVs obtained by extrusion of previously obtained LUVs through 50 nm membrane pores, were deposited on an electrode for 1.5/1 Chol/phosphatidylcholine (POPC) lipid mixture, and the results were compared. Electroformation using all three deposited vesicle types resulted in a high GUV yield, but the deposition of LUVs obtained by the extrusion of MLVs through 100 nm membrane pores provided the most reproducible results. Using the deposition of these LUVs, we produced high yield GUVs for six different Chol concentrations (from 0% to 71.4%). Using a protocol that included dry lipid film GUVs resulted in lower yields and induced the Chol demixing artifact, proving that the lipid film should never be subjected to drying when the Chol content is high.
Collapse
Affiliation(s)
- Ivan Mardešić
- Department of Medical Physics and Biophysics, University of Split School of Medicine, 21000 Split, Croatia; (I.M.); (Z.B.)
- Doctoral Study of Biophysics, Faculty of Science, University of Split, 21000 Split, Croatia
| | - Zvonimir Boban
- Department of Medical Physics and Biophysics, University of Split School of Medicine, 21000 Split, Croatia; (I.M.); (Z.B.)
| | - Marija Raguz
- Department of Medical Physics and Biophysics, University of Split School of Medicine, 21000 Split, Croatia; (I.M.); (Z.B.)
| |
Collapse
|
6
|
Otrin N, Otrin L, Bednarz C, Träger TK, Hamdi F, Kastritis PL, Ivanov I, Sundmacher K. Protein-Rich Rafts in Hybrid Polymer/Lipid Giant Unilamellar Vesicles. Biomacromolecules 2024; 25:778-791. [PMID: 38190609 PMCID: PMC10865357 DOI: 10.1021/acs.biomac.3c00972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 12/20/2023] [Accepted: 12/20/2023] [Indexed: 01/10/2024]
Abstract
Considerable attention has been dedicated to lipid rafts due to their importance in numerous cell functions such as membrane trafficking, polarization, and signaling. Next to studies in living cells, artificial micrometer-sized vesicles with a minimal set of components are established as a major tool to understand the phase separation dynamics and their intimate interplay with membrane proteins. In parallel, mixtures of phospholipids and certain amphiphilic polymers simultaneously offer an interface for proteins and mimic this segregation behavior, presenting a tangible synthetic alternative for fundamental studies and bottom-up design of cellular mimics. However, the simultaneous insertion of complex and sensitive membrane proteins is experimentally challenging and thus far has been largely limited to natural lipids. Here, we present the co-reconstitution of the proton pump bo3 oxidase and the proton consumer ATP synthase in hybrid polymer/lipid giant unilamellar vesicles (GUVs) via fusion/electroformation. Variations of the current method allow for tailored reconstitution protocols and control of the vesicle size. In particular, mixing of protein-free and protein-functionalized nanosized vesicles in the electroformation film results in larger GUVs, while separate reconstitution of the respiratory enzymes enables higher ATP synthesis rates. Furthermore, protein labeling provides a synthetic mechanism for phase separation and protein sequestration, mimicking lipid- and protein-mediated domain formation in nature. The latter means opens further possibilities for re-enacting phenomena like supercomplex assembly or symmetry breaking and enriches the toolbox of bottom-up synthetic biology.
Collapse
Affiliation(s)
- Nika Otrin
- Process
Systems Engineering, Max Planck Institute
for Dynamics of Complex Technical Systems, Sandtorstrasse 1, 39106 Magdeburg, Germany
| | - Lado Otrin
- Process
Systems Engineering, Max Planck Institute
for Dynamics of Complex Technical Systems, Sandtorstrasse 1, 39106 Magdeburg, Germany
| | - Claudia Bednarz
- Process
Systems Engineering, Max Planck Institute
for Dynamics of Complex Technical Systems, Sandtorstrasse 1, 39106 Magdeburg, Germany
| | - Toni K. Träger
- Interdisciplinary
Research Center HALOmem and Institute of Biochemistry and Biotechnology, Martin Luther University Halle-Wittenberg, Biozentrum, 06120 Halle/Saale, Germany
| | - Farzad Hamdi
- Interdisciplinary
Research Center HALOmem and Institute of Biochemistry and Biotechnology, Martin Luther University Halle-Wittenberg, Biozentrum, 06120 Halle/Saale, Germany
| | - Panagiotis L. Kastritis
- Interdisciplinary
Research Center HALOmem and Institute of Biochemistry and Biotechnology, Martin Luther University Halle-Wittenberg, Biozentrum, 06120 Halle/Saale, Germany
- Institute
of Chemical Biology, National Hellenic Research
Foundation, 11635 Athens, Greece
| | - Ivan Ivanov
- Process
Systems Engineering, Max Planck Institute
for Dynamics of Complex Technical Systems, Sandtorstrasse 1, 39106 Magdeburg, Germany
- Grup
de Biotecnologia Molecular i Industrial, Department of Chemical Engineering, Universitat Politècnica de Catalunya, Rambla Sant Nebridi 22, 08222 Terrassa, Spain
| | - Kai Sundmacher
- Process
Systems Engineering, Max Planck Institute
for Dynamics of Complex Technical Systems, Sandtorstrasse 1, 39106 Magdeburg, Germany
| |
Collapse
|
7
|
Maffeis V, Heuberger L, Nikoletić A, Schoenenberger C, Palivan CG. Synthetic Cells Revisited: Artificial Cells Construction Using Polymeric Building Blocks. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305837. [PMID: 37984885 PMCID: PMC10885666 DOI: 10.1002/advs.202305837] [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: 08/18/2023] [Revised: 10/06/2023] [Indexed: 11/22/2023]
Abstract
The exponential growth of research on artificial cells and organelles underscores their potential as tools to advance the understanding of fundamental biological processes. The bottom-up construction from a variety of building blocks at the micro- and nanoscale, in combination with biomolecules is key to developing artificial cells. In this review, artificial cells are focused upon based on compartments where polymers are the main constituent of the assembly. Polymers are of particular interest due to their incredible chemical variety and the advantage of tuning the properties and functionality of their assemblies. First, the architectures of micro- and nanoscale polymer assemblies are introduced and then their usage as building blocks is elaborated upon. Different membrane-bound and membrane-less compartments and supramolecular structures and how they combine into advanced synthetic cells are presented. Then, the functional aspects are explored, addressing how artificial organelles in giant compartments mimic cellular processes. Finally, how artificial cells communicate with their surrounding and each other such as to adapt to an ever-changing environment and achieve collective behavior as a steppingstone toward artificial tissues, is taken a look at. Engineering artificial cells with highly controllable and programmable features open new avenues for the development of sophisticated multifunctional systems.
Collapse
Affiliation(s)
- Viviana Maffeis
- Department of ChemistryUniversity of BaselMattenstrasse 22BaselCH‐4002Switzerland
- NCCR‐Molecular Systems EngineeringBPR 1095, Mattenstrasse 24aBaselCH‐4058Switzerland
| | - Lukas Heuberger
- Department of ChemistryUniversity of BaselMattenstrasse 22BaselCH‐4002Switzerland
| | - Anamarija Nikoletić
- Department of ChemistryUniversity of BaselMattenstrasse 22BaselCH‐4002Switzerland
- Swiss Nanoscience InstituteUniversity of BaselKlingelbergstrasse 82BaselCH‐4056Switzerland
| | | | - Cornelia G. Palivan
- Department of ChemistryUniversity of BaselMattenstrasse 22BaselCH‐4002Switzerland
- NCCR‐Molecular Systems EngineeringBPR 1095, Mattenstrasse 24aBaselCH‐4058Switzerland
- Swiss Nanoscience InstituteUniversity of BaselKlingelbergstrasse 82BaselCH‐4056Switzerland
| |
Collapse
|
8
|
Giordani S, Marassi V, Zattoni A, Roda B, Reschiglian P. Liposomes characterization for market approval as pharmaceutical products: Analytical methods, guidelines and standardized protocols. J Pharm Biomed Anal 2023; 236:115751. [PMID: 37778202 DOI: 10.1016/j.jpba.2023.115751] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 09/13/2023] [Accepted: 09/24/2023] [Indexed: 10/03/2023]
Abstract
Liposomes are nano-sized lipid-based vesicles widely studied for their drug delivery capabilities. Compared to standard carries they exhibit better properties such as improved site-targeting and drug release, protection of drugs from degradation and clearance, and lower toxic side effects. At present, scientific literature is rich of studies regarding liposomes-based systems, while 14 types of liposomal products have been authorized to the market by EMA and FDA and many others have been approved by national agencies. Although the interest in nanodevices and nanomedicine has steadily increased in the last two decades the development of documentation regulating and standardizing all the phases of their development and quality control still suffers from major inadequacy due to the intrinsic complexity of nano-systems characterization. Many generic documents (Type 1) discussing guidelines for the study of nano-systems (lipidic and not) have been proposed while there is a lack of robust and standardized methods (Type 2 documents). As a result, a widespread of different techniques, approaches and methodologies are being used, generating results of variable quality and hard to compare with each other. Additionally, such documents are often subject to updates and rewriting further complicating the topic. Within this context the aim of this work is focused on bridging the gap in liposome characterization: the most recent standardized methodologies suitable for liposomes characterization are here reported (with the corresponding Type 2 documents) and revised in a short and pragmatical way focused on providing the reader with a practical background of the state of the art. In particular, this paper will put the accent on the methodologies developed to evaluate the main critical quality attributes (CQAs) necessary for liposomes market approval.
Collapse
Affiliation(s)
- Stefano Giordani
- Department of Chemistry "Giacomo Ciamician", University of Bologna, 40126 Bologna, Italy
| | - Valentina Marassi
- Department of Chemistry "Giacomo Ciamician", University of Bologna, 40126 Bologna, Italy; byFlow srl, 40129 Bologna, Italy.
| | - Andrea Zattoni
- Department of Chemistry "Giacomo Ciamician", University of Bologna, 40126 Bologna, Italy; byFlow srl, 40129 Bologna, Italy
| | - Barbara Roda
- Department of Chemistry "Giacomo Ciamician", University of Bologna, 40126 Bologna, Italy; byFlow srl, 40129 Bologna, Italy.
| | - Pierluigi Reschiglian
- Department of Chemistry "Giacomo Ciamician", University of Bologna, 40126 Bologna, Italy; byFlow srl, 40129 Bologna, Italy
| |
Collapse
|
9
|
Nair KS, Bajaj H. Advances in giant unilamellar vesicle preparation techniques and applications. Adv Colloid Interface Sci 2023; 318:102935. [PMID: 37320960 DOI: 10.1016/j.cis.2023.102935] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 05/23/2023] [Accepted: 06/05/2023] [Indexed: 06/17/2023]
Abstract
Giant unilamellar vesicles (GUVs) are versatile and promising cell-sized bio-membrane mimetic platforms. Their applications range from understanding and quantifying membrane biophysical processes to acting as elementary blocks in the bottom-up assembly of synthetic cells. Definite properties and requisite goals in GUVs are dictated by the preparation techniques critical to the success of their applications. Here, we review key advances in giant unilamellar vesicle preparation techniques and discuss their formation mechanisms. Developments in lipid hydration and emulsion techniques for GUV preparation are described. Novel microfluidic-based techniques involving lipid or surfactant-stabilized emulsions are outlined. GUV immobilization strategies are summarized, including gravity-based settling, covalent linking, and immobilization by microfluidic, electric, and magnetic barriers. Moreover, some of the key applications of GUVs as biomimetic and synthetic cell platforms during the last decade have been identified. Membrane interface processes like phase separation, membrane protein reconstitution, and membrane bending have been deciphered using GUVs. In addition, vesicles are also employed as building blocks to construct synthetic cells with defined cell-like functions comprising compartments, metabolic reactors, and abilities to grow and divide. We critically discuss the pros and cons of preparation technologies and the properties they confer to the GUVs and identify potential techniques for dedicated applications.
Collapse
Affiliation(s)
- Karthika S Nair
- Microbial Processes and Technology Division, CSIR- National Institute for Interdisciplinary Science and Technology (NIIST), Trivandrum 695019, Kerala, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-Human Resource Development Centre, Ghaziabad 201002, India
| | - Harsha Bajaj
- Microbial Processes and Technology Division, CSIR- National Institute for Interdisciplinary Science and Technology (NIIST), Trivandrum 695019, Kerala, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-Human Resource Development Centre, Ghaziabad 201002, India.
| |
Collapse
|
10
|
Lin AJ, Sihorwala AZ, Belardi B. Engineering Tissue-Scale Properties with Synthetic Cells: Forging One from Many. ACS Synth Biol 2023; 12:1889-1907. [PMID: 37417657 PMCID: PMC11017731 DOI: 10.1021/acssynbio.3c00061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/08/2023]
Abstract
In metazoans, living cells achieve capabilities beyond individual cell functionality by assembling into multicellular tissue structures. These higher-order structures represent dynamic, heterogeneous, and responsive systems that have evolved to regenerate and coordinate their actions over large distances. Recent advances in constructing micrometer-sized vesicles, or synthetic cells, now point to a future where construction of synthetic tissue can be pursued, a boon to pressing material needs in biomedical implants, drug delivery systems, adhesives, filters, and storage devices, among others. To fully realize the potential of synthetic tissue, inspiration has been and will continue to be drawn from new molecular findings on its natural counterpart. In this review, we describe advances in introducing tissue-scale features into synthetic cell assemblies. Beyond mere complexation, synthetic cells have been fashioned with a variety of natural and engineered molecular components that serve as initial steps toward morphological control and patterning, intercellular communication, replication, and responsiveness in synthetic tissue. Particular attention has been paid to the dynamics, spatial constraints, and mechanical strengths of interactions that drive the synthesis of this next-generation material, describing how multiple synthetic cells can act as one.
Collapse
Affiliation(s)
- Alexander J Lin
- Department of Chemistry, University of Texas at Austin, Austin, Texas 78712, United States
| | - Ahmed Z Sihorwala
- McKetta Department of Chemical Engineering, University of Texas at Austin, Austin, Texas 78712, United States
| | - Brian Belardi
- McKetta Department of Chemical Engineering, University of Texas at Austin, Austin, Texas 78712, United States
| |
Collapse
|
11
|
Chen J, Agrawal S, Yi H, Vallejo D, Agrawal A, Lee AP. Cell-Sized Lipid Vesicles as Artificial Antigen-Presenting Cells for Antigen-Specific T Cell Activation. Adv Healthc Mater 2023; 12:e2203163. [PMID: 36645182 PMCID: PMC10175210 DOI: 10.1002/adhm.202203163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 12/22/2022] [Indexed: 01/17/2023]
Abstract
In this study, efficient T cell activation is demonstrated using cell-sized artificial antigen-presenting cells (aAPCs) with protein-conjugated bilayer lipid membranes that mimic biological cell membranes. The highly uniform aAPCs are generated by a facile method based on standard droplet microfluidic devices. These aAPCs are able to activate the T cells in peripheral blood mononuclear cells, showing a 28-fold increase in interferon gamma (IFNγ) secretion, a 233-fold increase in antigen-specific CD8 T cells expansion, and a 16-fold increase of CD4 T cell expansion. The aAPCs do not require repetitive boosting or additional stimulants and can function at a relatively low aAPC-to-T cell ratio (1:17). The research presents strong evidence that the surface fluidity and size of the aAPCs are critical to the effective formation of immune synapses essential for T cell activation. The findings demonstrate that the microfluidic-generated aAPCs can be instrumental in investigating the physiological conditions and mechanisms for T cell activation. Finally, this method demonstrates the feasibility of customizable aAPCs for a cost-effective off-the-shelf approach to immunotherapy.
Collapse
Affiliation(s)
- Jui‐Yi Chen
- Biomedical EngineeringUniversity of CaliforniaIrvineCA92617USA
| | | | - Hsiu‐Ping Yi
- Biomedical EngineeringUniversity of CaliforniaIrvineCA92617USA
| | - Derek Vallejo
- Biomedical EngineeringUniversity of CaliforniaIrvineCA92617USA
| | - Anshu Agrawal
- Department of MedicineUniversity of CaliforniaIrvineCA92617USA
| | - Abraham P. Lee
- Biomedical EngineeringUniversity of CaliforniaIrvineCA92617USA
| |
Collapse
|
12
|
Bailoni E, Partipilo M, Coenradij J, Grundel DAJ, Slotboom DJ, Poolman B. Minimal Out-of-Equilibrium Metabolism for Synthetic Cells: A Membrane Perspective. ACS Synth Biol 2023; 12:922-946. [PMID: 37027340 PMCID: PMC10127287 DOI: 10.1021/acssynbio.3c00062] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Indexed: 04/08/2023]
Abstract
Life-like systems need to maintain a basal metabolism, which includes importing a variety of building blocks required for macromolecule synthesis, exporting dead-end products, and recycling cofactors and metabolic intermediates, while maintaining steady internal physical and chemical conditions (physicochemical homeostasis). A compartment, such as a unilamellar vesicle, functionalized with membrane-embedded transport proteins and metabolic enzymes encapsulated in the lumen meets these requirements. Here, we identify four modules designed for a minimal metabolism in a synthetic cell with a lipid bilayer boundary: energy provision and conversion, physicochemical homeostasis, metabolite transport, and membrane expansion. We review design strategies that can be used to fulfill these functions with a focus on the lipid and membrane protein composition of a cell. We compare our bottom-up design with the equivalent essential modules of JCVI-syn3a, a top-down genome-minimized living cell with a size comparable to that of large unilamellar vesicles. Finally, we discuss the bottlenecks related to the insertion of a complex mixture of membrane proteins into lipid bilayers and provide a semiquantitative estimate of the relative surface area and lipid-to-protein mass ratios (i.e., the minimal number of membrane proteins) that are required for the construction of a synthetic cell.
Collapse
Affiliation(s)
- Eleonora Bailoni
- Department
of Biochemistry and Molecular Systems Biology, Groningen Biomolecular
Sciences and Biotechnology Institute, University
of Groningen, Nijenborgh
4, 9747 AG Groningen, The Netherlands
| | - Michele Partipilo
- Department
of Biochemistry and Molecular Systems Biology, Groningen Biomolecular
Sciences and Biotechnology Institute, University
of Groningen, Nijenborgh
4, 9747 AG Groningen, The Netherlands
| | - Jelmer Coenradij
- Department
of Biochemistry and Molecular Systems Biology, Groningen Biomolecular
Sciences and Biotechnology Institute, University
of Groningen, Nijenborgh
4, 9747 AG Groningen, The Netherlands
| | - Douwe A. J. Grundel
- Department
of Biochemistry and Molecular Systems Biology, Groningen Biomolecular
Sciences and Biotechnology Institute, University
of Groningen, Nijenborgh
4, 9747 AG Groningen, The Netherlands
| | - Dirk J. Slotboom
- Department
of Biochemistry and Molecular Systems Biology, Groningen Biomolecular
Sciences and Biotechnology Institute, University
of Groningen, Nijenborgh
4, 9747 AG Groningen, The Netherlands
| | - Bert Poolman
- Department
of Biochemistry and Molecular Systems Biology, Groningen Biomolecular
Sciences and Biotechnology Institute, University
of Groningen, Nijenborgh
4, 9747 AG Groningen, The Netherlands
| |
Collapse
|
13
|
Tosaka T, Kamiya K. Function Investigations and Applications of Membrane Proteins on Artificial Lipid Membranes. Int J Mol Sci 2023; 24:ijms24087231. [PMID: 37108393 PMCID: PMC10138308 DOI: 10.3390/ijms24087231] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/05/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
Membrane proteins play an important role in key cellular functions, such as signal transduction, apoptosis, and metabolism. Therefore, structural and functional studies of these proteins are essential in fields such as fundamental biology, medical science, pharmacology, biotechnology, and bioengineering. However, observing the precise elemental reactions and structures of membrane proteins is difficult, despite their functioning through interactions with various biomolecules in living cells. To investigate these properties, methodologies have been developed to study the functions of membrane proteins that have been purified from biological cells. In this paper, we introduce various methods for creating liposomes or lipid vesicles, from conventional to recent approaches, as well as techniques for reconstituting membrane proteins into artificial membranes. We also cover the different types of artificial membranes that can be used to observe the functions of reconstituted membrane proteins, including their structure, number of transmembrane domains, and functional type. Finally, we discuss the reconstitution of membrane proteins using a cell-free synthesis system and the reconstitution and function of multiple membrane proteins.
Collapse
Affiliation(s)
- Toshiyuki Tosaka
- Division of Molecular Science, Graduate School of Science and Technology, Gunma University, Gunma 376-8515, Japan
| | - Koki Kamiya
- Division of Molecular Science, Graduate School of Science and Technology, Gunma University, Gunma 376-8515, Japan
| |
Collapse
|
14
|
Boban Z, Mardešić I, Jozić SP, Šumanovac J, Subczynski WK, Raguz M. Electroformation of Giant Unilamellar Vesicles from Damp Lipid Films Formed by Vesicle Fusion. MEMBRANES 2023; 13:352. [PMID: 36984739 PMCID: PMC10059949 DOI: 10.3390/membranes13030352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/14/2023] [Accepted: 03/16/2023] [Indexed: 06/18/2023]
Abstract
Giant unilamellar vesicles (GUVs) are artificial membrane models which are of special interest to researchers because of their similarity in size to eukaryotic cells. The most commonly used method for GUVs production is electroformation. However, the traditional electroformation protocol involves a step in which the organic solvent is completely evaporated, leaving behind a dry lipid film. This leads to artifactual demixing of cholesterol (Chol) in the form of anhydrous crystals. These crystals do not participate in the formation of the lipid bilayer, resulting in a decrease of Chol concentration in the bilayer compared to the initial lipid solution. We propose a novel electroformation protocol which addresses this issue by combining the rapid solvent exchange, plasma cleaning and spin-coating techniques to produce GUVs from damp lipid films in a fast and reproducible manner. We have tested the protocol efficiency using 1/1 phosphatidylcholine/Chol and 1/1/1 phosphatidylcholine/sphingomyelin/Chol lipid mixtures and managed to produce a GUV population of an average diameter around 40 µm, with many GUVs being larger than 100 µm. Additionally, compared to protocols that include the dry film step, the sizes and quality of vesicles determined from fluorescence microscopy images were similar or better, confirming the benefits of our protocol in that regard as well.
Collapse
Affiliation(s)
- Zvonimir Boban
- Department of Medical Physics and Biophysics, University of Split School of Medicine, 21000 Split, Croatia; (Z.B.); (I.M.)
- Faculty of Science, University of Split, Doctoral Study of Biophysics, 21000 Split, Croatia
| | - Ivan Mardešić
- Department of Medical Physics and Biophysics, University of Split School of Medicine, 21000 Split, Croatia; (Z.B.); (I.M.)
- Faculty of Science, University of Split, Doctoral Study of Biophysics, 21000 Split, Croatia
| | - Sanja Perinović Jozić
- Department of Organic Technology, Faculty of Chemistry and Technology, University of Split, 21000 Split, Croatia
| | - Josipa Šumanovac
- Department of Physics, Faculty of Science, University of Split, 21000 Split, Croatia
| | | | - Marija Raguz
- Department of Medical Physics and Biophysics, University of Split School of Medicine, 21000 Split, Croatia; (Z.B.); (I.M.)
| |
Collapse
|
15
|
van Buren L, Koenderink GH, Martinez-Torres C. DisGUVery: A Versatile Open-Source Software for High-Throughput Image Analysis of Giant Unilamellar Vesicles. ACS Synth Biol 2023; 12:120-135. [PMID: 36508359 PMCID: PMC9872171 DOI: 10.1021/acssynbio.2c00407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Indexed: 12/14/2022]
Abstract
Giant unilamellar vesicles (GUVs) are cell-sized aqueous compartments enclosed by a phospholipid bilayer. Due to their cell-mimicking properties, GUVs have become a widespread experimental tool in synthetic biology to study membrane properties and cellular processes. In stark contrast to the experimental progress, quantitative analysis of GUV microscopy images has received much less attention. Currently, most analysis is performed either manually or with custom-made scripts, which makes analysis time-consuming and results difficult to compare across studies. To make quantitative GUV analysis accessible and fast, we present DisGUVery, an open-source, versatile software that encapsulates multiple algorithms for automated detection and analysis of GUVs in microscopy images. With a performance analysis, we demonstrate that DisGUVery's three vesicle detection modules successfully identify GUVs in images obtained with a wide range of imaging sources, in various typical GUV experiments. Multiple predefined analysis modules allow the user to extract properties such as membrane fluorescence, vesicle shape, and internal fluorescence from large populations. A new membrane segmentation algorithm facilitates spatial fluorescence analysis of nonspherical vesicles. Altogether, DisGUVery provides an accessible tool to enable high-throughput automated analysis of GUVs, and thereby to promote quantitative data analysis in synthetic cell research.
Collapse
Affiliation(s)
- Lennard van Buren
- Department
of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, 2629 HZDelft, The Netherlands
| | - Gijsje Hendrika Koenderink
- Department
of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, 2629 HZDelft, The Netherlands
| | - Cristina Martinez-Torres
- Department
of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, 2629 HZDelft, The Netherlands
| |
Collapse
|
16
|
Lo CH, Zeng J. Application of polymersomes in membrane protein study and drug discovery: Progress, strategies, and perspectives. Bioeng Transl Med 2023; 8:e10350. [PMID: 36684106 PMCID: PMC9842050 DOI: 10.1002/btm2.10350] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 05/08/2022] [Accepted: 05/10/2022] [Indexed: 01/25/2023] Open
Abstract
Membrane proteins (MPs) play key roles in cellular signaling pathways and are responsible for intercellular and intracellular interactions. Dysfunctional MPs are directly related to the pathogenesis of various diseases, and they have been exploited as one of the most sought-after targets in the pharmaceutical industry. However, working with MPs is difficult given that their amphiphilic nature requires protection from biological membrane or membrane mimetics. Polymersomes are bilayered nano-vesicles made of self-assembled block copolymers that have been widely used as cell membrane mimetics for MP reconstitution and in engineering of artificial cells. This review highlights the prevailing trend in the application of polymersomes in MP study and drug discovery. We begin with a review on the techniques for synthesis and characterization of polymersomes as well as methods of MP insertion to form proteopolymersomes. Next, we review the structural and functional analysis of the different types of MPs reconstituted in polymersomes, including membrane transport proteins, MP complexes, and membrane receptors. We then summarize the factors affecting reconstitution efficiency and the quality of reconstituted MPs for structural and functional studies. Additionally, we discuss the potential in using proteopolymersomes as platforms for high-throughput screening (HTS) in drug discovery to identify modulators of MPs. We conclude by providing future perspectives and recommendations on advancing the study of MPs and drug development using proteopolymersomes.
Collapse
Affiliation(s)
- Chih Hung Lo
- Lee Kong Chian School of MedicineNanyang Technological UniversitySingaporeSingapore
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Jialiu Zeng
- Lee Kong Chian School of MedicineNanyang Technological UniversitySingaporeSingapore
- Department of Biomedical EngineeringBoston UniversityBostonMassachusettsUSA
- Department of ChemistryBoston UniversityBostonMassachusettsUSA
| |
Collapse
|
17
|
Fletcher M, Zhu J, Rubio-Sánchez R, Sandler SE, Nahas KA, Michele LD, Keyser UF, Tivony R. DNA-Based Optical Quantification of Ion Transport across Giant Vesicles. ACS NANO 2022; 16:17128-17138. [PMID: 36222833 PMCID: PMC9620405 DOI: 10.1021/acsnano.2c07496] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 10/06/2022] [Indexed: 06/16/2023]
Abstract
Accurate measurements of ion permeability through cellular membranes remains challenging due to the lack of suitable ion-selective probes. Here we use giant unilamellar vesicles (GUVs) as membrane models for the direct visualization of mass translocation at the single-vesicle level. Ion transport is indicated with a fluorescently adjustable DNA-based sensor that accurately detects sub-millimolar variations in K+ concentration. In combination with microfluidics, we employed our DNA-based K+ sensor for extraction of the permeation coefficient of potassium ions. We measured K+ permeability coefficients at least 1 order of magnitude larger than previously reported values from bulk experiments and show that permeation rates across the lipid bilayer increase in the presence of octanol. In addition, an analysis of the K+ flux in different concentration gradients allows us to estimate the complementary H+ flux that dissipates the charge imbalance across the GUV membrane. Subsequently, we show that our sensor can quantify the K+ transport across prototypical cation-selective ion channels, gramicidin A and OmpF, revealing their relative H+/K+ selectivity. Our results show that gramicidin A is much more selective to protons than OmpF with a H+/K+ permeability ratio of ∼104.
Collapse
Affiliation(s)
- Marcus Fletcher
- Cavendish
Laboratory, University of Cambridge, J.J. Thomson Avenue, CambridgeCB3 0HE, U.K.
| | - Jinbo Zhu
- Cavendish
Laboratory, University of Cambridge, J.J. Thomson Avenue, CambridgeCB3 0HE, U.K.
| | - Roger Rubio-Sánchez
- Department
of Chemistry, Molecular Sciences Research Hub, Imperial College London, LondonW12 0BZ, U.K.
- fabriCELL,
Molecular Sciences Research Hub, Imperial
College London, LondonW12 0BZ, U.K.
| | - Sarah E Sandler
- Cavendish
Laboratory, University of Cambridge, J.J. Thomson Avenue, CambridgeCB3 0HE, U.K.
| | - Kareem Al Nahas
- Cavendish
Laboratory, University of Cambridge, J.J. Thomson Avenue, CambridgeCB3 0HE, U.K.
| | - Lorenzo Di Michele
- Department
of Chemistry, Molecular Sciences Research Hub, Imperial College London, LondonW12 0BZ, U.K.
- fabriCELL,
Molecular Sciences Research Hub, Imperial
College London, LondonW12 0BZ, U.K.
| | - Ulrich F Keyser
- Cavendish
Laboratory, University of Cambridge, J.J. Thomson Avenue, CambridgeCB3 0HE, U.K.
| | - Ran Tivony
- Cavendish
Laboratory, University of Cambridge, J.J. Thomson Avenue, CambridgeCB3 0HE, U.K.
| |
Collapse
|
18
|
Dolder N, Müller P, von Ballmoos C. Experimental platform for the functional investigation of membrane proteins in giant unilamellar vesicles. SOFT MATTER 2022; 18:5877-5893. [PMID: 35916307 PMCID: PMC9364335 DOI: 10.1039/d2sm00551d] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Giant unilamellar vesicles (GUVs) are micrometer-sized model membrane systems that can be viewed directly under the microscope. They serve as scaffolds for the bottom-up creation of synthetic cells, targeted drug delivery and have been widely used to study membrane related phenomena in vitro. GUVs are also of interest for the functional investigation of membrane proteins that carry out many key cellular functions. A major hurdle to a wider application of GUVs in this field is the diversity of existing protocols that are optimized for individual proteins. Here, we compare PVA assisted and electroformation techniques for GUV formation under physiologically relevant conditions, and analyze the effect of immobilization on vesicle structure and membrane tightness towards small substrates and protons. There, differences in terms of yield, size, and leakage of GUVs produced by PVA assisted swelling and electroformation were found, dependent on salt and buffer composition. Using fusion of oppositely charged membranes to reconstitute a model membrane protein, we find that empty vesicles and proteoliposomes show similar fusion behavior, which allows for a rapid estimation of protein incorporation using fluorescent lipids.
Collapse
Affiliation(s)
- Nicolas Dolder
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, 3012 Bern, Switzerland.
| | - Philipp Müller
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, 3012 Bern, Switzerland.
| | - Christoph von Ballmoos
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, 3012 Bern, Switzerland.
| |
Collapse
|
19
|
Han WB, Kang DH, Kim TS. 3D Artificial Cell Membranes as Versatile Platforms for Biological Applications. BIOCHIP JOURNAL 2022. [DOI: 10.1007/s13206-022-00066-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
20
|
Podolsky KA, Masubuchi T, Debelouchina GT, Hui E, Devaraj NK. In Situ Assembly of Transmembrane Proteins from Expressed and Synthetic Components in Giant Unilamellar Vesicles. ACS Chem Biol 2022; 17:1015-1021. [PMID: 35482050 PMCID: PMC9255206 DOI: 10.1021/acschembio.2c00013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Reconstituting functional transmembrane (TM) proteins into model membranes is challenging due to the difficulty of expressing hydrophobic TM domains, which often require stabilizing detergents that can perturb protein structure and function. Recent model systems solve this problem by linking the soluble domains of membrane proteins to lipids, using noncovalent conjugation. Herein, we test an alternative solution involving the in vitro assembly of TM proteins from synthetic TM domains and expressed soluble domains using chemoselective peptide ligation. We developed an intein mediated ligation strategy to semisynthesize single-pass TM proteins in synthetic giant unilamellar vesicle (GUV) membranes by covalently attaching soluble protein domains to a synthetic TM polypeptide, avoiding the requirement for detergent. We show that the extracellular domain of programmed cell death protein 1, a mammalian immune checkpoint receptor, retains its ligand-binding function at a membrane interface after ligation to a synthetic TM peptide in GUVs, facilitating the study of receptor-ligand interactions.
Collapse
Affiliation(s)
- K. A. Podolsky
- Department of Chemistry and Biochemistry, University of California, San Diego, CA, U.S.A
| | - T. Masubuchi
- Section of Cell and Developmental Biology, Division of Biological Sciences, University of California, San Diego, CA, U.S.A
| | - G. T. Debelouchina
- Department of Chemistry and Biochemistry, University of California, San Diego, CA, U.S.A
| | - E. Hui
- Section of Cell and Developmental Biology, Division of Biological Sciences, University of California, San Diego, CA, U.S.A
| | - N. K. Devaraj
- Department of Chemistry and Biochemistry, University of California, San Diego, CA, U.S.A.,Corresponding Author: Neal K. Devaraj,
| |
Collapse
|
21
|
Laurence MJ, Carpenter TS, Laurence TA, Coleman MA, Shelby M, Liu C. Biophysical Characterization of Membrane Proteins Embedded in Nanodiscs Using Fluorescence Correlation Spectroscopy. MEMBRANES 2022; 12:membranes12040392. [PMID: 35448362 PMCID: PMC9028781 DOI: 10.3390/membranes12040392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 03/29/2022] [Accepted: 03/30/2022] [Indexed: 02/04/2023]
Abstract
Proteins embedded in biological membranes perform essential functions in all organisms, serving as receptors, transporters, channels, cell adhesion molecules, and other supporting cellular roles. These membrane proteins comprise ~30% of all human proteins and are the targets of ~60% of FDA-approved drugs, yet their extensive characterization using established biochemical and biophysical methods has continued to be elusive due to challenges associated with the purification of these insoluble proteins. In response, the development of nanodisc techniques, such as nanolipoprotein particles (NLPs) and styrene maleic acid polymers (SMALPs), allowed membrane proteins to be expressed and isolated in solution as part of lipid bilayer rafts with defined, consistent nanometer sizes and compositions, thus enabling solution-based measurements. Fluorescence correlation spectroscopy (FCS) is a relatively simple yet powerful optical microscopy-based technique that yields quantitative biophysical information, such as diffusion kinetics and concentrations, about individual or interacting species in solution. Here, we first summarize current nanodisc techniques and FCS fundamentals. We then provide a focused review of studies that employed FCS in combination with nanodisc technology to investigate a handful of membrane proteins, including bacteriorhodopsin, bacterial division protein ZipA, bacterial membrane insertases SecYEG and YidC, Yersinia pestis type III secretion protein YopB, yeast cell wall stress sensor Wsc1, epidermal growth factor receptor (EGFR), ABC transporters, and several G protein-coupled receptors (GPCRs).
Collapse
Affiliation(s)
- Matthew J. Laurence
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, CA 94550, USA; (M.J.L.); (T.S.C.); (M.A.C.)
| | - Timothy S. Carpenter
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, CA 94550, USA; (M.J.L.); (T.S.C.); (M.A.C.)
| | - Ted A. Laurence
- Materials Science Division, Lawrence Livermore National Laboratory, Livermore, CA 94550, USA;
| | - Matthew A. Coleman
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, CA 94550, USA; (M.J.L.); (T.S.C.); (M.A.C.)
- Department of Radiation Oncology, University of California Davis, Sacramento, CA 95616, USA
| | - Megan Shelby
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, CA 94550, USA; (M.J.L.); (T.S.C.); (M.A.C.)
- Correspondence: (M.S.); (C.L.)
| | - Chao Liu
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, CA 94550, USA; (M.J.L.); (T.S.C.); (M.A.C.)
- Correspondence: (M.S.); (C.L.)
| |
Collapse
|
22
|
Mathiassen PPM, Pomorski TG. A Fluorescence-based Assay for Measuring Phospholipid Scramblase Activity in Giant Unilamellar Vesicles. Bio Protoc 2022; 12:e4366. [PMID: 35434199 PMCID: PMC8983165 DOI: 10.21769/bioprotoc.4366] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 11/09/2021] [Accepted: 03/01/2022] [Indexed: 07/28/2023] Open
Abstract
Transbilayer movement of phospholipids in biological membranes is mediated by a diverse set of lipid transporters. Among them are scramblases that facilitate rapid bi-directional movement of lipids without metabolic energy input. In this protocol, we describe the incorporation of phospholipid scramblases into giant unilamellar vesicles (GUVs) formed from scramblase-containing large unilamellar vesicles by electroformation. We also describe how to analyze their activity using membrane-impermeant sodium dithionite, to bleach symmetrically incorporated fluorescent ATTO488-conjugated phospholipids. The fluorescence-based readout allows single vesicle tracking for a large number of settled/immobilized GUVs, and provides a well-defined experimental setup to directly characterize these lipid transporters at the molecular level. Graphic abstract: Giant unilamellar vesicles (GUVs) are formed by electroformation from large unilamellar vesicles (LUVs) containing phospholipid scramblases (purple) and trace amounts of a fluorescent lipid reporter (green). The scramblase activity is analyzed by a fluorescence-based assay of single GUVs, using the membrane-impermeant quencher dithionite. Sizes not to scale. Modified from Mathiassen et al. (2021).
Collapse
Affiliation(s)
- Patricia P. M. Mathiassen
- Department of Molecular Biochemistry, Faculty of Chemistry and Biochemistry, Ruhr University Bochum, 44780, Bochum, Germany
| | - Thomas Günther Pomorski
- Department of Molecular Biochemistry, Faculty of Chemistry and Biochemistry, Ruhr University Bochum, 44780, Bochum, Germany
- Department of Plant and Environmental Sciences, University of Copenhagen, 1871, Frederiksberg C, Denmark
| |
Collapse
|
23
|
Sato W, Zajkowski T, Moser F, Adamala KP. Synthetic cells in biomedical applications. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1761. [PMID: 34725945 PMCID: PMC8918002 DOI: 10.1002/wnan.1761] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 08/23/2021] [Accepted: 08/30/2021] [Indexed: 12/12/2022]
Abstract
Synthetic cells are engineered vesicles that can mimic one or more salient features of life. These features include directed localization, sense-and-respond behavior, gene expression, metabolism, and high stability. In nanomedicine, many of these features are desirable capabilities of drug delivery vehicles but are difficult to engineer. In this focus article, we discuss where synthetic cells offer unique advantages over nanoparticle and living cell therapies. We review progress in the engineering of the above life-like behaviors and how they are deployed in nanomedicine. Finally, we assess key challenges synthetic cells face before being deployed as drugs and suggest ways to overcome these challenges. This article is categorized under: Therapeutic Approaches and Drug Discovery > Emerging Technologies Biology-Inspired Nanomaterials > Lipid-Based Structures.
Collapse
Affiliation(s)
- Wakana Sato
- 1 Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN US
| | - Tomasz Zajkowski
- Centre of New Technologies, University of Warsaw, S. Banacha 2c, 02-097 Warsaw, Poland
- USRA at NASA Ames Research Center, Mountain View, CA 94035
- Blue Marble Space Institute of Science, 600 1st Avenue, Seattle WA 98104
| | - Felix Moser
- Synlife, Inc., One Kendall Square Suite B4401, Cambridge, MA 20139
| | - Katarzyna P. Adamala
- 1 Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN US
| |
Collapse
|
24
|
Lussier F, Schröter M, Diercks NJ, Jahnke K, Weber C, Frey C, Platzman I, Spatz JP. pH-Triggered Assembly of Endomembrane Multicompartments in Synthetic Cells. ACS Synth Biol 2022; 11:366-382. [PMID: 34889607 PMCID: PMC8787813 DOI: 10.1021/acssynbio.1c00472] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Indexed: 11/29/2022]
Abstract
By using electrostatic interactions as driving force to assemble vesicles, the droplet-stabilized method was recently applied to reconstitute and encapsulate proteins, or compartments, inside giant unilamellar vesicles (GUVs) to act as minimal synthetic cells. However, the droplet-stabilized approach exhibits low production efficiency associated with the troublesome release of the GUVs from the stabilized droplets, corresponding to a major hurdle for the droplet-stabilized approach. Herein, we report the use of pH as a potential trigger to self-assemble droplet-stabilized GUVs (dsGUVs) by either bulk or droplet-based microfluidics. Moreover, pH enables the generation of compartmentalized GUVs with flexibility and robustness. By co-encapsulating pH-sensitive small unilamellar vesicles (SUVs), negatively charged SUVs, and/or proteins, we show that acidification of the droplets efficiently produces dsGUVs while sequestrating the co-encapsulated material. Most importantly, the pH-mediated assembly of dsGUVs significantly improves the production efficiency of free-standing GUVs (i.e., released from the stabilizing-droplets) compared to its previous implementation.
Collapse
Affiliation(s)
- Félix Lussier
- Department
of Cellular Biophysics, Max Planck Institute
for Medical Research, Jahnstraße 29, D-69120 Heidelberg, Germany
- Institute
for Molecular Systems Engineering (IMSE), Heidelberg University, Im Neuenheimer Feld 225, D-69120 Heidelberg, Germany
| | - Martin Schröter
- Department
of Cellular Biophysics, Max Planck Institute
for Medical Research, Jahnstraße 29, D-69120 Heidelberg, Germany
- Institute
for Molecular Systems Engineering (IMSE), Heidelberg University, Im Neuenheimer Feld 225, D-69120 Heidelberg, Germany
| | - Nicolas J. Diercks
- Department
of Cellular Biophysics, Max Planck Institute
for Medical Research, Jahnstraße 29, D-69120 Heidelberg, Germany
- Institute
for Molecular Systems Engineering (IMSE), Heidelberg University, Im Neuenheimer Feld 225, D-69120 Heidelberg, Germany
| | - Kevin Jahnke
- Biophysical
Engineering Group, Max Planck Institute
for Medical Research, Jahnstraße 29, D-69120 Heidelberg, Germany
- Department
of Physics and Astronomy, Heidelberg University, D-69120 Heidelberg, Germany
| | - Cornelia Weber
- Department
of Cellular Biophysics, Max Planck Institute
for Medical Research, Jahnstraße 29, D-69120 Heidelberg, Germany
- Institute
for Molecular Systems Engineering (IMSE), Heidelberg University, Im Neuenheimer Feld 225, D-69120 Heidelberg, Germany
| | - Christoph Frey
- Department
of Cellular Biophysics, Max Planck Institute
for Medical Research, Jahnstraße 29, D-69120 Heidelberg, Germany
- Institute
for Molecular Systems Engineering (IMSE), Heidelberg University, Im Neuenheimer Feld 225, D-69120 Heidelberg, Germany
| | - Ilia Platzman
- Department
of Cellular Biophysics, Max Planck Institute
for Medical Research, Jahnstraße 29, D-69120 Heidelberg, Germany
- Institute
for Molecular Systems Engineering (IMSE), Heidelberg University, Im Neuenheimer Feld 225, D-69120 Heidelberg, Germany
| | - Joachim P. Spatz
- Department
of Cellular Biophysics, Max Planck Institute
for Medical Research, Jahnstraße 29, D-69120 Heidelberg, Germany
- Institute
for Molecular Systems Engineering (IMSE), Heidelberg University, Im Neuenheimer Feld 225, D-69120 Heidelberg, Germany
- Max
Planck School Matter to Life, Jahnstraße 29, D-69120 Heidelberg, Germany
| |
Collapse
|
25
|
Carvalho BG, Ceccato BT, Michelon M, Han SW, de la Torre LG. Advanced Microfluidic Technologies for Lipid Nano-Microsystems from Synthesis to Biological Application. Pharmaceutics 2022; 14:141. [PMID: 35057037 PMCID: PMC8781930 DOI: 10.3390/pharmaceutics14010141] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 12/23/2021] [Accepted: 12/30/2021] [Indexed: 12/17/2022] Open
Abstract
Microfluidics is an emerging technology that can be employed as a powerful tool for designing lipid nano-microsized structures for biological applications. Those lipid structures can be used as carrying vehicles for a wide range of drugs and genetic materials. Microfluidic technology also allows the design of sustainable processes with less financial demand, while it can be scaled up using parallelization to increase production. From this perspective, this article reviews the recent advances in the synthesis of lipid-based nanostructures through microfluidics (liposomes, lipoplexes, lipid nanoparticles, core-shell nanoparticles, and biomimetic nanovesicles). Besides that, this review describes the recent microfluidic approaches to produce lipid micro-sized structures as giant unilamellar vesicles. New strategies are also described for the controlled release of the lipid payloads using microgels and droplet-based microfluidics. To address the importance of microfluidics for lipid-nanoparticle screening, an overview of how microfluidic systems can be used to mimic the cellular environment is also presented. Future trends and perspectives in designing novel nano and micro scales are also discussed herein.
Collapse
Affiliation(s)
- Bruna G. Carvalho
- Department of Material and Bioprocess Engineering, School of Chemical Engineering, University of Campinas (UNICAMP), Campinas 13083-852, Brazil; (B.G.C.); (B.T.C.)
| | - Bruno T. Ceccato
- Department of Material and Bioprocess Engineering, School of Chemical Engineering, University of Campinas (UNICAMP), Campinas 13083-852, Brazil; (B.G.C.); (B.T.C.)
| | - Mariano Michelon
- School of Chemical and Food Engineering, Federal University of Rio Grande (FURG), Rio Grande 96203-900, Brazil;
| | - Sang W. Han
- Center for Cell Therapy and Molecular, Department of Biophysics, Federal University of São Paulo (UNIFESP), São Paulo 04044-010, Brazil;
| | - Lucimara G. de la Torre
- Department of Material and Bioprocess Engineering, School of Chemical Engineering, University of Campinas (UNICAMP), Campinas 13083-852, Brazil; (B.G.C.); (B.T.C.)
| |
Collapse
|
26
|
Abstract
Hierarchic self-assembly underpins much of the form and function seen in synthetic or biological soft materials. Lipids are paramount examples, building themselves in nature or synthetically in a variety of meso/nanostructures. Synthetic block copolymers capture many of lipid's structural and functional properties. Lipids are typically biocompatible and high molecular weight polymers are mechanically robust and chemically versatile. The development of new materials for applications like controlled drug/gene/protein delivery, biosensors, and artificial cells often requires the combination of lipids and polymers. The emergent composite material, a "polymer-lipid hybrid membrane", displays synergistic properties not seen in pure components. Specific examples include the observation that hybrid membranes undergo lateral phase separation that can correlate in registry across multiple layers into a three-dimensional phase-separated system with enhanced permeability of encapsulated drugs. It is timely to underpin these emergent properties in several categories of hybrid systems ranging from colloidal suspensions to supported hybrid films. In this review, we discuss the form and function of a vast number of polymer-lipid hybrid systems published to date. We rationalize the results to raise new fundamental understanding of hybrid self-assembling soft materials as well as to enable the design of new supramolecular systems and applications.
Collapse
Affiliation(s)
- Yoo Kyung Go
- Department of Materials Science and Engineering, University of Illinois at Urbana─Champaign, Urbana, Illinois 61801, United States
| | - Cecilia Leal
- Department of Materials Science and Engineering, University of Illinois at Urbana─Champaign, Urbana, Illinois 61801, United States
| |
Collapse
|
27
|
Boban Z, Mardešić I, Subczynski WK, Raguz M. Giant Unilamellar Vesicle Electroformation: What to Use, What to Avoid, and How to Quantify the Results. MEMBRANES 2021; 11:membranes11110860. [PMID: 34832088 PMCID: PMC8622294 DOI: 10.3390/membranes11110860] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/03/2021] [Accepted: 11/05/2021] [Indexed: 12/12/2022]
Abstract
Since its inception more than thirty years ago, electroformation has become the most commonly used method for growing giant unilamellar vesicles (GUVs). Although the method seems quite straightforward at first, researchers must consider the interplay of a large number of parameters, different lipid compositions, and internal solutions in order to avoid artifactual results or reproducibility problems. These issues motivated us to write a short review of the most recent methodological developments and possible pitfalls. Additionally, since traditional manual analysis can lead to biased results, we have included a discussion on methods for automatic analysis of GUVs. Finally, we discuss possible improvements in the preparation of GUVs containing high cholesterol contents in order to avoid the formation of artifactual cholesterol crystals. We intend this review to be a reference for those trying to decide what parameters to use as well as an overview providing insight into problems not yet addressed or solved.
Collapse
Affiliation(s)
- Zvonimir Boban
- Department of Medical Physics and Biophysics, University of Split School of Medicine, 21000 Split, Croatia; (Z.B.); (I.M.)
- Doctoral Study of Biophysics, Faculty of Science, University of Split, 21000 Split, Croatia
| | - Ivan Mardešić
- Department of Medical Physics and Biophysics, University of Split School of Medicine, 21000 Split, Croatia; (Z.B.); (I.M.)
- Doctoral Study of Biophysics, Faculty of Science, University of Split, 21000 Split, Croatia
| | | | - Marija Raguz
- Department of Medical Physics and Biophysics, University of Split School of Medicine, 21000 Split, Croatia; (Z.B.); (I.M.)
- Correspondence: ; Tel.: +385-98-768-819
| |
Collapse
|
28
|
Islam MS, Gaston JP, Baker MAB. Fluorescence Approaches for Characterizing Ion Channels in Synthetic Bilayers. MEMBRANES 2021; 11:857. [PMID: 34832086 PMCID: PMC8619978 DOI: 10.3390/membranes11110857] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 10/25/2021] [Accepted: 10/28/2021] [Indexed: 12/12/2022]
Abstract
Ion channels are membrane proteins that play important roles in a wide range of fundamental cellular processes. Studying membrane proteins at a molecular level becomes challenging in complex cellular environments. Instead, many studies focus on the isolation and reconstitution of the membrane proteins into model lipid membranes. Such simpler, in vitro, systems offer the advantage of control over the membrane and protein composition and the lipid environment. Rhodopsin and rhodopsin-like ion channels are widely studied due to their light-interacting properties and are a natural candidate for investigation with fluorescence methods. Here we review techniques for synthesizing liposomes and for reconstituting membrane proteins into lipid bilayers. We then summarize fluorescence assays which can be used to verify the functionality of reconstituted membrane proteins in synthetic liposomes.
Collapse
Affiliation(s)
- Md. Sirajul Islam
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Kensington, NSW 2052, Australia; (M.S.I.); (J.P.G.)
| | - James P. Gaston
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Kensington, NSW 2052, Australia; (M.S.I.); (J.P.G.)
| | - Matthew A. B. Baker
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Kensington, NSW 2052, Australia; (M.S.I.); (J.P.G.)
- CSIRO Synthetic Biology Future Science Platform, GPO Box 2583, Brisbane, QLD 4001, Australia
| |
Collapse
|
29
|
Lopez Mora N, Findlay HE, Brooks NJ, Purushothaman S, Ces O, Booth PJ. The membrane transporter lactose permease increases lipid bilayer bending rigidity. Biophys J 2021; 120:3787-3794. [PMID: 34273316 PMCID: PMC8456183 DOI: 10.1016/j.bpj.2021.06.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 06/23/2021] [Accepted: 06/28/2021] [Indexed: 11/26/2022] Open
Abstract
Cellular life relies on membranes, which provide a resilient and adaptive cell boundary. Many essential processes depend upon the ease with which the membrane is able to deform and bend, features that can be characterized by the bending rigidity. Quantitative investigations of such mechanical properties of biological membranes have primarily been undertaken in solely lipid bilayers and frequently in the absence of buffers. In contrast, much less is known about the influence of integral membrane proteins on bending rigidity under physiological conditions. We focus on an exemplar member of the ubiquitous major facilitator superfamily of transporters and assess the influence of lactose permease on the bending rigidity of lipid bilayers. Fluctuation analysis of giant unilamellar vesicles (GUVs) is a useful means to measure bending rigidity. We find that using a hydrogel substrate produces GUVs that are well suited to fluctuation analysis. Moreover, the hydrogel method is amenable to both physiological salt concentrations and anionic lipids, which are important to mimic key aspects of the native lactose permease membrane. Varying the fraction of the anionic lipid in the lipid mixture DOPC/DOPE/DOPG allows us to assess the dependence of membrane bending rigidity on the topology and concentration of an integral membrane protein in the lipid bilayer of GUVs. The bending rigidity gradually increases with the incorporation of lactose permease, but there is no further increase with greater amounts of the protein in the membrane.
Collapse
Affiliation(s)
- Nestor Lopez Mora
- Department of Chemistry, Kings College London, London, United Kingdom
| | - Heather E Findlay
- Department of Chemistry, Kings College London, London, United Kingdom
| | - Nicholas J Brooks
- Department of Chemistry, Imperial College London, London, United Kingdom
| | - Sowmya Purushothaman
- Department of Chemistry, Imperial College London, London, United Kingdom; Beyond Meat, El Segundo, California
| | - Oscar Ces
- Department of Chemistry, Imperial College London, London, United Kingdom
| | - Paula J Booth
- Department of Chemistry, Kings College London, London, United Kingdom.
| |
Collapse
|
30
|
Majeed S, Ahmad AB, Sehar U, Georgieva ER. Lipid Membrane Mimetics in Functional and Structural Studies of Integral Membrane Proteins. MEMBRANES 2021; 11:685. [PMID: 34564502 PMCID: PMC8470526 DOI: 10.3390/membranes11090685] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/18/2021] [Accepted: 08/30/2021] [Indexed: 12/12/2022]
Abstract
Integral membrane proteins (IMPs) fulfill important physiological functions by providing cell-environment, cell-cell and virus-host communication; nutrients intake; export of toxic compounds out of cells; and more. However, some IMPs have obliterated functions due to polypeptide mutations, modifications in membrane properties and/or other environmental factors-resulting in damaged binding to ligands and the adoption of non-physiological conformations that prevent the protein from returning to its physiological state. Thus, elucidating IMPs' mechanisms of function and malfunction at the molecular level is important for enhancing our understanding of cell and organism physiology. This understanding also helps pharmaceutical developments for restoring or inhibiting protein activity. To this end, in vitro studies provide invaluable information about IMPs' structure and the relation between structural dynamics and function. Typically, these studies are conducted on transferred from native membranes to membrane-mimicking nano-platforms (membrane mimetics) purified IMPs. Here, we review the most widely used membrane mimetics in structural and functional studies of IMPs. These membrane mimetics are detergents, liposomes, bicelles, nanodiscs/Lipodisqs, amphipols, and lipidic cubic phases. We also discuss the protocols for IMPs reconstitution in membrane mimetics as well as the applicability of these membrane mimetic-IMP complexes in studies via a variety of biochemical, biophysical, and structural biology techniques.
Collapse
Affiliation(s)
- Saman Majeed
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX 79409, USA
| | - Akram Bani Ahmad
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX 79409, USA
| | - Ujala Sehar
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX 79409, USA
| | - Elka R Georgieva
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX 79409, USA
- Department of Cell Physiology and Molecular Biophysics, Texas Tech University Health Science Center, Lubbock, TX 79409, USA
| |
Collapse
|
31
|
Endoplasmic reticulum phospholipid scramblase activity revealed after protein reconstitution into giant unilamellar vesicles containing a photostable lipid reporter. Sci Rep 2021; 11:14364. [PMID: 34257324 PMCID: PMC8277826 DOI: 10.1038/s41598-021-93664-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 06/28/2021] [Indexed: 02/04/2023] Open
Abstract
Transbilayer movement of phospholipids in biological membranes is mediated by a diverse set of lipid transporters. Among them are scramblases that facilitate a rapid bi-directional movement of lipids without metabolic energy input. Here, we established a new fluorescence microscopy-based assay for detecting phospholipid scramblase activity of membrane proteins upon their reconstitution into giant unilamellar vesicles formed from proteoliposomes by electroformation. The assay is based on chemical bleaching of fluorescence of a photostable ATTO-dye labeled phospholipid with the membrane-impermeant reductant sodium dithionite. We demonstrate that this new methodology is suitable for the study of the scramblase activity of the yeast endoplasmic reticulum at single vesicle level.
Collapse
|
32
|
Souissi M, Pernier J, Rossier O, Giannone G, Le Clainche C, Helfer E, Sengupta K. Integrin-Functionalised Giant Unilamellar Vesicles via Gel-Assisted Formation: Good Practices and Pitfalls. Int J Mol Sci 2021; 22:6335. [PMID: 34199292 PMCID: PMC8231826 DOI: 10.3390/ijms22126335] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/26/2021] [Accepted: 06/08/2021] [Indexed: 01/16/2023] Open
Abstract
Giant unilamellar vesicles (GUV) are powerful tools to explore physics and biochemistry of the cell membrane in controlled conditions. For example, GUVs were extensively used to probe cell adhesion, but often using non-physiological linkers, due to the difficulty of incorporating transmembrane adhesion proteins into model membranes. Here we describe a new protocol for making GUVs incorporating the transmembrane protein integrin using gel-assisted swelling. We report an optimised protocol, enumerating the pitfalls encountered and precautions to be taken to maintain the robustness of the protocol. We characterise intermediate steps of small proteoliposome formation and the final formed GUVs. We show that the integrin molecules are successfully incorporated and are functional.
Collapse
Affiliation(s)
- Mariem Souissi
- Aix Marseille Univ, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille (CINAM), Turing Centre for Living Systems, 13009 Marseille, France;
| | - Julien Pernier
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198 Gif-sur-Yvette, France; (J.P.); (C.L.C.)
| | - Olivier Rossier
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, 33000 Bordeaux, France; (O.R.); (G.G.)
| | - Gregory Giannone
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, 33000 Bordeaux, France; (O.R.); (G.G.)
| | - Christophe Le Clainche
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198 Gif-sur-Yvette, France; (J.P.); (C.L.C.)
| | - Emmanuèle Helfer
- Aix Marseille Univ, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille (CINAM), Turing Centre for Living Systems, 13009 Marseille, France;
| | - Kheya Sengupta
- Aix Marseille Univ, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille (CINAM), Turing Centre for Living Systems, 13009 Marseille, France;
| |
Collapse
|
33
|
Mahapatra A, Uysalel C, Rangamani P. The Mechanics and Thermodynamics of Tubule Formation in Biological Membranes. J Membr Biol 2021; 254:273-291. [PMID: 33462667 PMCID: PMC8184589 DOI: 10.1007/s00232-020-00164-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 12/11/2020] [Indexed: 02/07/2023]
Abstract
Membrane tubulation is a ubiquitous process that occurs both at the plasma membrane and on the membranes of intracellular organelles. These tubulation events are known to be mediated by forces applied on the membrane either due to motor proteins, by polymerization of the cytoskeleton, or due to the interactions between membrane proteins binding onto the membrane. The numerous experimental observations of tube formation have been amply supported by mathematical modeling of the associated membrane mechanics and have provided insights into the force-displacement relationships of membrane tubes. Recent advances in quantitative biophysical measurements of membrane-protein interactions and tubule formation have necessitated the need for advances in modeling that will account for the interplay of multiple aspects of physics that occur simultaneously. Here, we present a comprehensive review of experimental observations of tubule formation and provide context from the framework of continuum modeling. Finally, we explore the scope for future research in this area with an emphasis on iterative modeling and experimental measurements that will enable us to expand our mechanistic understanding of tubulation processes in cells.
Collapse
Affiliation(s)
- Arijit Mahapatra
- Department of Mechanical and Aerospace Engineering, University of California San Diego, 9500 Gilman Dr, La Jolla, CA, 92093, USA
| | - Can Uysalel
- Department of Mechanical and Aerospace Engineering, University of California San Diego, 9500 Gilman Dr, La Jolla, CA, 92093, USA
| | - Padmini Rangamani
- Department of Mechanical and Aerospace Engineering, University of California San Diego, 9500 Gilman Dr, La Jolla, CA, 92093, USA.
| |
Collapse
|
34
|
Guo X, Steinkühler J, Marin M, Li X, Lu W, Dimova R, Melikyan GB. Interferon-Induced Transmembrane Protein 3 Blocks Fusion of Diverse Enveloped Viruses by Altering Mechanical Properties of Cell Membranes. ACS NANO 2021; 15:8155-8170. [PMID: 33656312 PMCID: PMC8159881 DOI: 10.1021/acsnano.0c10567] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Interferon-induced transmembrane protein 3 (IFITM3) potently inhibits entry of diverse enveloped viruses by trapping the viral fusion at a hemifusion stage, but the underlying mechanism remains unclear. Here, we show that recombinant IFITM3 reconstituted into lipid vesicles induces negative membrane curvature and that this effect maps to its small amphipathic helix (AH). We demonstrate that AH (i) partitions into lipid-disordered domains where IAV fusion occurs, (ii) induces negative membrane curvature, and (iii) increases lipid order and membrane stiffness. These effects on membrane properties correlate with the fusion-inhibitory activity, as targeting the ectopically expressed AH peptide to the cytoplasmic leaflet of the cell plasma membrane diminishes IAV-cell surface fusion induced by exposure to acidic pH. Our results thus imply that IFITM3 inhibits the transition from hemifusion to full fusion by imposing an unfavorable membrane curvature and increasing the order and stiffness of the cytoplasmic leaflet of endosomal membranes. Our findings reveal a universal mechanism by which cells block entry of diverse enveloped viruses.
Collapse
Affiliation(s)
- Xiangyang Guo
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, United States
| | - Jan Steinkühler
- Max Planck Institute of Colloids and Interfaces, 14424 Potsdam, Germany
| | - Mariana Marin
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, United States
| | - Xiang Li
- School of Pharmacy, Second Military Medical University, Shanghai, 200433, China
| | - Wuyuan Lu
- Institute of Human Virology and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
| | - Rumiana Dimova
- Max Planck Institute of Colloids and Interfaces, 14424 Potsdam, Germany
| | - Gregory B. Melikyan
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, United States
- Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
- Corresponding author: Gregory B. Melikyan,
| |
Collapse
|
35
|
Sych T, Gurdap CO, Wedemann L, Sezgin E. How Does Liquid-Liquid Phase Separation in Model Membranes Reflect Cell Membrane Heterogeneity? MEMBRANES 2021; 11:323. [PMID: 33925240 PMCID: PMC8146956 DOI: 10.3390/membranes11050323] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 04/22/2021] [Accepted: 04/26/2021] [Indexed: 11/22/2022]
Abstract
Although liquid-liquid phase separation of cytoplasmic or nuclear components in cells has been a major focus in cell biology, it is only recently that the principle of phase separation has been a long-standing concept and extensively studied in biomembranes. Membrane phase separation has been reconstituted in simplified model systems, and its detailed physicochemical principles, including essential phase diagrams, have been extensively explored. These model membrane systems have proven very useful to study the heterogeneity in cellular membranes, however, concerns have been raised about how reliably they can represent native membranes. In this review, we will discuss how phase-separated membrane systems can mimic cellular membranes and where they fail to reflect the native cell membrane heterogeneity. We also include a few humble suggestions on which phase-separated systems should be used for certain applications, and which interpretations should be avoided to prevent unreliable conclusions.
Collapse
Affiliation(s)
| | | | | | - Erdinc Sezgin
- Science for Life Laboratory, Department of Women’s and Children’s Health, Karolinska Institutet, 17165 Solna, Sweden; (T.S.); (C.O.G.); (L.W.)
| |
Collapse
|
36
|
Diederichs T, Tampé R. Single Cell-like Systems Reveal Active Unidirectional and Light-Controlled Transport by Nanomachineries. ACS NANO 2021; 15:6747-6755. [PMID: 33724767 PMCID: PMC8157534 DOI: 10.1021/acsnano.0c10139] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Cellular life depends on transport and communication across membranes, which is emphasized by the fact that membrane proteins are prime drug targets. The cell-like environment of membrane proteins has gained increasing attention based on its important role in function and regulation. As a versatile scaffold for bottom-up synthetic biology and nanoscience, giant liposomes represent minimalistic models of living cells. Nevertheless, the incorporation of fragile multiprotein membrane complexes still remains a major challenge. Here, we report on an approach for the functional reconstitution of membrane assemblies exemplified by human and bacterial ATP-binding cassette (ABC) transporters. We reveal that these nanomachineries transport substrates unidirectionally against a steep concentration gradient. Active substrate transport can be spatiotemporally resolved in single cell-like compartments by light, enabling real-time tracking of substrate export and import in individual liposomes. This approach will help to construct delicate artificial cell-like systems.
Collapse
Affiliation(s)
- Tim Diederichs
- Institute of Biochemistry, Biocenter,
Goethe-University Frankfurt, Max-von Laue-Straße 9,
60438 Frankfurt a.M., Germany
| | - Robert Tampé
- Institute of Biochemistry, Biocenter,
Goethe-University Frankfurt, Max-von Laue-Straße 9,
60438 Frankfurt a.M., Germany
| |
Collapse
|
37
|
Abstract
Giant unilamellar vesicles (GUVs) have gained great popularity as mimicries for cellular membranes. As their sizes are comfortably above the optical resolution limit, and their lipid composition is easily controlled, they are ideal for quantitative light microscopic investigation of dynamic processes in and on membranes. However, reconstitution of functional proteins into the lumen or the GUV membrane itself has proven technically challenging. In recent years, a selection of techniques has been introduced that tremendously improve GUV-assay development and enable the precise investigation of protein-membrane interactions under well-controlled conditions. Moreover, due to these methodological advances, GUVs are considered important candidates as protocells in bottom-up synthetic biology. In this review, we discuss the state of the art of the most important vesicle production and protein encapsulation methods and highlight some key protein systems whose functional reconstitution has advanced the field.
Collapse
Affiliation(s)
- Thomas Litschel
- Department of Cellular and Molecular Biophysics, Max Planck Institute of Biochemistry, Martinsried 82152, Germany; ,
| | - Petra Schwille
- Department of Cellular and Molecular Biophysics, Max Planck Institute of Biochemistry, Martinsried 82152, Germany; ,
| |
Collapse
|
38
|
Shukla S, Baumgart T. Enzymatic trans-bilayer lipid transport: Mechanisms, efficiencies, slippage, and membrane curvature. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2021; 1863:183534. [PMID: 33340491 PMCID: PMC8351443 DOI: 10.1016/j.bbamem.2020.183534] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 12/03/2020] [Accepted: 12/07/2020] [Indexed: 12/12/2022]
Abstract
The eukaryotic plasma membrane's lipid composition is found to be ubiquitously asymmetric comparing inner and outer leaflets. This membrane lipid asymmetry plays a crucial role in diverse cellular processes critical for cell survival. A specialized set of transmembrane proteins called translocases, or flippases, have evolved to maintain this membrane lipid asymmetry in an energy-dependent manner. One potential consequence of local variations in membrane lipid asymmetry is membrane remodeling, which is essential for cellular processes such as intracellular trafficking. Recently, there has been a surge in the identification and characterization of flippases, which has significantly advanced the understanding of their functional mechanisms. Furthermore, there are intriguing possibilities for a coupling between membrane curvature and flippase activity. In this review we highlight studies that link membrane shape and remodeling to differential stresses generated by the activity of lipid flippases with an emphasis on data obtained through model membrane systems. We review the common mechanistic models of flippase-mediated lipid flipping and discuss common techniques used to test lipid flippase activity. We then compare the existing data on lipid translocation rates by flippases and conclude with potential future directions for this field.
Collapse
Affiliation(s)
- Sankalp Shukla
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Tobias Baumgart
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA 19104, United States.
| |
Collapse
|
39
|
Giuliano CB, Cvjetan N, Ayache J, Walde P. Multivesicular Vesicles: Preparation and Applications. CHEMSYSTEMSCHEM 2021. [DOI: 10.1002/syst.202000049] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Camila Betterelli Giuliano
- Elvesys – Microfluidics Innovation Center 172 Rue de Charonne 75011 Paris France
- University of Strasbourg CNRS ISIS UMR 7006 67000 Strasbourg France
| | - Nemanja Cvjetan
- ETH Zürich Department of Materials Laboratory for Multifunctional Materials Vladimir-Prelog-Weg 5 8093 Zürich Switzerland
| | - Jessica Ayache
- Elvesys – Microfluidics Innovation Center 172 Rue de Charonne 75011 Paris France
| | - Peter Walde
- ETH Zürich Department of Materials Laboratory for Multifunctional Materials Vladimir-Prelog-Weg 5 8093 Zürich Switzerland
| |
Collapse
|
40
|
Wang X, Du H, Wang Z, Mu W, Han X. Versatile Phospholipid Assemblies for Functional Synthetic Cells and Artificial Tissues. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2002635. [PMID: 32830387 DOI: 10.1002/adma.202002635] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 06/09/2020] [Indexed: 06/11/2023]
Abstract
The bottom-up construction of a synthetic cell from nonliving building blocks capable of mimicking cellular properties and behaviors helps to understand the particular biophysical properties and working mechanisms of a cell. A synthetic cell built in this way possesses defined chemical composition and structure. Since phospholipids are native biomembrane components, their assemblies are widely used to mimic cellular structures. Here, recent developments in the formation of versatile phospholipid assemblies are described, together with the applications of these assemblies for functional membranes (protein reconstituted giant unilamellar vesicles), spherical and nonspherical protoorganelles, and functional synthetic cells, as well as the high-order hierarchical structures of artificial tissues. Their biomedical applications are also briefly summarized. Finally, the challenges and future directions in the field of synthetic cells and artificial tissues based on phospholipid assemblies are proposed.
Collapse
Affiliation(s)
- Xuejing Wang
- State Key Laboratory of Urban Water Resource and Environment, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China
| | - Hang Du
- State Key Laboratory of Urban Water Resource and Environment, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China
- Marine Antifouling Engineering Technology Center of Shangdong Province, Harbin Institute of Technology, Weihai, 264209, China
| | - Zhao Wang
- State Key Laboratory of Urban Water Resource and Environment, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China
| | - Wei Mu
- State Key Laboratory of Urban Water Resource and Environment, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China
| | - Xiaojun Han
- State Key Laboratory of Urban Water Resource and Environment, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China
| |
Collapse
|
41
|
Cho E, Lu Y. Compartmentalizing Cell-Free Systems: Toward Creating Life-Like Artificial Cells and Beyond. ACS Synth Biol 2020; 9:2881-2901. [PMID: 33095011 DOI: 10.1021/acssynbio.0c00433] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Building an artificial cell is a research area that is rigorously studied in the field of synthetic biology. It has brought about much attention with the aim of ultimately constructing a natural cell-like structure. In particular, with the more mature cell-free platforms and various compartmentalization methods becoming available, achieving this aim seems not far away. In this review, we discuss the various types of artificial cells capable of hosting several cellular functions. Different compartmental boundaries and the mature and evolving technologies that are used for compartmentalization are examined, and exciting recent advances that overcome or have the potential to address current challenges are discussed. Ultimately, we show how compartmentalization and cell-free systems have, and will, come together to fulfill the goal to assemble a fully synthetic cell that displays functionality and complexity as advanced as that in nature. The development of such artificial cell systems will offer insight into the fundamental study of evolutionary biology and the sea of applications as a result. Although several challenges remain, emerging technologies such as artificial intelligence also appear to help pave the way to address them and achieve the ultimate goal.
Collapse
Affiliation(s)
- Eunhee Cho
- Key Lab of Industrial Biocatalysis, Ministry of Education, Department of Chemical Engineering, Tsinghua University, Beijing 100084, China
| | - Yuan Lu
- Key Lab of Industrial Biocatalysis, Ministry of Education, Department of Chemical Engineering, Tsinghua University, Beijing 100084, China
| |
Collapse
|
42
|
Schmid YRF, Scheller L, Buchmann S, Dittrich PS. Calcium-Mediated Liposome Fusion to Engineer Giant Lipid Vesicles with Cytosolic Proteins and Reconstituted Mammalian Proteins. ACTA ACUST UNITED AC 2020; 4:e2000153. [PMID: 33084207 DOI: 10.1002/adbi.202000153] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 09/25/2020] [Indexed: 12/16/2022]
Abstract
Giant unilamellar lipid vesicles (GUVs) are widely used as model membrane systems and provide an excellent basis to construct artificial cells. To construct more sophisticated artificial cells, proteins-in particular membrane proteins-need to be incorporated in GUVs. However, current methods for protein reconstitution have limited throughput or are not generally applicable for all proteins because they depend on detergent solubilization. This limitation is addressed here by introducing calcium-mediated membrane fusion to transfer proteins between negatively charged GUVs and cell-derived plasma membrane vesicles (CDVs), derived from HEK293T cells overexpressing a membrane receptor protein. Fusion conditions are optimized using large unilamellar vesicles and GUVs containing phosphatidylserines and fusogenic lipids. The approach is then applied to induce lipid mixing and subsequent transfer of the overexpressed membrane receptor from CDVs into GUVs. The membrane receptor is detected by immunofluorescence on GUVs that underwent lipid mixing with CDVs. Those GUVs also exhibit esterase activity because cytosolic esterases entrapped in the CDVs are transferred during membrane fusion. Thus, content mixing is demonstrated. Using CDVs circumvents the need to purify or solubilize proteins. Moreover, calcium-mediated fusion allows transfer of lipids, water-soluble and membrane bound proteins in one step, resulting in a semi-synthetic cell.
Collapse
Affiliation(s)
- Yannick R F Schmid
- Department of Biosystems Science and Engineering, Bioanalytics Group, ETH Zürich, Mattenstrasse 26, Basel, CH-4058, Switzerland
| | - Leo Scheller
- Department of Biosystems Science and Engineering, Biotechnology and Bioengineering Group, ETH Zürich, Mattenstrasse 26, Basel, CH-4058, Switzerland
| | - Sebastian Buchmann
- Department of Biosystems Science and Engineering, Bioanalytics Group, ETH Zürich, Mattenstrasse 26, Basel, CH-4058, Switzerland
| | - Petra S Dittrich
- Department of Biosystems Science and Engineering, Bioanalytics Group, ETH Zürich, Mattenstrasse 26, Basel, CH-4058, Switzerland
| |
Collapse
|
43
|
Fan Y, Marioli M, Zhang K. Analytical characterization of liposomes and other lipid nanoparticles for drug delivery. J Pharm Biomed Anal 2020; 192:113642. [PMID: 33011580 DOI: 10.1016/j.jpba.2020.113642] [Citation(s) in RCA: 177] [Impact Index Per Article: 35.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 09/11/2020] [Accepted: 09/13/2020] [Indexed: 12/14/2022]
Abstract
Lipid nanoparticles, especially liposomes and lipid/nucleic acid complexed nanoparticles have shown great success in the pharmaceutical industry. Their success is attributed to stable drug loading, extended pharmacokinetics, reduced off-target side effects, and enhanced delivery efficiency to disease targets with formidable blood-brain or plasma membrane barriers. Therefore, they offer promising formulation options for drugs limited by low therapeutic indexes in traditional dosage forms and current "undruggable" targets. Recent development of siRNA, antisense oligonucleotide, or the CRISPR complex-loaded lipid nanoparticles and liposomal vaccines also shed light on their potential in enabling versatile formulation platforms for new pharmaceutical modalities. Analytical characterization of these nanoparticles is critical to drug design, formulation development, understanding in vivo performance, as well as quality control. The multi-lipid excipients, unique core-bilayer structure, and nanoscale size all underscore their complicated critical quality attributes, including lipid species, drug encapsulation efficiency, nanoparticle characteristics, product stability, and drug release. To address these challenges and facilitate future applications of lipid nanoparticles in drug development, we summarize available analytical approaches for physicochemical characterizations of lipid nanoparticle-based pharmaceutical modalities. Furthermore, we compare advantages and challenges of different techniques, and highlight the promise of new strategies for automated high-throughput screening and future development.
Collapse
Affiliation(s)
- Yuchen Fan
- Research and Early Development, Genentech Inc., 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Maria Marioli
- Pharma Technical Development Europe Analytics, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | - Kelly Zhang
- Research and Early Development, Genentech Inc., 1 DNA Way, South San Francisco, CA, 94080, USA.
| |
Collapse
|
44
|
Extraction of Membrane Components from Neisseria gonorrhoeae Using Catanionic Surfactant Vesicles: A New Approach for the Study of Bacterial Surface Molecules. Pharmaceutics 2020; 12:pharmaceutics12090787. [PMID: 32825235 PMCID: PMC7559012 DOI: 10.3390/pharmaceutics12090787] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/13/2020] [Accepted: 08/16/2020] [Indexed: 01/11/2023] Open
Abstract
Identification of antigens is important for vaccine production. We tested extraction protocols using cetyltrimethylammonium tosylate (CTAT) and sodium dodecylbenzenesulfonate (SDBS) to formulate surfactant vesicles (SVs) containing components from Neisseria gonorrhoeae. Carbohydrate and protein assays demonstrated that protein and carbohydrates were incorporated into the vesicle leaflet. Depending on the extraction protocol utilized, 100–400 µg of protein/mL of SVs solution was obtained. Gel electrophoresis followed by silver staining demonstrated that SV extracts contained lipooligosaccharide and a subset of bacterial proteins and lipoproteins. Western blotting and mass spectral analysis indicated that the majority of the proteins were derived from the outer membrane. Mass spectrometric and bioinformatics analysis of SVs identified 29 membrane proteins, including porin and opacity-associated protein. Proteins embedded in the SVs leaflet could be degraded by the addition of trypsin or proteinase K. Our data showed that the incorporation of CTAT and SDBS into vesicles eliminated their toxicity as measured by a THP-1 killing assay. Incorporation of gonococcal cell surface components into SVs reduced toxicity as compared to the whole cell extracts, as measured by cytokine induction, while retaining the immunogenicity. This process constitutes a general method for extracting bacterial surface components and identification of antigens that might be included in vaccines.
Collapse
|
45
|
Céspedes PF, Beckers D, Dustin ML, Sezgin E. Model membrane systems to reconstitute immune cell signaling. FEBS J 2020; 288:1070-1090. [DOI: 10.1111/febs.15488] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 06/26/2020] [Accepted: 07/14/2020] [Indexed: 12/26/2022]
Affiliation(s)
- Pablo F. Céspedes
- Kennedy Institute of Rheumatology Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences University of Oxford UK
| | - Daniel Beckers
- MRC Human Immunology Unit MRC Weatherall Institute of Molecular Medicine University of Oxford UK
| | - Michael L. Dustin
- Kennedy Institute of Rheumatology Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences University of Oxford UK
| | - Erdinc Sezgin
- MRC Human Immunology Unit MRC Weatherall Institute of Molecular Medicine University of Oxford UK
- Science for Life Laboratory Department of Women's and Children's Health Karolinska Institutet Stockholm Sweden
| |
Collapse
|
46
|
Kim YK, Choi Y, Nam GH, Kim IS. Functionalized exosome harboring bioactive molecules for cancer therapy. Cancer Lett 2020; 489:155-162. [PMID: 32623071 DOI: 10.1016/j.canlet.2020.05.036] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/26/2020] [Accepted: 05/27/2020] [Indexed: 12/17/2022]
Abstract
Exosomes are nanosized vesicles with a lipid membrane that are secreted by most cells and play a crucial role as intermediates of intercellular communication because they carry bioactive molecules. Exosomes are promising for drug delivery of chemicals, proteins, and nucleic acids owing to their inherent properties such as excellent biocompatibility, high tumor targetability, and prolonged circulation in vivo. In this review, we cover recent approaches and advances made in the field of exosome-mediated delivery of bioactive molecules for cancer therapy and factors that affect the clinical use of exosomes. This review can be used as a guideline for further study in expanding the utility of therapeutic exosomes.
Collapse
Affiliation(s)
- Yoon Kyoung Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea; Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea.
| | - Yoonjeong Choi
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea; Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Gi-Hoon Nam
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea; Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea.
| | - In-San Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea; Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea.
| |
Collapse
|
47
|
Marušič N, Otrin L, Zhao Z, Lira RB, Kyrilis FL, Hamdi F, Kastritis PL, Vidaković-Koch T, Ivanov I, Sundmacher K, Dimova R. Constructing artificial respiratory chain in polymer compartments: Insights into the interplay between bo3 oxidase and the membrane. Proc Natl Acad Sci U S A 2020; 117:15006-15017. [PMID: 32554497 PMCID: PMC7334566 DOI: 10.1073/pnas.1919306117] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Cytochrome bo3 ubiquinol oxidase is a transmembrane protein, which oxidizes ubiquinone and reduces oxygen, while pumping protons. Apart from its combination with F1Fo-ATPase to assemble a minimal ATP regeneration module, the utility of the proton pump can be extended to other applications in the context of synthetic cells such as transport, signaling, and control of enzymatic reactions. In parallel, polymers have been speculated to be phospholipid mimics with respect to their ability to self-assemble in compartments with increased stability. However, their usability as interfaces for complex membrane proteins has remained questionable. In the present work, we optimized a fusion/electroformation approach to reconstitute bo3 oxidase in giant unilamellar vesicles made of PDMS-g-PEO and/or phosphatidylcholine (PC). This enabled optical access, while microfluidic trapping allowed for online analysis of individual vesicles. The tight polymer membranes and the inward oriented enzyme caused 1 pH unit difference in 30 min, with an initial rate of 0.35 pH·min-1 To understand the interplay in these composite systems, we studied the relevant mechanical and rheological membrane properties. Remarkably, the proton permeability of polymer/lipid hybrids decreased after protein insertion, while the latter also led to a 20% increase of the polymer diffusion coefficient in polymersomes. In addition, PDMS-g-PEO increased the activity lifetime and the resistance to free radicals. These advantageous properties may open diverse applications, ranging from cell-free biotechnology to biomedicine. Furthermore, the presented study serves as a comprehensive road map for studying the interactions between membrane proteins and synthetic membranes, which will be fundamental for the successful engineering of such hybrid systems.
Collapse
Affiliation(s)
- Nika Marušič
- Process Systems Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, 39106 Magdeburg, Germany
| | - Lado Otrin
- Electrochemical Energy Conversion, Max Planck Institute for Dynamics of Complex Technical Systems, 39106 Magdeburg, Germany
| | - Ziliang Zhao
- Department of Theory and Bio-Systems, Max Planck Institute of Colloids and Interfaces, 14424 Potsdam, Germany
| | - Rafael B Lira
- Department of Theory and Bio-Systems, Max Planck Institute of Colloids and Interfaces, 14424 Potsdam, Germany
| | - Fotis L Kyrilis
- Interdisciplinary Research Center HALOmem, Martin Luther University Halle-Wittenberg, 06120 Halle/Saale, Germany
- Institute of Biochemistry and Biotechnology, Martin Luther University Halle-Wittenberg, 06120 Halle/Saale, Germany
| | - Farzad Hamdi
- Interdisciplinary Research Center HALOmem, Martin Luther University Halle-Wittenberg, 06120 Halle/Saale, Germany
- Institute of Biochemistry and Biotechnology, Martin Luther University Halle-Wittenberg, 06120 Halle/Saale, Germany
| | - Panagiotis L Kastritis
- Interdisciplinary Research Center HALOmem, Martin Luther University Halle-Wittenberg, 06120 Halle/Saale, Germany
- Institute of Biochemistry and Biotechnology, Martin Luther University Halle-Wittenberg, 06120 Halle/Saale, Germany
| | - Tanja Vidaković-Koch
- Electrochemical Energy Conversion, Max Planck Institute for Dynamics of Complex Technical Systems, 39106 Magdeburg, Germany;
| | - Ivan Ivanov
- Process Systems Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, 39106 Magdeburg, Germany;
| | - Kai Sundmacher
- Process Systems Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, 39106 Magdeburg, Germany
| | - Rumiana Dimova
- Department of Theory and Bio-Systems, Max Planck Institute of Colloids and Interfaces, 14424 Potsdam, Germany
| |
Collapse
|
48
|
Fusion Pore Formation Observed during SNARE-Mediated Vesicle Fusion with Pore-Spanning Membranes. Biophys J 2020; 119:151-161. [PMID: 32533941 DOI: 10.1016/j.bpj.2020.05.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 05/22/2020] [Accepted: 05/26/2020] [Indexed: 12/27/2022] Open
Abstract
Planar pore-spanning membranes (PSMs) have been shown to be a versatile tool to resolve elementary steps of the neuronal fusion process. However, in previous studies, we monitored only lipid mixing between fusing large unilamellar vesicles and PSMs and did not gather information about the formation of fusion pores. To address this important step of the fusion process, we entrapped sulforhodamine B at self-quenching concentrations into large unilamellar vesicles containing the v-SNARE synaptobrevin 2, which were docked and fused with lipid-labeled PSMs containing the t-SNARE acceptor complex ΔN49 prepared on gold-coated porous silicon substrates. By dual-color spinning disk fluorescence microscopy with a time resolution of ∼20 ms, we could unambiguously distinguish between bursting vesicles, which was only rarely observed (<0.01%), and fusion pore formation. From the time-resolved dual-color fluorescence time traces, we were able to identify different fusion pathways, including remaining three-dimensional postfusion structures with released content and transient openings and closings of the fusion pores. Our results on fusion pore formation and lipid diffusion from the PSM into the fusing vesicle let us conclude that the content release, i.e., fusion pore formation after the merger of the two lipid membranes occurs almost simultaneously.
Collapse
|
49
|
Kamiya K. Development of Artificial Cell Models Using Microfluidic Technology and Synthetic Biology. MICROMACHINES 2020; 11:E559. [PMID: 32486297 PMCID: PMC7345299 DOI: 10.3390/mi11060559] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 05/28/2020] [Accepted: 05/29/2020] [Indexed: 02/07/2023]
Abstract
Giant lipid vesicles or liposomes are primarily composed of phospholipids and form a lipid bilayer structurally similar to that of the cell membrane. These vesicles, like living cells, are 5-100 μm in diameter and can be easily observed using an optical microscope. As their biophysical and biochemical properties are similar to those of the cell membrane, they serve as model cell membranes for the investigation of the biophysical or biochemical properties of the lipid bilayer, as well as its dynamics and structure. Investigation of membrane protein functions and enzyme reactions has revealed the presence of soluble or membrane proteins integrated in the giant lipid vesicles. Recent developments in microfluidic technologies and synthetic biology have enabled the development of well-defined artificial cell models with complex reactions based on the giant lipid vesicles. In this review, using microfluidics, the formations of giant lipid vesicles with asymmetric lipid membranes or complex structures have been described. Subsequently, the roles of these biomaterials in the creation of artificial cell models including nanopores, ion channels, and other membrane and soluble proteins have been discussed. Finally, the complex biological functions of giant lipid vesicles reconstituted with various types of biomolecules has been communicated. These complex artificial cell models contribute to the production of minimal cells or protocells for generating valuable or rare biomolecules and communicating between living cells and artificial cell models.
Collapse
Affiliation(s)
- Koki Kamiya
- Division of Molecular Science, Graduate School of Science and Technology, Gunma University, 1-5-1 Tenjin-cho, Kiryu city, Gunma 376-8515, Japan
| |
Collapse
|
50
|
Hindley JW, Law RV, Ces O. Membrane functionalization in artificial cell engineering. SN APPLIED SCIENCES 2020. [DOI: 10.1007/s42452-020-2357-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
AbstractBottom-up synthetic biology aims to construct mimics of cellular structure and behaviour known as artificial cells from a small number of molecular components. The development of this nascent field has coupled new insights in molecular biology with large translational potential for application in fields such as drug delivery and biosensing. Multiple approaches have been applied to create cell mimics, with many efforts focusing on phospholipid-based systems. This mini-review focuses on different approaches to incorporating molecular motifs as tools for lipid membrane functionalization in artificial cell construction. Such motifs range from synthetic chemical functional groups to components from extant biology that can be arranged in a ‘plug-and-play’ approach which is hard to replicate in living systems. Rationally designed artificial cells possess the promise of complex biomimetic behaviour from minimal, highly engineered chemical networks.
Collapse
|