1
|
Ji J, Kawai S, Takagi R, Koiwai K, Kawano R, Tagawa Y. Generation of cell-sized liposomes using laser-induced microjets. LAB ON A CHIP 2025. [PMID: 40302460 DOI: 10.1039/d5lc00149h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2025]
Abstract
Cell-sized liposomes are microcapsules composed of a lipid bilayer, with potential applications in membrane science and synthetic biology. In this study, we present a novel method that employs high-speed laser-induced microjets to penetrate a lipid-carrying oil phase, thereby generating cell-sized liposomes. By simply triggering the microjets, we can reliably and repeatedly generate cell-sized liposomes, thereby enabling on-demand liposome production. We employed a high-speed camera to capture and analyze the penetration behavior of microjets. Additionally, we confirmed the unilamellarity of the liposomes using melittin, confirming their suitability for various biochemical applications. Furthermore, we conducted a numerical analysis to investigate potential factors influencing liposome formation in detail. These findings hold promise for advancing on-demand liposome production and contributing to the development of biochemical research.
Collapse
Affiliation(s)
- Jiajue Ji
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, Koganei, Japan.
| | - Shuma Kawai
- Mechanical Systems Engineering, Tokyo University of Agriculture and Technology, Koganei, Japan.
| | - Rina Takagi
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, Koganei, Japan.
| | - Keiichiro Koiwai
- Laboratory of Genome Science, Tokyo University of Marine Science and Technology, Minato, Japan
| | - Ryuji Kawano
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, Koganei, Japan.
| | - Yoshiyuki Tagawa
- Mechanical Systems Engineering, Tokyo University of Agriculture and Technology, Koganei, Japan.
| |
Collapse
|
2
|
Chen C, Love CM, Carnahan CF, Ganar KA, Parikh AN, Deshpande S. Regulating Biocondensates within Synthetic Cells via Segregative Phase Separation. ACS NANO 2025. [PMID: 40293809 DOI: 10.1021/acsnano.4c18971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Living cells orchestrate a myriad of biological reactions within a highly complex and crowded environment. A major factor responsible for such seamless assembly is the preferential interactions between the constituent macromolecules, that can drive demixing to produce coexisting phases and thus provide dynamic intracellular compartmentalization. However, the way multiple-phase separation phenomena, occurring simultaneously within the cytoplasmic space, influence each other is still largely unknown. Here, we show that the interplay between segregative and associative phase separation within cell-mimicking confinements can lead to rich dynamics between multiple phases and the lipid boundary. Using on-chip microfluidic systems, we encapsulate the associative and segregative components and externally trigger their phase separation within cell-sized vesicles. We find that segregative phases create microdomains and tend to dictate the fate of associative components by acting as molecular recruiters, membrane-targeting agents, and initiators of condensation. The obtained multiphase architecture provides an isolated microenvironment for condensates, restricting their molecular communication as well as diffusive motion, and can further lead to global shape transformation of the confinement itself in the form of wetted, hierarchical domains at the lipid membrane. In conclusion, we propose segregative phase separation as a universal condensation regulation strategy by managing their molecular distribution, process initiation, and spatial localization, including membrane interaction. The presented interplay between the two phase separation systems suggests a distinct design principle in constructing complex synthetic cells and controlling the behavior of artificial membraneless organelles within.
Collapse
Affiliation(s)
- Chang Chen
- Laboratory of Physical Chemistry and Soft Matter, Wageningen University & Research, 6708 WE Wageningen, The Netherlands
| | - Caroline M Love
- Department of Materials Science and Engineering, University of California, Davis, Davis, California 95616, United States
| | - Christopher F Carnahan
- Biophysics Graduate Group, University of California, Davis, Davis, California 95616, United States
| | - Ketan A Ganar
- Laboratory of Physical Chemistry and Soft Matter, Wageningen University & Research, 6708 WE Wageningen, The Netherlands
| | - Atul N Parikh
- Department of Materials Science and Engineering, University of California, Davis, Davis, California 95616, United States
- Biophysics Graduate Group, University of California, Davis, Davis, California 95616, United States
- Department of Biomedical Engineering, University of California, Davis, Davis, California 95616, United States
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, 636921 Singapore
- Institute for Digital Molecular Analytics and Science, Nanyang Technological University, 637551 Singapore
| | - Siddharth Deshpande
- Laboratory of Physical Chemistry and Soft Matter, Wageningen University & Research, 6708 WE Wageningen, The Netherlands
| |
Collapse
|
3
|
Yandrapalli N. Bottom-up development of lipid-based synthetic cells for practical applications. Trends Biotechnol 2025:S0167-7799(25)00094-0. [PMID: 40263003 DOI: 10.1016/j.tibtech.2025.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 03/10/2025] [Accepted: 03/11/2025] [Indexed: 04/24/2025]
Abstract
Synthetic cells (SCs) can be engineered from the bottom up to recapitulate the functional properties of natural cells while performing specialized tasks such as drug delivery, biosensors, bioproduction, vaccine development, and even environmental remediation. Recent advances in synthetic biology, biomaterials, and microfluidics have enabled the development of increasingly sophisticated SCs. Transitioning from proof-of-concept demonstrations to practical applications requires a deep understanding of the design principles, materials, and fabrication techniques involved. This review provides a comprehensive overview of the current state of bottom-up SC technology and highlights the most promising approaches and applications. Challenges in the implementation of SCs and their prospects for future applications are also discussed.
Collapse
Affiliation(s)
- Naresh Yandrapalli
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany.
| |
Collapse
|
4
|
Vink P, Honaker LW, Deshpande S. Towards electrospray-assisted production of lipid-based synthetic cell assemblies. SOFT MATTER 2025; 21:2977-2985. [PMID: 40035737 PMCID: PMC11878373 DOI: 10.1039/d4sm01284d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 02/25/2025] [Indexed: 03/06/2025]
Abstract
Lipid-based vesicles are widely used, minimalistic model containers for in vitro reconstitution of biological systems and engineering synthetic cells. These containers provide a micro-chassis to encapsulate biomolecules and study biochemical interactions. Liposomes are often the most sought-after vesicles owing to their cell-mimicking nature, and numerous bulk and on-chip methods exist for their production. However, exploring the scope of synthetic containers, both in terms of the alternative lipid assemblies as well as newer production methods is useful for expanding the toolbox for synthetic biology. In this paper, we report the development of an electrospray-based technique, which we term "ATPS-templated lipid assemblies via electrofusion of SUVs" (ATLAES), to form lipid-based vesicles. Using an aqueous two-phase system (ATPS), free of organic solvents, we demonstrate efficient formation of microscopic vesicles stabilized via interfacial lipid assembly. Interestingly, the formed vesicles exhibit a nebulous and disordered, but highly stable coating of lipids, and tend to form interconnected vesicle populations. Remarkably, the lipid assemblies can continue to rearrange and reconfigure over time, leading to spherical vesicles with ultra-thin and smooth lipid coating, suggestive of liposomes. Our work provides a new avenue, in the form of electrospray, to form various lipid-based assemblies using all-aqueous systems and we believe this platform can be further exploited for high-throughput vesicle production and higher-order assemblies.
Collapse
Affiliation(s)
- Pim Vink
- Laboratory of Physical Chemistry and Soft Matter, Wageningen University & Research, Wageningen, The Netherlands.
| | - Lawrence W Honaker
- Laboratory of Physical Chemistry and Soft Matter, Wageningen University & Research, Wageningen, The Netherlands.
| | - Siddharth Deshpande
- Laboratory of Physical Chemistry and Soft Matter, Wageningen University & Research, Wageningen, The Netherlands.
| |
Collapse
|
5
|
Dong D, Zhu T, Liao G, Tan F, Chen L, Yu Q, Wang L. Microfluidics-driven templating preparation of polymer vesicles with tailorable dimensions and rapid cellular internalization. Biomater Sci 2025. [PMID: 40237355 DOI: 10.1039/d5bm00377f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2025]
Abstract
Polymer vesicles hold immense potential in biomedicine and nanotechnology, yet conventional rehydration methods face critical limitations in controlling the vesicle architecture due to stochastic block copolymer (BCP) self-assembly. Here, we present a first-reported microsphere-templated strategy that synergizes microfluidic precision with BCP assembly to overcome these constraints. By engineering emulsion templates via flow rate, BCP concentration and collection distance optimization, we established a method based on the radius-square law governing the evolution of uniform vesicles (size range diameter: 70-170 nm, PDI: 0.16), enabling on-demand size tuning, a capability unattainable with traditional approaches. Multi-scale characterization (DLS, OM, SEM and TEM) elucidates the non-equilibrium templating-to-vesicle transition, revealing critical dynamics of BCP film reorganization. The resultant nano-scale vesicles exhibit rapid cellular uptake (>95% in 3 h) by HUVECs and 4T1 cells with exceptional biocompatibility (>85% viability, 36 h), outperforming many cytotoxic counterparts. This work not only provides a scalable platform for precision vesicle fabrication but also establishes foundational principles for templated self-assembly, bridging microfluidics and soft matter science. Our methodology opens avenues for tailored vesicles in drug delivery, nanoreactors and synthetic biology, addressing the persistent demand for functionally adaptive polymeric nanostructures.
Collapse
Affiliation(s)
- Donghua Dong
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, Guangdong Basic Research Center of Excellence for Energy and Information Polymer Materials, South China University of Technology, Guangzhou 510640, China.
| | - Tong Zhu
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, Guangdong Basic Research Center of Excellence for Energy and Information Polymer Materials, South China University of Technology, Guangzhou 510640, China.
| | - Guoxing Liao
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, Guangdong Basic Research Center of Excellence for Energy and Information Polymer Materials, South China University of Technology, Guangzhou 510640, China.
| | - Fangrong Tan
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, Guangdong Basic Research Center of Excellence for Energy and Information Polymer Materials, South China University of Technology, Guangzhou 510640, China.
| | - Lei Chen
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, Guangdong Basic Research Center of Excellence for Energy and Information Polymer Materials, South China University of Technology, Guangzhou 510640, China.
| | - Qianqian Yu
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, Guangdong Basic Research Center of Excellence for Energy and Information Polymer Materials, South China University of Technology, Guangzhou 510640, China.
| | - LinGe Wang
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, Guangdong Basic Research Center of Excellence for Energy and Information Polymer Materials, South China University of Technology, Guangzhou 510640, China.
| |
Collapse
|
6
|
Fletcher M, Elani Y. On-the-Fly Microfluidic Control of Giant Vesicle Compositions Validated by DNA Surface Charge Sensors. ACS NANO 2025; 19:13768-13778. [PMID: 40183490 PMCID: PMC12004935 DOI: 10.1021/acsnano.4c16289] [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: 11/13/2024] [Revised: 03/18/2025] [Accepted: 03/19/2025] [Indexed: 04/05/2025]
Abstract
The specific lipid composition of cell membrane microenvironments plays a critical role in regulating a range of cellular processes such as integral and peripheral membrane protein function, cell morphology, and membrane potential. However, harnessing similar complex capabilities in artificial membrane mimics remains challenging. In large part, progress has been slow due to a scarcity of techniques for both (i) accurately quantifying composition-dependent properties of artificial cell models at the single-vesicle level and (ii) efficiently exploring large multidimensional composition spaces. Here, we address both challenges by first developing an assay for quantitatively sensing giant unilamellar vesicle (GUV) membrane surface potentials using a fluorescent cholesterol-labeled DNA duplex sensor. We then devised a microfluidic vesicle assembly line enabling the continuous, on-chip production of lipid vesicles with variable compositions. This enabled real-time, on-the-fly adjustment of membrane compositions and biophysical properties as vesicles were being produced, followed by membrane analysis using our assay. Analysis of the association of our DNA fluorescent probe with single vesicles reveals that we may quantify the surface potential of vesicle membranes in situ through quantification of the membrane-probe binding constant. Our work paves the way for the production and biophysical analysis of artificial cell libraries that can enable rational artificial cell engineering.
Collapse
Affiliation(s)
- Marcus Fletcher
- Department of Chemical Engineering, Imperial College London, Exhibition Road, London SW7 2AZ, United
Kingdom
| | - Yuval Elani
- Department of Chemical Engineering, Imperial College London, Exhibition Road, London SW7 2AZ, United
Kingdom
| |
Collapse
|
7
|
Kwon H, Shin J, Sun S, Zhu R, Stainer S, Hinterdorfer P, Cho SJ, Kim DH, Oh YJ. Vertical DNA Nanostructure Arrays: Facilitating Functionalization on Macro-Scale Surfaces. ACS NANO 2025. [PMID: 40200829 DOI: 10.1021/acsnano.5c03100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/10/2025]
Abstract
The capability for varied functionalization and precise control at the nanoscale are significant advantages of DNA nanostructures. In the assembly of DNA nanostructure, the surface-assisted growth method utilizing double-crossover (DX) tile structures facilitates nucleation at relatively low concentrations on the surface based on electrostatic interactions, thereby enabling crystal growth over large areas. However, in surface-assisted growth, the geometrical hindrance of vertical structures on the DX tile structure surface makes it challenging to conjugate DNA nanostructures into fabricated surfaces. Here, the surface-assisted growth method was employed to extend the DX tile growth for forming vertical structure arrays on the substrate, providing attachment sites for functionalization on uniformly covered substrates at the macroscopic scale. Additionally, the spacing of the vertical structure arrays was demonstrated to be controllable through the strategic design of the repeating unit tiles that construct the DX crystals.
Collapse
Affiliation(s)
- Hyeonjun Kwon
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jihoon Shin
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Siqi Sun
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Rong Zhu
- Department of Applied Experimental Biophysics, Institute of Biophysics, Johannes Kepler University Linz, Gruberstrasse 40, A-4020 Linz, Austria
| | - Sarah Stainer
- Department of Applied Experimental Biophysics, Institute of Biophysics, Johannes Kepler University Linz, Gruberstrasse 40, A-4020 Linz, Austria
| | - Peter Hinterdorfer
- Department of Applied Experimental Biophysics, Institute of Biophysics, Johannes Kepler University Linz, Gruberstrasse 40, A-4020 Linz, Austria
| | - Sang-Joon Cho
- Park Systems, Corp., KANC 15F, Gwanggyo-ro 109, Suwon 16229, Republic of Korea
| | - Dong-Hwan Kim
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Yoo Jin Oh
- Department of Applied Experimental Biophysics, Institute of Biophysics, Johannes Kepler University Linz, Gruberstrasse 40, A-4020 Linz, Austria
| |
Collapse
|
8
|
Strutt R, Jusková P, Berlanda SF, Krämer SD, Dittrich PS. Engineering a Biohybrid System to Link Antibiotic Efficacy to Membrane Depth in Bacterial Infections. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2412399. [PMID: 40143780 DOI: 10.1002/smll.202412399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Indexed: 03/28/2025]
Abstract
Treating bacterial infections is dependent upon their site within a biological system, where the cumulative role of membrane transport is challenging to resolve. In this work, a cultivation method based on droplet interface bilayers (DIBs) is established. The architecture of infections in both cellular and tissue contexts is crafted where individual droplets serve as artificial cells infected by intracellular bacteria, or as interconnected units in a tissue-like structure. Through spatio-temporal control over droplets, addition, withdrawal, and sequential antibiotic gradients are tailored acting upon living bacteria. With droplet networks mimicking tissues, it is showed that the treatment response is dependent on the number of the cell-like barriers, corresponding to the number of membranes from an antibiotic source, here described as the membrane depth. Through mathematical modelling a correlation is revealed between the membrane depth of each bacterial population, the antibiotic distribution and thus the treatment efficacy. Ultimately, this approach holds promise as an in vitro bioassay for understanding the response of intracellular bacteria to antibiotics, developing new antibiotics, designing biologically inspired materials, and underpinning emerging bioprinting approaches.
Collapse
Affiliation(s)
- Robert Strutt
- Department of Biosystems Science and Engineering, ETH Zürich, Schanzenstrasse 44, Basel, 4056, Switzerland
| | - Petra Jusková
- Department of Biosystems Science and Engineering, ETH Zürich, Schanzenstrasse 44, Basel, 4056, Switzerland
| | - Simon F Berlanda
- Department of Biosystems Science and Engineering, ETH Zürich, Schanzenstrasse 44, Basel, 4056, Switzerland
| | - Stefanie D Krämer
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zürich, Vladimir-Prelog-Weg 4, Zürich, 8093, Switzerland
| | - Petra S Dittrich
- Department of Biosystems Science and Engineering, ETH Zürich, Schanzenstrasse 44, Basel, 4056, Switzerland
| |
Collapse
|
9
|
Yin C, Yu X, Wu B, Tian L. Spontaneous Emergence of Lipid Vesicles in a Coacervate-Based Compartmentalized System. Angew Chem Int Ed Engl 2025; 64:e202414372. [PMID: 39656166 DOI: 10.1002/anie.202414372] [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: 07/29/2024] [Revised: 12/06/2024] [Indexed: 12/22/2024]
Abstract
The spontaneous emergence of lipid vesicles in the absence of evolved biological machinery represents a major challenge for bottom-up synthetic biology. We show that coacervate microdroplets could create a compartmentalized environment that enriches lipid molecules and facilitates their spontaneous assembly into lipid vesicles. These vesicles can escape from the coacervate microdroplets in a continuous process under non-equilibrium conditions, resembling a constant production process akin to a "primitive enzyme" factory assembly line. These findings significantly extend our understanding of the intricate interaction between lipid molecules and coacervate microdroplets, shedding light on the emergence of cellular systems and offering a new perspective on the conditions necessary for the development of life on Earth.
Collapse
Affiliation(s)
- Chengying Yin
- Key Laboratory of Biomedical Engineering of Ministry of Education, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
- Innovation Center for Smart Medical Technologies & Devices, Binjiang Institute of Zhejiang University, Hangzhou, 310053, China
- Department of Ambulatory Surgery, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, 310027, China
| | - Xinran Yu
- Key Laboratory of Biomedical Engineering of Ministry of Education, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Baohu Wu
- MLZ, JCNS, JCNS-4, Forschungszentrum Jülich, Lichtenbergstr. 1, 85748, Garching, Germany
| | - Liangfei Tian
- Key Laboratory of Biomedical Engineering of Ministry of Education, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
- Innovation Center for Smart Medical Technologies & Devices, Binjiang Institute of Zhejiang University, Hangzhou, 310053, China
- Department of Ambulatory Surgery, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, 310027, China
| |
Collapse
|
10
|
Huang Y, Xu Z, Celik U, Carnahan CF, Faller R, Parikh AN, Liu GY. A New Means to Generate Liposomes by Rehydrating Engineered Lipid Nanoconstructs. MICROMACHINES 2025; 16:138. [PMID: 40047589 PMCID: PMC11857817 DOI: 10.3390/mi16020138] [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: 10/05/2024] [Revised: 01/09/2025] [Accepted: 01/21/2025] [Indexed: 03/09/2025]
Abstract
The concept and feasibility of producing liposomes by rehydrating engineered lipid nanoconstructs are demonstrated in this study. Nanoconstructs of 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC) were produced using a microfluidic delivery probe integrated with an atomic force microscope. The subsequent rehydration of these POPC constructs led to the formation of liposomes, most of which remained adhered to the surface. The size (e.g., diameter) of the liposomes could be tuned by varying the lateral dimension of the lipid constructs. Hierarchical liposomal structures, such as pentagons containing five liposomes at the corners, could also be designed and produced by depositing lipid constructs to designated locations on the surfaces, followed by rehydration. This new means allows for regulating liposomal sizes, distributions, and compositions. The outcomes benefit applications of liposomes as delivery vehicles, sensors, and building blocks in biomaterials design. The ability to produce hierarchical liposomal structures benefits numerous applications such as proto-cell development, multiplexed bio-composite materials, and the engineering of local bio-environments.
Collapse
Affiliation(s)
- Yuqi Huang
- Department of Chemistry, University of California, Davis, CA 95616, USA; (Y.H.); (Z.X.)
| | - Ziqian Xu
- Department of Chemistry, University of California, Davis, CA 95616, USA; (Y.H.); (Z.X.)
| | - Umit Celik
- Department of Chemistry, University of California, Davis, CA 95616, USA; (Y.H.); (Z.X.)
| | - Christopher F. Carnahan
- Biophysics Graduate Group, University of California, Davis, CA 95616, USA; (C.F.C.); (A.N.P.)
| | - Roland Faller
- Department of Chemical Engineering, University of California, Davis, CA 95616, USA
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409, USA;
| | - Atul N. Parikh
- Biophysics Graduate Group, University of California, Davis, CA 95616, USA; (C.F.C.); (A.N.P.)
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA
| | - Gang-yu Liu
- Department of Chemistry, University of California, Davis, CA 95616, USA; (Y.H.); (Z.X.)
- Biophysics Graduate Group, University of California, Davis, CA 95616, USA; (C.F.C.); (A.N.P.)
| |
Collapse
|
11
|
Hindley JW. Constructing mechanosensitive signalling pathways de novo in synthetic cells. Biochem Soc Trans 2025:BST20231285. [PMID: 39838922 DOI: 10.1042/bst20231285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 11/11/2024] [Accepted: 11/27/2024] [Indexed: 01/23/2025]
Abstract
Biological mechanotransduction enables cells to sense and respond to mechanical forces in their local environment through changes in cell structure and gene expression, resulting in downstream changes in cell function. However, the complexity of living systems obfuscates the mechanisms of mechanotransduction, and hence the study of these processes in vitro has been critical in characterising the function of existing mechanosensitive membrane proteins. Synthetic cells are biomolecular compartments that aim to mimic the organisation, functionality and behaviours of biological systems, and represent the next step in the development of in vitro cell models. In recent years, mechanosensitive channels have been incorporated into synthetic cells to create de novo mechanosensitive signalling pathways. Here, I will discuss these developments, from the molecular parts used to construct existing pathways, the functionality of such systems, and potential future directions in engineering synthetic mechanotransduction. The recapitulation of mechanotransduction in synthetic biology will facilitate an improved understanding of biological signalling through the study of molecular interactions across length scales, whilst simultaneously generating new biotechnologies that can be applied as diagnostics, microreactors and therapeutics.
Collapse
Affiliation(s)
- James W Hindley
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, 80 Wood Lane, London W12 0BZ, U.K
- Institute of Chemical Biology, Molecular Sciences Research Hub, Imperial College London, 80 Wood Lane, London W12 0BZ, U.K
- fabriCELL, Molecular Sciences Research Hub, Imperial College London, 80 Wood Lane, London W12 0BZ, U.K
| |
Collapse
|
12
|
Tan CWD, Schöller M, Ehmoser EK. Rapid Multi-Well Evaluation of Assorted Materials for Hydrogel-Assisted Giant Unilamellar Vesicle Production: Empowering Bottom-Up Synthetic Biology. Gels 2025; 11:29. [PMID: 39852000 PMCID: PMC11765364 DOI: 10.3390/gels11010029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 12/28/2024] [Accepted: 12/30/2024] [Indexed: 01/26/2025] Open
Abstract
Giant unilamellar vesicles (GUVs) are versatile cell models in biomedical and environmental research. Of the various GUV production methods, hydrogel-assisted GUV production is most easily implemented in a typical biological laboratory. To date, agarose, polyvinyl alcohol, cross-linked dextran-PEG, polyacrylamide, and starch hydrogels have been used to produce GUVs. Some leach and contaminate the GUVs, while others require handling toxic material or specialised chemistry, thus limiting their use by novices. Alternative hydrogel materials could address these issues or even offer novel advantages. To facilitate discovery, we replaced the manual spreading of reagents with controlled drop-casting in glass Petri dishes and polystyrene multi-well plates, allowing us to rapidly screen up to 96 GUV-production formulations simultaneously. Exploiting this, we rapidly evaluated assorted biomedical hydrogels, including PEG-DA, cross-linked hyaluronic acid, Matrigel, and cross-linked DNA. All of these alternatives successfully produced GUVs. In the process, we also developed a treatment for recycling agarose and polyvinyl alcohol hydrogels for GUV production, and successfully encapsulated porcine liver esterase (PLE-GUVs). PLE-GUVs offer a novel method of GUV labelling and tracing, which emulates the calcein-AM staining behaviour of cells. Our results highlight the utility of our protocol for potentiating substrate material discovery, as well as protocol and product development.
Collapse
Affiliation(s)
- Cherng-Wen Darren Tan
- Institute of Synthetic Bioarchitectures, Department of Bionanosciences, University of Natural Resources and Life Sciences, Vienna, Muthgasse 11, Level 2, 1190 Vienna, Austria; (M.S.); (E.-K.E.)
| | | | | |
Collapse
|
13
|
Heuberger L, Korpidou M, Guinart A, Doellerer D, López DM, Schoenenberger C, Milinkovic D, Lörtscher E, Feringa BL, Palivan CG. Photoreceptor-Like Signal Transduction Between Polymer-Based Protocells. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2413981. [PMID: 39491508 PMCID: PMC11756044 DOI: 10.1002/adma.202413981] [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: 09/16/2024] [Revised: 10/14/2024] [Indexed: 11/05/2024]
Abstract
Deciphering inter- and intracellular signaling pathways is pivotal for understanding the intricate communication networks that orchestrate life's dynamics. Communication models involving bottom-up construction of protocells are emerging but often lack specialized compartments sufficiently robust and hierarchically organized to perform spatiotemporally defined signaling. Here, the modular construction of communicating polymer-based protocells designed to mimic the transduction of information in retinal photoreceptors is presented. Microfluidics is used to generate polymeric protocells subcompartmentalized by specialized artificial organelles. In one protocell population, light triggers artificial organelles with membrane-embedded photoresponsive rotary molecular motors to set off a sequence of reactions starting with the release of encapsulated signaling molecules into the lumen. Intercellular communication is mediated by signal transfer across membranes to protocells containing catalytic artificial organelles as subcompartments, whose signal conversion can be modulated by environmental calcium. Signal propagation also requires selective permeability of the diverse compartments. By segregating artificial organelles in distinct protocells, a sequential chain of reactions mediating intercellular communication is created that is further modulated by adding extracellular messengers. This connective behavior offers the potential for a deeper understanding of signaling pathways and faster integration of proto- and living cells, with the unique advantage of controlling each step by bio-relevant signals.
Collapse
Affiliation(s)
- Lukas Heuberger
- Department of ChemistryUniversity of BaselBasel4002Switzerland
| | - Maria Korpidou
- Department of ChemistryUniversity of BaselBasel4002Switzerland
| | - Ainoa Guinart
- Faculty of Science and EngineeringStratingh Institute for ChemistryUniversity of GroningenAG Groningen9747The Netherlands
| | - Daniel Doellerer
- Faculty of Science and EngineeringStratingh Institute for ChemistryUniversity of GroningenAG Groningen9747The Netherlands
| | | | | | | | - Emanuel Lörtscher
- IBM Research Europe–ZürichSäumerstrasse 4Rüschlikon8803Switzerland
- NCCR – Molecular Systems EngineeringMattenstrasse 22Basel4002Switzerland
| | - Ben L. Feringa
- Faculty of Science and EngineeringStratingh Institute for ChemistryUniversity of GroningenAG Groningen9747The Netherlands
| | - Cornelia G. Palivan
- Department of ChemistryUniversity of BaselBasel4002Switzerland
- NCCR – Molecular Systems EngineeringMattenstrasse 22Basel4002Switzerland
- Swiss Nanoscience Institute (SNI)University of BaselKlingelbergstrasse 80Basel4056Switzerland
| |
Collapse
|
14
|
Bakouei M, Kalantarifard A, Sundara Raju I, Avsievich T, Rannaste L, Kreivi M, Elbuken C. Facile and versatile PDMS-glass capillary double emulsion formation device coupled with rapid purification toward microfluidic giant liposome generation. MICROSYSTEMS & NANOENGINEERING 2024; 10:183. [PMID: 39632792 PMCID: PMC11618511 DOI: 10.1038/s41378-024-00815-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 09/11/2024] [Accepted: 09/29/2024] [Indexed: 12/07/2024]
Abstract
The exceptional ability of liposomes to mimic a cellular lipid membrane makes them invaluable tools in biomembrane studies and bottom-up synthetic biology. Microfluidics provides a promising toolkit for creating giant liposomes in a controlled manner. Nevertheless, challenges associated with the microfluidic formation of double emulsions, as precursors to giant liposomes, limit the full exploration of this potential. In this study, we propose a PDMS-glass capillary hybrid device as a facile and versatile tool for the formation of double emulsions which not only eliminates the need for selective surface treatment, a well-known problem with PDMS formation chips, but also provides fabrication simplicity and reusability compared to the glass-capillary formation chips. These advantages make the presented device a versatile tool for forming double emulsions with varying sizes (spanning two orders of magnitude in diameter), shell thickness, number of compartments, and choice of solvents. We achieved robust thin shell double emulsion formation by operating the hybrid chip in double dripping mode without performing hydrophilic/phobic treatment a priori. In addition, as an alternative to the conventional, time-consuming density-based separation method, a tandem separation chip is developed to deliver double emulsions free of any oil droplet contamination in a continuous and rapid manner without any need for operator handling. The applicability of the device was demonstrated by forming giant liposomes using the solvent extraction method. This easy-to-replicate, flexible, and reliable microfluidic platform for the formation and separation of double emulsion templates paves the way for the high-throughput microfluidic generation of giant liposomes and synthetic cells, opening exciting avenues for biomimetic research. The presented giant liposome assembly line features a novel treatment-free hybrid chip for double emulsion formation coupled with a high throughput separation chip for sample purification.
Collapse
Affiliation(s)
- Mostafa Bakouei
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Ali Kalantarifard
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Indraja Sundara Raju
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Tatiana Avsievich
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Lauri Rannaste
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
- VTT Technical Research Centre of Finland, Oulu, Finland
| | - Marjut Kreivi
- VTT Technical Research Centre of Finland, Oulu, Finland
| | - Caglar Elbuken
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland.
- VTT Technical Research Centre of Finland, Oulu, Finland.
| |
Collapse
|
15
|
Yahyazadeh Shourabi A, Kieffer R, de Jong D, Tam D, Aubin-Tam ME. Mechanical characterization of freestanding lipid bilayers with temperature-controlled phase. SOFT MATTER 2024; 20:8524-8537. [PMID: 39417217 DOI: 10.1039/d4sm00706a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Coexistence of lipid domains in cell membranes is associated with vital biological processes. Here, we investigate two such membranes: a multi-component membrane composed of DOPC and DPPC lipids with gel and fluid separated domains, and a single component membrane composed of PMPC lipids forming ripples. We characterize their mechanical properties below their melting point, where ordered and disordered regions coexist, and above their melting point, where they are in fluid phase. To conduct these inquiries, we create lipid bilayers in a microfluidic chip interfaced with a heating system and optical tweezers. The chip features a bubble trap and enables high-throughput formation of planar bilayers. Optical tweezers experiments reveal interfacial hydrodynamics (fluid-slip) and elastic properties (membrane tension and bending rigidity) at various temperatures. For PMPC bilayers, we demonstrate a higher fluid slip at the interface in the fluid-phase compared to the ripple phase, while for the DOPC:DPPC mixture, similar fluid slip is measured below and above the transition point. Membrane tension for both compositions increases after thermal fluidization. Bending rigidity is also measured using the forces required to extend a lipid nanotube pushed out of the freestanding membranes. This novel temperature-controlled microfluidic platform opens numerous possibilities for thermomechanical studies on freestanding planar membranes.
Collapse
Affiliation(s)
- Arash Yahyazadeh Shourabi
- Department of Bionanoscience, Kavli institute of Nanoscience, Delft University of Technology, Van der Maasweg 9, 2629 HZ, Delft, The Netherlands.
| | - Roland Kieffer
- Department of Bionanoscience, Kavli institute of Nanoscience, Delft University of Technology, Van der Maasweg 9, 2629 HZ, Delft, The Netherlands.
| | - Djanick de Jong
- Department of Bionanoscience, Kavli institute of Nanoscience, Delft University of Technology, Van der Maasweg 9, 2629 HZ, Delft, The Netherlands.
| | - Daniel Tam
- Laboratory for Aero and Hydrodynamics, Faculty of Mechanical Engineering, Delft University of Technology, 2628 CD, Delft, The Netherlands.
| | - Marie-Eve Aubin-Tam
- Department of Bionanoscience, Kavli institute of Nanoscience, Delft University of Technology, Van der Maasweg 9, 2629 HZ, Delft, The Netherlands.
| |
Collapse
|
16
|
Yamada T, Suzuki H. Microfluidics-based stable production of monodisperse giant unilamellar vesicles by oil-phase removal from double emulsion. J Liposome Res 2024:1-7. [PMID: 39470184 DOI: 10.1080/08982104.2024.2420337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 10/17/2024] [Accepted: 10/18/2024] [Indexed: 10/30/2024]
Abstract
Giant liposomes, or giant unilamellar vesicles (GUVs), have been utilized as cell-size bioreactors to replicate the physical and chemical properties of biological cells. However, conventional methods for preparing GUVs typically lack precise control over their size. Several research groups have recently developed microfluidic techniques to create monodisperse GUVs by generating water-in-oil-in-water (W/O/W) droplets with a thin oil layer that subsequently transform into GUVs. However, the formation of a thin oil shell requires the intricate control of the flow rate, which can be both challenging and unstable. In this study, we investigated the design of a two-step flow-focusing microfluidic channel to produce stable W/O/W droplets. These droplets underwent substantial oil layer reduction through spontaneous removal by fluidic shear forces. Consequently, the majority of the oil layer in the W/O/W droplets was reduced, improving uniformity of GUVs.
Collapse
Affiliation(s)
- Tomoki Yamada
- Department of Precision Mechanics, Faculty of Science and Engineering, Chuo University, Tokyo, Japan
| | - Hiroaki Suzuki
- Department of Precision Mechanics, Faculty of Science and Engineering, Chuo University, Tokyo, Japan
| |
Collapse
|
17
|
Van de Cauter L, Jawale YK, Tam D, Baldauf L, van Buren L, Koenderink GH, Dogterom M, Ganzinger KA. High-Speed Imaging of Giant Unilamellar Vesicle Formation in cDICE. ACS OMEGA 2024; 9:42278-42288. [PMID: 39431092 PMCID: PMC11483911 DOI: 10.1021/acsomega.4c04825] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 08/26/2024] [Accepted: 09/17/2024] [Indexed: 10/22/2024]
Abstract
Giant unilamellar vesicles (GUVs) are widely used as in vitro model membranes in biophysics and as cell-sized containers in synthetic biology. Despite their ubiquitous use, there is no one-size-fits-all method for their production. Numerous methods have been developed to meet the demanding requirements of reproducibility, reliability, and high yield while simultaneously achieving robust encapsulation. Emulsion-based methods are often praised for their apparent simplicity and good yields; hence, methods like continuous droplet interface crossing encapsulation (cDICE), which make use of this principle, have gained popularity. However, the underlying physical principles governing the formation of GUVs in cDICE and related methods remain poorly understood. To this end, we have developed a high-speed microscopy setup that allows us to visualize GUV formation in real time. Our experiments reveal a complex droplet formation process occurring at the capillary orifice, generating >30 μm-sized droplets and only in some cases GUV-sized (∼15 μm) satellite droplets. According to existing theoretical models, the oil-water interface should allow for the crossing of all droplets, but based on our observations and scaling arguments on the fluid dynamics within the system, we find a size-selective crossing of GUV-sized droplets only. The origin of these droplets remains partly unclear; we hypothesize that some small GUVs might be formed from large droplets sitting at the second interface. Finally, we demonstrate that proteins in the inner solution affect GUV formation by increasing the viscosity and altering the lipid adsorption kinetics. These results will not only contribute to a better understanding of GUV formation processes in cDICE but ultimately also aid in the development of more reliable and efficient methods for GUV production.
Collapse
Affiliation(s)
| | - Yash K. Jawale
- Department
of Bionanoscience, Kavli Institute of Nanoscience, Delft University of Technology, Delft 2629 HZ, The Netherlands
| | - Daniel Tam
- Laboratory
for Aero and Hydrodynamics, Delft University
of Technology, Delft 2629 HZ, The Netherlands
| | - Lucia Baldauf
- Department
of Bionanoscience, Kavli Institute of Nanoscience, Delft University of Technology, Delft 2629 HZ, The Netherlands
| | - Lennard van Buren
- Department
of Bionanoscience, Kavli Institute of Nanoscience, Delft University of Technology, Delft 2629 HZ, The Netherlands
| | - Gijsje H. Koenderink
- Department
of Bionanoscience, Kavli Institute of Nanoscience, Delft University of Technology, Delft 2629 HZ, The Netherlands
| | - Marileen Dogterom
- Department
of Bionanoscience, Kavli Institute of Nanoscience, Delft University of Technology, Delft 2629 HZ, The Netherlands
| | | |
Collapse
|
18
|
Cheng Y, Hay CD, Mahuttanatan SM, Hindley JW, Ces O, Elani Y. Microfluidic technologies for lipid vesicle generation. LAB ON A CHIP 2024; 24:4679-4716. [PMID: 39323383 PMCID: PMC11425070 DOI: 10.1039/d4lc00380b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 08/12/2024] [Indexed: 09/27/2024]
Abstract
Encapsulating biological and non-biological materials in lipid vesicles presents significant potential in both industrial and academic settings. When smaller than 100 nm, lipid vesicles and lipid nanoparticles are ideal vehicles for drug delivery, facilitating the delivery of payloads, improving pharmacokinetics, and reducing the off-target effects of therapeutics. When larger than 1 μm, vesicles are useful as model membranes for biophysical studies, as synthetic cell chassis, as bio-inspired supramolecular devices, and as the basis of protocells to explore the origin of life. As applications of lipid vesicles gain prominence in the fields of nanomedicine, biotechnology, and synthetic biology, there is a demand for advanced technologies for their controlled construction, with microfluidic methods at the forefront of these developments. Compared to conventional bulk methods, emerging microfluidic methods offer advantages such as precise size control, increased production throughput, high encapsulation efficiency, user-defined membrane properties (i.e., lipid composition, vesicular architecture, compartmentalisation, membrane asymmetry, etc.), and potential integration with lab-on-chip manipulation and analysis modules. We provide a review of microfluidic lipid vesicle generation technologies, focusing on recent advances and state-of-the-art techniques. Principal technologies are described, and key research milestones are highlighted. The advantages and limitations of each approach are evaluated, and challenges and opportunities for microfluidic engineering of lipid vesicles to underpin a new generation of therapeutics, vaccines, sensors, and bio-inspired technologies are presented.
Collapse
Affiliation(s)
- Yu Cheng
- Institute of Chemical Biology, Molecular Sciences Research Hub, Imperial College London, London, UK.
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London, UK
| | - Callum D Hay
- Institute of Chemical Biology, Molecular Sciences Research Hub, Imperial College London, London, UK.
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London, UK
| | - Suchaya M Mahuttanatan
- Institute of Chemical Biology, Molecular Sciences Research Hub, Imperial College London, London, UK.
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London, UK
| | - James W Hindley
- Institute of Chemical Biology, Molecular Sciences Research Hub, Imperial College London, London, UK.
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London, UK
| | - Oscar Ces
- Institute of Chemical Biology, Molecular Sciences Research Hub, Imperial College London, London, UK.
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London, UK
| | - Yuval Elani
- Institute of Chemical Biology, Molecular Sciences Research Hub, Imperial College London, London, UK.
- Department of Chemical Engineering, Imperial College London, London, UK
| |
Collapse
|
19
|
Fleury JB, Baulin VA. Synergistic Effects of Microplastics and Marine Pollutants on the Destabilization of Lipid Bilayers. J Phys Chem B 2024; 128:8753-8761. [PMID: 39219546 PMCID: PMC11403677 DOI: 10.1021/acs.jpcb.4c03290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Microplastics have been detected in diverse environments, including soil, snowcapped mountains, and even within human organs and blood. These findings have sparked extensive research into the health implications of microplastics for living organisms. Recent studies have shown that microplastics can adsorb onto lipid membranes and induce mechanical stress. In controlled laboratory conditions, the behavior and effects of microplastics can differ markedly from those in natural environments. In this study, we investigate how exposure of microplastics to pollutants affects their interactions with lipid bilayers. Our findings reveal that pollutants, such as chemical solvents, significantly enhance the mechanical stretching effects of microplastics. This suggests that microplastics can act as vectors for harmful pollutants, facilitating their penetration through lipid membranes and thus strongly affect their biophysical properties. This research underscores the complex interplay between microplastics and environmental contaminants.
Collapse
Affiliation(s)
- Jean-Baptiste Fleury
- Experimental Physics and Center for Biophysics, Universitat des Saarlandes, 66123 Saarbruecken, Germany
| | - Vladimir A Baulin
- Departament d'Enginyeria Química, Universitat Rovira i Virgili, Av. dels Països Catalans, 26, 43007 Tarragona, Spain
| |
Collapse
|
20
|
Chen C, Ganar KA, de Haas RJ, Jarnot N, Hogeveen E, de Vries R, Deshpande S. Elastin-like polypeptide coacervates as reversibly triggerable compartments for synthetic cells. Commun Chem 2024; 7:198. [PMID: 39232074 PMCID: PMC11374812 DOI: 10.1038/s42004-024-01270-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 08/05/2024] [Indexed: 09/06/2024] Open
Abstract
Compartmentalization is a vital aspect of living cells to orchestrate intracellular processes. In a similar vein, constructing dynamic and responsive sub-compartments is key to synthetic cell engineering. In recent years, liquid-liquid phase separation via coacervation has offered an innovative avenue for creating membraneless organelles (MOs) within artificial cells. Here, we present a lab-on-a-chip system to reversibly trigger peptide-based coacervates within cell-mimicking confinements. We use double emulsion droplets (DEs) as our synthetic cell containers while pH-responsive elastin-like polypeptides (ELPs) act as the coacervate system. We first present a high-throughput microfluidic DE production enabling efficient encapsulation of the ELPs. The DEs are then harvested to perform multiple MO formation-dissolution cycles using pH as well as temperature variation. For controlled long-term visualization and modulation of the external environment, we developed an integrated microfluidic device for trapping and environmental stimulation of DEs, with negligible mechanical force, and demonstrated a proof-of-principle osmolyte-based triggering to induce multiple MO formation-dissolution cycles. In conclusion, our work showcases the use of DEs and ELPs in designing membraneless reversible compartmentalization within synthetic cells via physicochemical triggers. Additionally, presented on-chip platform can be applied over a wide range of phase separation and vesicle systems for applications in synthetic cells and beyond.
Collapse
Affiliation(s)
- Chang Chen
- Laboratory of Physical Chemistry and Soft Matter, Wageningen University and Research, Stippeneng 4, 6708 WE, Wageningen, The Netherlands
| | - Ketan A Ganar
- Laboratory of Physical Chemistry and Soft Matter, Wageningen University and Research, Stippeneng 4, 6708 WE, Wageningen, The Netherlands
| | - Robbert J de Haas
- Laboratory of Physical Chemistry and Soft Matter, Wageningen University and Research, Stippeneng 4, 6708 WE, Wageningen, The Netherlands
| | - Nele Jarnot
- Laboratory of Physical Chemistry and Soft Matter, Wageningen University and Research, Stippeneng 4, 6708 WE, Wageningen, The Netherlands
| | - Erwin Hogeveen
- Laboratory of Physical Chemistry and Soft Matter, Wageningen University and Research, Stippeneng 4, 6708 WE, Wageningen, The Netherlands
| | - Renko de Vries
- Laboratory of Physical Chemistry and Soft Matter, Wageningen University and Research, Stippeneng 4, 6708 WE, Wageningen, The Netherlands
| | - Siddharth Deshpande
- Laboratory of Physical Chemistry and Soft Matter, Wageningen University and Research, Stippeneng 4, 6708 WE, Wageningen, The Netherlands.
| |
Collapse
|
21
|
Cochereau R, Voisin H, Solé-Jamault V, Novales B, Davy J, Jamme F, Renard D, Boire A. Influence of pH and lipid membrane on the liquid-liquid phase separation of wheat γ-gliadin in aqueous conditions. J Colloid Interface Sci 2024; 668:252-263. [PMID: 38678881 DOI: 10.1016/j.jcis.2024.04.136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 03/25/2024] [Accepted: 04/19/2024] [Indexed: 05/01/2024]
Abstract
Protein body (PB) formation in wheat seeds is a critical process influencing seed content and nutritional quality. In this study, we investigate the potential mechanisms governing PB formation through an in vitro approach, focusing on γ-gliadin, a key wheat storage protein. We used a microfluidic technique to encapsulate γ-gliadin within giant unilamellar vesicles (GUVs) and tune the physicochemical conditions in a controlled and rapid way. We examined the influence of pH and protein concentration on LLPS and protein-membrane interactions using various microscopy and spectroscopy techniques. We showed that γ-gliadin encapsulated in GUVs can undergo a pH-triggered liquid-liquid phase separation (LLPS) by two distinct mechanisms depending on the γ-gliadin concentration. At low protein concentrations, γ-gliadins phase separate by a nucleation and growth-like process, while, at higher protein concentration and pH above 6.0, γ-gliadin formed a bi-continuous phase suggesting a spinodal decomposition-like mechanism. Fluorescence and microscopy data suggested that γ-gliadin dense phase exhibited affinity for the GUV membrane, forming a layer at the interface and affecting the reversibility of the phase separation.
Collapse
Affiliation(s)
| | | | | | - Bruno Novales
- INRAE, UR 1268 BIA, F-44300 Nantes, France; INRAE, PROBE/CALIS Research Infrastructures, BIBS Facility, F-44300 Nantes, France
| | | | - Frédéric Jamme
- DISCO Beamline, SOLEIL Synchrotron, 91192 Gif-sur-Yvette, France
| | | | | |
Collapse
|
22
|
Ngocho K, Yang X, Wang Z, Hu C, Yang X, Shi H, Wang K, Liu J. Synthetic Cells from Droplet-Based Microfluidics for Biosensing and Biomedical Applications. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2400086. [PMID: 38563581 DOI: 10.1002/smll.202400086] [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: 01/04/2024] [Revised: 03/13/2024] [Indexed: 04/04/2024]
Abstract
Synthetic cells function as biological mimics of natural cells by mimicking salient features of cells such as metabolism, response to stimuli, gene expression, direct metabolism, and high stability. Droplet-based microfluidic technology presents the opportunity for encapsulating biological functional components in uni-lamellar liposome or polymer droplets. Verified by its success in the fabrication of synthetic cells, microfluidic technology is widely replacing conventional labor-intensive, expensive, and sophisticated techniques justified by its ability to miniaturize and perform batch production operations. In this review, an overview of recent research on the preparation of synthetic cells through droplet-based microfluidics is provided. Different synthetic cells including lipid vesicles (liposome), polymer vesicles (polymersome), coacervate microdroplets, and colloidosomes, are systematically discussed. Efforts are then made to discuss the design of a variety of microfluidic chips for synthetic cell preparation since the combination of microfluidics with bottom-up synthetic biology allows for reproductive and tunable construction of batches of synthetic cell models from simple structures to higher hierarchical structures. The recent advances aimed at exploiting them in biosensors and other biomedical applications are then discussed. Finally, some perspectives on the challenges and future developments of synthetic cell research with microfluidics for biomimetic science and biomedical applications are provided.
Collapse
Affiliation(s)
- Kleins Ngocho
- State key laboratory of Chemo/Biosensing and Chemometrics College of Chemistry and Chemical Engineering Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha, 410082, P. R. China
| | - Xilei Yang
- State key laboratory of Chemo/Biosensing and Chemometrics College of Chemistry and Chemical Engineering Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha, 410082, P. R. China
| | - Zefeng Wang
- State key laboratory of Chemo/Biosensing and Chemometrics College of Chemistry and Chemical Engineering Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha, 410082, P. R. China
| | - Cunjie Hu
- State key laboratory of Chemo/Biosensing and Chemometrics College of Chemistry and Chemical Engineering Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha, 410082, P. R. China
| | - Xiaohai Yang
- State key laboratory of Chemo/Biosensing and Chemometrics College of Chemistry and Chemical Engineering Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha, 410082, P. R. China
| | - Hui Shi
- State key laboratory of Chemo/Biosensing and Chemometrics College of Chemistry and Chemical Engineering Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha, 410082, P. R. China
| | - Kemin Wang
- State key laboratory of Chemo/Biosensing and Chemometrics College of Chemistry and Chemical Engineering Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha, 410082, P. R. China
| | - Jianbo Liu
- State key laboratory of Chemo/Biosensing and Chemometrics College of Chemistry and Chemical Engineering Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha, 410082, P. R. China
| |
Collapse
|
23
|
Vladisaljević GT. Droplet Microfluidics for High-Throughput Screening and Directed Evolution of Biomolecules. MICROMACHINES 2024; 15:971. [PMID: 39203623 PMCID: PMC11356158 DOI: 10.3390/mi15080971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 07/23/2024] [Accepted: 07/26/2024] [Indexed: 09/03/2024]
Abstract
Directed evolution is a powerful technique for creating biomolecules such as proteins and nucleic acids with tailor-made properties for therapeutic and industrial applications by mimicking the natural evolution processes in the laboratory. Droplet microfluidics improved classical directed evolution by enabling time-consuming and laborious steps in this iterative process to be performed within monodispersed droplets in a highly controlled and automated manner. Droplet microfluidic chips can generate, manipulate, and sort individual droplets at kilohertz rates in a user-defined microchannel geometry, allowing new strategies for high-throughput screening and evolution of biomolecules. In this review, we discuss directed evolution studies in which droplet-based microfluidic systems were used to screen and improve the functional properties of biomolecules. We provide a systematic overview of basic on-chip fluidic operations, including reagent mixing by merging continuous fluid streams and droplet pairs, reagent addition by picoinjection, droplet generation, droplet incubation in delay lines, chambers and hydrodynamic traps, and droplet sorting techniques. Various microfluidic strategies for directed evolution using single and multiple emulsions and biomimetic materials (giant lipid vesicles, microgels, and microcapsules) are highlighted. Completely cell-free microfluidic-assisted in vitro compartmentalization methods that eliminate the need to clone DNA into cells after each round of mutagenesis are also presented.
Collapse
Affiliation(s)
- Goran T Vladisaljević
- Department of Chemical Engineering, Loughborough University, Loughborough LE11 3TU, UK
| |
Collapse
|
24
|
Selivanovitch E, Ostwalt A, Chao Z, Daniel S. Emerging Designs and Applications for Biomembrane Biosensors. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2024; 17:339-366. [PMID: 39018354 PMCID: PMC11913122 DOI: 10.1146/annurev-anchem-061622-042618] [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] [Indexed: 07/19/2024]
Abstract
Nature has inspired the development of biomimetic membrane sensors in which the functionalities of biological molecules, such as proteins and lipids, are harnessed for sensing applications. This review provides an overview of the recent developments for biomembrane sensors compatible with either bulk or planar sensing applications, namely using lipid vesicles or supported lipid bilayers, respectively. We first describe the individual components required for these sensing platforms and the design principles that are considered when constructing them, and we segue into recent applications being implemented across multiple fields. Our goal for this review is to illustrate the versatility of nature's biomembrane toolbox and simultaneously highlight how biosensor platforms can be enhanced by harnessing it.
Collapse
Affiliation(s)
- Ekaterina Selivanovitch
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York, USA;
| | - Alexis Ostwalt
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York, USA;
| | - Zhongmou Chao
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York, USA;
| | - Susan Daniel
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York, USA;
| |
Collapse
|
25
|
Ernits M, Reinsalu O, Yandrapalli N, Kopanchuk S, Moradpur-Tari E, Sanka I, Scheler O, Rinken A, Kurg R, Kyritsakis A, Linko V, Zadin V. Microfluidic production, stability and loading of synthetic giant unilamellar vesicles. Sci Rep 2024; 14:14071. [PMID: 38890456 PMCID: PMC11189546 DOI: 10.1038/s41598-024-64613-4] [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: 06/02/2023] [Accepted: 06/11/2024] [Indexed: 06/20/2024] Open
Abstract
In advanced drug delivery, versatile liposomal formulations are commonly employed for safer and more accurate therapies. Here we report a method that allows a straightforward production of synthetic monodisperse (~ 100 μm) giant unilamellar vesicles (GUVs) using a microfluidic system. The stability analysis based on the microscopy imaging showed that at ambient conditions the produced GUVs had a half-life of 61 ± 2 h. However, it was observed that ~ 90% of the calcein dye that was loaded into GUVs was transported into a surrounding medium in 24 h, thus indicating that the GUVs may release these small dye molecules without distinguishable membrane disruption. We further demonstrated the feasibility of our method by loading GUVs with larger and very different cargo objects; small soluble fluorescent proteins and larger magnetic microparticles in a suspension. Compared to previously reported microfluidics-based production techniques, the obtained results indicate that our simplified method could be equally harnessed in creating GUVs with less cost, effort and time, which could further benefit studying closed membrane systems.
Collapse
Affiliation(s)
- Mart Ernits
- Institute of Technology, University of Tartu, Nooruse 1, 50411, Tartu, Estonia
| | - Olavi Reinsalu
- Institute of Technology, University of Tartu, Nooruse 1, 50411, Tartu, Estonia
| | - Naresh Yandrapalli
- Department of Colloid Chemistry, Max Planck Institute of Colloids and Interfaces, Am Mühlenberg 1, 14476, Potsdam, Germany
| | - Sergei Kopanchuk
- Institute of Chemistry, University of Tartu, Ravila 14a, 50411, Tartu, Estonia
| | - Ehsan Moradpur-Tari
- Institute of Technology, University of Tartu, Nooruse 1, 50411, Tartu, Estonia
| | - Immanuel Sanka
- Department of Chemistry and Biotechnology, Tallinn University of Technology (TalTech), Akadeemia Tee 15, 12618, Tallinn, Estonia
| | - Ott Scheler
- Department of Chemistry and Biotechnology, Tallinn University of Technology (TalTech), Akadeemia Tee 15, 12618, Tallinn, Estonia
| | - Ago Rinken
- Institute of Chemistry, University of Tartu, Ravila 14a, 50411, Tartu, Estonia
| | - Reet Kurg
- Institute of Technology, University of Tartu, Nooruse 1, 50411, Tartu, Estonia
| | - Andreas Kyritsakis
- Institute of Technology, University of Tartu, Nooruse 1, 50411, Tartu, Estonia.
| | - Veikko Linko
- Institute of Technology, University of Tartu, Nooruse 1, 50411, Tartu, Estonia.
- Biohybrid Materials, Department of Bioproducts and Biosystems, Aalto University School of Chemical Engineering, Kemistintie 1, 02150, Espoo, Finland.
| | - Veronika Zadin
- Institute of Technology, University of Tartu, Nooruse 1, 50411, Tartu, Estonia.
| |
Collapse
|
26
|
Ben Trad F, Delacotte J, Lemaître F, Guille-Collignon M, Arbault S, Sojic N, Labbé E, Buriez O. Shadow electrochemiluminescence imaging of giant liposomes opening at polarized electrodes. Analyst 2024; 149:3317-3324. [PMID: 38742381 DOI: 10.1039/d4an00470a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
In this work, the release of giant liposome (∼100 μm in diameter) content was imaged by shadow electrochemiluminescence (ECL) microscopy. Giant unilamellar liposomes were pre-loaded with a sucrose solution and allowed to sediment at an ITO electrode surface immersed in a solution containing a luminophore ([Ru(bpy)3]2+) and a sacrificial co-reactant (tri-n-propylamine). Upon polarization, the electrode exhibited illumination over its entire surface thanks to the oxidation of ECL reagents. However, as soon as liposomes reached the electrode surface, dark spots appeared and then spread over time on the surface. This observation reflected a blockage of the electrode surface at the contact point between the liposome and the electrode surface, followed by the dilution of ECL reagents after the rupture of the liposome membrane and release of its internal ECL-inactive solution. Interestingly, ECL reappeared in areas where it initially faded, indicating back-diffusion of ECL reagents towards the previously diluted area and thus confirming liposome permeabilization. The whole process was analyzed qualitatively and quantitatively within the defined region of interest. Two mass transport regimes were identified: a gravity-driven spreading process when the liposome releases its content leading to ECL vanishing and a diffusive regime when ECL recovers. The reported shadow ECL microscopy should find promising applications for the imaging of transient events such as molecular species released by artificial or biological vesicles.
Collapse
Affiliation(s)
- Fatma Ben Trad
- PASTEUR, Département de Chimie, Ecole Normale Supérieure, PSL University, Sorbonne Université, CNRS, 75005 Paris, France.
| | - Jérôme Delacotte
- PASTEUR, Département de Chimie, Ecole Normale Supérieure, PSL University, Sorbonne Université, CNRS, 75005 Paris, France.
| | - Frédéric Lemaître
- PASTEUR, Département de Chimie, Ecole Normale Supérieure, PSL University, Sorbonne Université, CNRS, 75005 Paris, France.
| | - Manon Guille-Collignon
- PASTEUR, Département de Chimie, Ecole Normale Supérieure, PSL University, Sorbonne Université, CNRS, 75005 Paris, France.
| | - Stéphane Arbault
- Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, F-33600 Pessac, France
| | - Neso Sojic
- Univ. Bordeaux, CNRS, Bordeaux INP, ISM, UMR 5255 CNRS, 33400 Talence, France.
| | - Eric Labbé
- PASTEUR, Département de Chimie, Ecole Normale Supérieure, PSL University, Sorbonne Université, CNRS, 75005 Paris, France.
| | - Olivier Buriez
- PASTEUR, Département de Chimie, Ecole Normale Supérieure, PSL University, Sorbonne Université, CNRS, 75005 Paris, France.
| |
Collapse
|
27
|
Zizzari A, Arima V. Glass Microdroplet Generator for Lipid-Based Double Emulsion Production. MICROMACHINES 2024; 15:500. [PMID: 38675311 PMCID: PMC11052113 DOI: 10.3390/mi15040500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 03/27/2024] [Accepted: 04/01/2024] [Indexed: 04/28/2024]
Abstract
Microfluidics offers a highly controlled and reproducible route to synthesize lipid vesicles. In recent years, several microfluidic approaches have been introduced for this purpose, but double emulsions, such as Water-in-Oil-in-Water (W/O/W) droplets, are preferable to produce giant vesicles that are able to maximize material encapsulation. Flow focusing (FF) is a technique used to generate double emulsion droplets with high monodispersity, a controllable size, and good robustness. Many researchers use polydimethylsiloxane as a substrate material to fabricate microdroplet generators, but it has some limitations due to its hydrophobicity, incompatibility with organic solvents, and the molecular adsorption on the microchannel walls. Thus, specific surface modification and functionalization steps, which are uncomfortable to perform in closed microchannels, are required to overcome these shortcomings. Here, we propose glass as a material to produce a chip with a six-inlet junction geometry. The peculiar geometry and the glass physicochemical properties allow for W/O/W droplet formation without introducing microchannel wall functionalization and using a variety of reagents and organic solvents. The robust glass chip can be easily cleaned and used repeatedly, bringing advantages in terms of cost and reproducibility in emulsion preparation.
Collapse
Affiliation(s)
- Alessandra Zizzari
- CNR NANOTEC-Institute of Nanotechnology c/o Campus Ecotekne, via Monteroni, 73100 Lecce, Italy;
| | | |
Collapse
|
28
|
Zhang M, Qiu W, Nie R, Xia Q, Zhang D, Pan X. Macronutrient and PFOS bioavailability manipulated by aeration-driven rhizospheric organic nanocapsular assembly. WATER RESEARCH 2024; 253:121334. [PMID: 38382293 DOI: 10.1016/j.watres.2024.121334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 01/31/2024] [Accepted: 02/17/2024] [Indexed: 02/23/2024]
Abstract
Ubiquitous presence of the extremely persistent pollutants, per- and polyfluoroalkyl substances, is drawing ever-increasing concerns for their high eco-environmental risks which, however, are insufficiently considered based on the assembly characteristics of those amphiphilic molecules in environment. This study investigated the re-organization and self-assembly of perfluorooctane sulfonate (PFOS) and macronutrient molecules from rhizospheric organic (RhO) matter induced with a common operation of aeration. Atomic force microscopy (AFM) with infrared spectroscopy (IR)-mapping clearly showed that, after aeration and stabilization, RhO nanocapsules (∼ 1000 nm or smaller) with a core of PFOS-protein complexes coated by "lipid-carbohydrate" layers were observed whereas the capsule structure with a lipid core surrounded by "protein-carbohydrate-protein" multilayers was obtained in the absence of PFOS. It is aeration that exerted the disassociation of pristine RhO components, after which the environmental concentration PFOS restructured the self-assembly structure in a conspicuous "disorder-to-order" transition. AFM IR-mapping analysis of faeces combined with quantification of component uptake denoted the decreased ingestion and utilization of both PFOS and proteins compared with lipids and carbohydrates when Daphnia magna were fed with RhO nanocapsules. RhO nanocapsules acted as double-edged swords via simultaneously impeding the bioaccessibility of hazardous PFOS molecules and macronutrient proteins; and the latter might be more significant, which caused a malnutrition status within merely 48 h. Elucidating the assembly structure of natural organic matter and environmental concentration PFOS, the finding of this work could be a crucial supplementation to the high-dose-dependent eco-effect investigations on PFOS.
Collapse
Affiliation(s)
- Ming Zhang
- Key Laboratory of Microbial Technology for Industrial Pollution Control of Zhejiang Province, College of Environment, Zhejiang University of Technology, Hangzhou 310014, China
| | - Weifeng Qiu
- Key Laboratory of Microbial Technology for Industrial Pollution Control of Zhejiang Province, College of Environment, Zhejiang University of Technology, Hangzhou 310014, China
| | - Rui Nie
- Key Laboratory of Microbial Technology for Industrial Pollution Control of Zhejiang Province, College of Environment, Zhejiang University of Technology, Hangzhou 310014, China
| | - Qiaoyun Xia
- Key Laboratory of Microbial Technology for Industrial Pollution Control of Zhejiang Province, College of Environment, Zhejiang University of Technology, Hangzhou 310014, China
| | - Daoyong Zhang
- Key Laboratory of Microbial Technology for Industrial Pollution Control of Zhejiang Province, College of Environment, Zhejiang University of Technology, Hangzhou 310014, China.
| | - Xiangliang Pan
- Key Laboratory of Microbial Technology for Industrial Pollution Control of Zhejiang Province, College of Environment, Zhejiang University of Technology, Hangzhou 310014, China.
| |
Collapse
|
29
|
Fasciano S, Wang S. Recent advances of droplet-based microfluidics for engineering artificial cells. SLAS Technol 2024; 29:100090. [PMID: 37245659 DOI: 10.1016/j.slast.2023.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 05/09/2023] [Accepted: 05/24/2023] [Indexed: 05/30/2023]
Abstract
Artificial cells, synthetic cells, or minimal cells are microengineered cell-like structures that mimic the biological functions of cells. Artificial cells are typically biological or polymeric membranes where biologically active components, including proteins, genes, and enzymes, are encapsulated. The goal of engineering artificial cells is to build a living cell with the least amount of parts and complexity. Artificial cells hold great potential for several applications, including membrane protein interactions, gene expression, biomaterials, and drug development. It is critical to generate robust, stable artificial cells using high throughput, easy-to-control, and flexible techniques. Recently, droplet-based microfluidic techniques have shown great potential for the synthesis of vesicles and artificial cells. Here, we summarized the recent advances in droplet-based microfluidic techniques for the fabrication of vesicles and artificial cells. We first reviewed the different types of droplet-based microfluidic devices, including flow-focusing, T-junction, and coflowing. Next, we discussed the formation of multi-compartmental vesicles and artificial cells based on droplet-based microfluidics. The applications of artificial cells for studying gene expression dynamics, artificial cell-cell communications, and mechanobiology are highlighted and discussed. Finally, the current challenges and future outlook of droplet-based microfluidic methods for engineering artificial cells are discussed. This review will provide insights into scientific research in synthetic biology, microfluidic devices, membrane interactions, and mechanobiology.
Collapse
Affiliation(s)
- Samantha Fasciano
- Department of Cellular and Molecular Biology, University of New Haven, West Haven, CT, USA
| | - Shue Wang
- Department of Chemistry, Chemical and Biomedical Engineering, University of New Haven, West Haven, CT, USA.
| |
Collapse
|
30
|
Waeterschoot J, Gosselé W, Lemež Š, Casadevall I Solvas X. Artificial cells for in vivo biomedical applications through red blood cell biomimicry. Nat Commun 2024; 15:2504. [PMID: 38509073 PMCID: PMC10954685 DOI: 10.1038/s41467-024-46732-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 03/08/2024] [Indexed: 03/22/2024] Open
Abstract
Recent research in artificial cell production holds promise for the development of delivery agents with therapeutic effects akin to real cells. To succeed in these applications, these systems need to survive the circulatory conditions. In this review we present strategies that, inspired by the endurance of red blood cells, have enhanced the viability of large, cell-like vehicles for in vivo therapeutic use, particularly focusing on giant unilamellar vesicles. Insights from red blood cells can guide modifications that could transform these platforms into advanced drug delivery vehicles, showcasing biomimicry's potential in shaping the future of therapeutic applications.
Collapse
Affiliation(s)
- Jorik Waeterschoot
- Department of Biosystems - MeBioS, KU Leuven, Willem de Croylaan 42, 3001, Leuven, Belgium.
| | - Willemien Gosselé
- Department of Biosystems - MeBioS, KU Leuven, Willem de Croylaan 42, 3001, Leuven, Belgium
| | - Špela Lemež
- Department of Biosystems - MeBioS, KU Leuven, Willem de Croylaan 42, 3001, Leuven, Belgium
| | | |
Collapse
|
31
|
Heuberger L, Messmer D, dos Santos EC, Scherrer D, Lörtscher E, Schoenenberger C, Palivan CG. Microfluidic Giant Polymer Vesicles Equipped with Biopores for High-Throughput Screening of Bacteria. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307103. [PMID: 38158637 PMCID: PMC10953582 DOI: 10.1002/advs.202307103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Indexed: 01/03/2024]
Abstract
Understanding the mechanisms of antibiotic resistance is critical for the development of new therapeutics. Traditional methods for testing bacteria are often limited in their efficiency and reusability. Single bacterial cells can be studied at high throughput using double emulsions, although the lack of control over the oil shell permeability and limited access to the droplet interior present serious drawbacks. Here, a straightforward strategy for studying bacteria-encapsulating double emulsion-templated giant unilamellar vesicles (GUVs) is introduced. This microfluidic approach serves to simultaneously load bacteria inside synthetic GUVs and to permeabilize their membrane with the pore-forming peptide melittin. This enables antibiotic delivery or the influx of fresh medium into the GUV lumen for highly parallel cultivation and antimicrobial efficacy testing. Polymer-based GUVs proved to be efficient culture and analysis microvessels, as microfluidics allow easy selection and encapsulation of bacteria and rapid modification of culture conditions for antibiotic development. Further, a method for in situ profiling of biofilms within GUVs for high-throughput screening is demonstrated. Conceivably, synthetic GUVs equipped with biopores can serve as a foundation for the high-throughput screening of bacterial colony interactions during biofilm formation and for investigating the effect of antibiotics on biofilms.
Collapse
Affiliation(s)
- Lukas Heuberger
- Department of ChemistryUniversity of BaselMattenstrasse 22Basel4002Switzerland
| | - Daniel Messmer
- Department of ChemistryUniversity of BaselMattenstrasse 22Basel4002Switzerland
| | - Elena C. dos Santos
- Department of ChemistryUniversity of BaselMattenstrasse 22Basel4002Switzerland
| | - Dominik Scherrer
- IBM Research Europe–ZürichSäumerstrasse 4Rüschlikon8803Switzerland
| | - Emanuel Lörtscher
- IBM Research Europe–ZürichSäumerstrasse 4Rüschlikon8803Switzerland
- NCCR‐Molecular Systems EngineeringMattenstrasse 24a, BPR 1095Basel4058Switzerland
| | | | - Cornelia G. Palivan
- Department of ChemistryUniversity of BaselMattenstrasse 22Basel4002Switzerland
- NCCR‐Molecular Systems EngineeringMattenstrasse 24a, BPR 1095Basel4058Switzerland
- Swiss Nanoscience Institute (SNI)University of BaselKlingelbergstrasse 82Basel4056Switzerland
| |
Collapse
|
32
|
Peng Z, Iwabuchi S, Izumi K, Takiguchi S, Yamaji M, Fujita S, Suzuki H, Kambara F, Fukasawa G, Cooney A, Di Michele L, Elani Y, Matsuura T, Kawano R. Lipid vesicle-based molecular robots. LAB ON A CHIP 2024; 24:996-1029. [PMID: 38239102 PMCID: PMC10898420 DOI: 10.1039/d3lc00860f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 12/12/2023] [Indexed: 02/28/2024]
Abstract
A molecular robot, which is a system comprised of one or more molecular machines and computers, can execute sophisticated tasks in many fields that span from nanomedicine to green nanotechnology. The core parts of molecular robots are fairly consistent from system to system and always include (i) a body to encapsulate molecular machines, (ii) sensors to capture signals, (iii) computers to make decisions, and (iv) actuators to perform tasks. This review aims to provide an overview of approaches and considerations to develop molecular robots. We first introduce the basic technologies required for constructing the core parts of molecular robots, describe the recent progress towards achieving higher functionality, and subsequently discuss the current challenges and outlook. We also highlight the applications of molecular robots in sensing biomarkers, signal communications with living cells, and conversion of energy. Although molecular robots are still in their infancy, they will unquestionably initiate massive change in biomedical and environmental technology in the not too distant future.
Collapse
Affiliation(s)
- Zugui Peng
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei-shi, Tokyo185-8588, Japan.
| | - Shoji Iwabuchi
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei-shi, Tokyo185-8588, Japan.
| | - Kayano Izumi
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei-shi, Tokyo185-8588, Japan.
| | - Sotaro Takiguchi
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei-shi, Tokyo185-8588, Japan.
| | - Misa Yamaji
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei-shi, Tokyo185-8588, Japan.
| | - Shoko Fujita
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei-shi, Tokyo185-8588, Japan.
| | - Harune Suzuki
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei-shi, Tokyo185-8588, Japan.
| | - Fumika Kambara
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei-shi, Tokyo185-8588, Japan.
| | - Genki Fukasawa
- School of Life Science and Technology, Tokyo Institute of Technology, Ookayama 2-12-1, Meguro-Ku, Tokyo 152-8550, Japan
| | - Aileen Cooney
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, UK
| | - Lorenzo Di Michele
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge CB3 0AS, UK
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, UK
- FabriCELL, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, UK
| | - Yuval Elani
- Department of Chemical Engineering, Imperial College London, South Kensington, London SW7 2AZ, UK
- FabriCELL, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, UK
| | - Tomoaki Matsuura
- Earth-Life Science Institute, Tokyo Institute of Technology, Ookayama 2-12-1, Meguro-Ku, Tokyo 152-8550, Japan
| | - Ryuji Kawano
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei-shi, Tokyo185-8588, Japan.
| |
Collapse
|
33
|
Luo ZH, Chen C, Zhao QH, Deng NN. Functional metal-phenolic cortical cytoskeleton for artificial cells. SCIENCE ADVANCES 2024; 10:eadj4047. [PMID: 38363847 PMCID: PMC10871533 DOI: 10.1126/sciadv.adj4047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 01/16/2024] [Indexed: 02/18/2024]
Abstract
Cortex-like cytoskeleton, a thin layer of cross-linked cytoplasmic proteins underlying the cell membrane, plays an essential role in modulating membrane behavior and cell surface properties. However, bottom-up construction of functional cortex-like cytoskeleton in artificial cells remains a challenge. Here, we present metal-phenolic networks as artificial cortical cytoskeletons in liposome-based artificial cells. The metal-phenolic cytoskeleton-reinforced artificial cells exhibit long-term stability, enhanced resistance to a variety of harsh environments, tunable permeability, and well-controlled morphologies. We anticipate that our stable artificial cell models will stride forward to practical applications of liposome-based microsystem.
Collapse
Affiliation(s)
- Zhen-Hong Luo
- Shanghai Jiao Tong University, School of Chemistry and Chemical Engineering, Shanghai 200240, China
| | - Chen Chen
- Shanghai Jiao Tong University, School of Chemistry and Chemical Engineering, Shanghai 200240, China
| | - Qi-Hong Zhao
- Shanghai Jiao Tong University, School of Chemistry and Chemical Engineering, Shanghai 200240, China
| | - Nan-Nan Deng
- Shanghai Jiao Tong University, School of Chemistry and Chemical Engineering, Shanghai 200240, China
- Shanghai Jiao Tong University Sichuan Research Institute, Chengdu 610213, Sichuan, China
| |
Collapse
|
34
|
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
|
35
|
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: 15] [Impact Index Per Article: 15.0] [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
|
36
|
Ushiyama R, Nanjo S, Tsugane M, Sato R, Matsuura T, Suzuki H. Identifying Conditions for Protein Synthesis Inside Giant Vesicles Using Microfluidics toward Standardized Artificial Cell Production. ACS Synth Biol 2024; 13:68-76. [PMID: 38032418 DOI: 10.1021/acssynbio.3c00629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2023]
Abstract
To expand the range of practical applications of artificial cells, it is important to standardize the production process of giant (cell-sized) vesicles that encapsulate reconstituted biochemical reaction systems. For this purpose, a rapidly developing microfluidics-based giant vesicle generation system is a promising approach, similar to the droplet assay systems that are already widespread in the market. In this study, we examined the composition of the solutions used to generate vesicles encapsulating the in vitro transcription-translation (IVTT) system. We show that tuning of the lipid composition and adding poly(vinyl alcohol) to the outer solution improved the stability of the transition process into the lipid membrane so that protein synthesis proceeded in vesicles. The direct integration of α-hemolysin nanopores synthesized in situ was also demonstrated. These protein-synthesizing monodisperse giant vesicles can be prepared by using a simple microfluidic fabrication/operation with a commercial IVTT system.
Collapse
Affiliation(s)
- Ryota Ushiyama
- Department of Precision Mechanics, Faculty of Science and Engineering, Chuo University, 1-13-27 Kasuga, Bunkyo-ku, Tokyo 112-8551, Japan
| | - Satoshi Nanjo
- Department of Precision Mechanics, Faculty of Science and Engineering, Chuo University, 1-13-27 Kasuga, Bunkyo-ku, Tokyo 112-8551, Japan
| | - Mamiko Tsugane
- Department of Precision Mechanics, Faculty of Science and Engineering, Chuo University, 1-13-27 Kasuga, Bunkyo-ku, Tokyo 112-8551, Japan
| | - Reiko Sato
- Department of Precision Mechanics, Faculty of Science and Engineering, Chuo University, 1-13-27 Kasuga, Bunkyo-ku, Tokyo 112-8551, Japan
| | - Tomoaki Matsuura
- Earth-Life Science Institute, Tokyo Institute of Technology, 2-12-1-i7E Ookayama, Meguro-ku, Tokyo 152-8550, Japan
| | - Hiroaki Suzuki
- Department of Precision Mechanics, Faculty of Science and Engineering, Chuo University, 1-13-27 Kasuga, Bunkyo-ku, Tokyo 112-8551, Japan
| |
Collapse
|
37
|
Douliez JP. Double Emulsion Droplets as a Plausible Step to Fatty Acid Protocells. SMALL METHODS 2023; 7:e2300530. [PMID: 37574259 DOI: 10.1002/smtd.202300530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 07/07/2023] [Indexed: 08/15/2023]
Abstract
It is assumed that life originated on the Earth from vesicles made of fatty acids. These amphiphiles are the simplest chemicals, which can be present in the prebiotic soup, capable of self-assembling into compartments mimicking modern cells. Production of fatty acid vesicles is widely studied, as their growing and division. However, how prebiotic chemicals require to further yield living cells encapsulated within protocells remains unclear. Here, one suggests a scenario based on recent studies, which shows that phospholipid vesicles can form from double emulsions affording facile encapsulation of cargos. In these works, water-in-oil-in-water droplets are produced by microfluidics, having dispersed lipids in the oil. Dewetting of the oil droplet leaves the internal aqueous droplet covered by a lipid bilayer, entrapping cargos. In this review, formation of fatty acid protocells is briefly reviewed, together with the procedure for preparing double emulsions and vesicles from double emulsion and finally, it is proposed that double emulsion droplets formed in the deep ocean where undersea volcano expulsed materials, with fatty acids (under their carboxylic form) and alkanols as the oily phase, entrapping hydrosoluble prebiotic chemicals in a double emulsion droplet core. Once formed, double emulsion droplets can move up to the surface, where an increase of pH, variation of pressure and/or temperature may have allowed dewetting of the oily droplet, leaving a fatty acid vesicular protocell with encapsulated prebiotic materials.
Collapse
Affiliation(s)
- Jean-Paul Douliez
- Biologie du Fruit et Pathologie, UMR 1332, Institut National de Recherche Agronomique (INRAE), Université De Bordeaux, Villenave d'Ornon, F-33140, France
| |
Collapse
|
38
|
Xu Q, Zhang Z, Lui PPY, Lu L, Li X, Zhang X. Preparation and biomedical applications of artificial cells. Mater Today Bio 2023; 23:100877. [PMID: 38075249 PMCID: PMC10701372 DOI: 10.1016/j.mtbio.2023.100877] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 11/16/2023] [Accepted: 11/19/2023] [Indexed: 10/16/2024] Open
Abstract
Artificial cells have received much attention in recent years as cell mimics with typical biological functions that can be adapted for therapeutic and diagnostic applications, as well as having an unlimited supply. Although remarkable progress has been made to construct complex multifunctional artificial cells, there are still significant differences between artificial cells and natural cells. It is therefore important to understand the techniques and challenges for the fabrication of artificial cells and their applications for further technological advancement. The key concepts of top-down and bottom-up methods for preparing artificial cells are summarized, and the advantages and disadvantages of the bottom-up methods are compared and critically discussed in this review. Potential applications of artificial cells as drug carriers (microcapsules), as signaling regulators for coordinating cellular communication and as bioreactors for biomolecule fabrication, are further discussed. The challenges and future trends for the development of artificial cells simulating the real activities of natural cells are finally described.
Collapse
Affiliation(s)
- Qian Xu
- Department of Materials Physics and Chemistry, School of Materials Science and Engineering, Key Laboratory for Anisotropy and Texture of Materials, Ministry of Education, Northeastern University, Shenyang, Liaoning, 110819, China
- Institute of Metal Research, Chinese Academy of Sciences, Shenyang, Liaoning, 110016, China
| | - Zeping Zhang
- Institute of Metal Research, Chinese Academy of Sciences, Shenyang, Liaoning, 110016, China
- School of Materials Science and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Pauline Po Yee Lui
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, 999077, Hong Kong
| | - Liang Lu
- Institute of Metal Research, Chinese Academy of Sciences, Shenyang, Liaoning, 110016, China
- School of Materials Science and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Xiaowu Li
- Department of Materials Physics and Chemistry, School of Materials Science and Engineering, Key Laboratory for Anisotropy and Texture of Materials, Ministry of Education, Northeastern University, Shenyang, Liaoning, 110819, China
| | - Xing Zhang
- Institute of Metal Research, Chinese Academy of Sciences, Shenyang, Liaoning, 110016, China
- School of Materials Science and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, China
| |
Collapse
|
39
|
Van de Cauter L, van Buren L, Koenderink GH, Ganzinger KA. Exploring Giant Unilamellar Vesicle Production for Artificial Cells - Current Challenges and Future Directions. SMALL METHODS 2023; 7:e2300416. [PMID: 37464561 DOI: 10.1002/smtd.202300416] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/30/2023] [Indexed: 07/20/2023]
Abstract
Creating an artificial cell from the bottom up is a long-standing challenge and, while significant progress has been made, the full realization of this goal remains elusive. Arguably, one of the biggest hurdles that researchers are facing now is the assembly of different modules of cell function inside a single container. Giant unilamellar vesicles (GUVs) have emerged as a suitable container with many methods available for their production. Well-studied swelling-based methods offer a wide range of lipid compositions but at the expense of limited encapsulation efficiency. Emulsion-based methods, on the other hand, excel at encapsulation but are only effective with a limited set of membrane compositions and may entrap residual additives in the lipid bilayer. Since the ultimate artificial cell will need to comply with both specific membrane and encapsulation requirements, there is still no one-method-fits-all solution for GUV formation available today. This review discusses the state of the art in different GUV production methods and their compatibility with GUV requirements and operational requirements such as reproducibility and ease of use. It concludes by identifying the most pressing issues and proposes potential avenues for future research to bring us one step closer to turning artificial cells into a reality.
Collapse
Affiliation(s)
- Lori Van de Cauter
- Autonomous Matter Department, AMOLF, Amsterdam, 1098 XG, The Netherlands
| | - Lennard van Buren
- Department of Bionanoscience, Kavli Institute of Nanoscience, Delft University of Technology, Delft, 2629 HZ, The Netherlands
| | - Gijsje H Koenderink
- Department of Bionanoscience, Kavli Institute of Nanoscience, Delft University of Technology, Delft, 2629 HZ, The Netherlands
| | | |
Collapse
|
40
|
Douliez JP, Arlaut A, Beven L, Fameau AL, Saint-Jalmes A. One step generation of single-core double emulsions from polymer-osmose-induced aqueous phase separation in polar oil droplets. SOFT MATTER 2023; 19:7562-7569. [PMID: 37751151 DOI: 10.1039/d3sm00970j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/27/2023]
Abstract
Water-in-oil-in-water emulsions (W/O/W) are aqueous droplet(s) embedded within oil droplets dispersed in a continuous water phase. They are attracting interest due to their possible applications from cosmetic to food science since both hydrosoluble and liposoluble cargos can be encapsulated within. They are generally prepared using a one-step or a two-step method, phase inversion and also via spontaneous emulsification. Here, we describe a general and simple one-step method based on hydrophilic polymers dispersed in polar oils to generate osmose-induced diffusion of water into oil droplets, forming polymer-rich aqueous droplets inside the oil droplets. Polyethylene glycol, but also other hydrophilic polymers (branched polyethylene imine or polyvinyl pyrrolidone) were successfully dispersed in 1-octanol or other polar oils (oleic acid or tributyrin) to produce an O/W emulsion that spontaneously transformed into a W1/O/W2 emulsion, with the inner aqueous droplet (W1) only containing the hydrophilic polymer initially dispersed in oil. By combining single drop experiments, with macroscopic viscosity measurements, we demonstrated that the double emulsion resulted of water diffusion, which amplitude could be adjusted by the polymer concentration. The production of high internal phase emulsions was also achieved, together with a pH-induced transition from multiple to single core double emulsion. We expect this new method for producing double emulsions to find applications in domains of microencapsulation and materials chemistry.
Collapse
Affiliation(s)
- Jean-Paul Douliez
- Univ. Bordeaux, INRAE, Biologie du Fruit et Pathologie, UMR 1332, F-33140 Villenave dOrnon, France.
| | - Anais Arlaut
- Univ Rennes, CNRS, IPR (Institut de Physique de Rennes), UMR 6251, F-35000, Rennes, France.
| | - Laure Beven
- Univ. Bordeaux, INRAE, Biologie du Fruit et Pathologie, UMR 1332, F-33140 Villenave dOrnon, France.
| | - Anne-Laure Fameau
- University Lille, CNRS, INRAE, Centrale Lille, UMET, 369 Rue Jules Guesde, F-59000 Lille, France
| | - Arnaud Saint-Jalmes
- Univ Rennes, CNRS, IPR (Institut de Physique de Rennes), UMR 6251, F-35000, Rennes, France.
| |
Collapse
|
41
|
Jahnke K, Göpfrich K. Engineering DNA-based cytoskeletons for synthetic cells. Interface Focus 2023; 13:20230028. [PMID: 37577007 PMCID: PMC10415745 DOI: 10.1098/rsfs.2023.0028] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 06/30/2023] [Indexed: 08/15/2023] Open
Abstract
The development and bottom-up assembly of synthetic cells with a functional cytoskeleton sets a major milestone to understand cell mechanics and to develop man-made machines on the nano- and microscale. However, natural cytoskeletal components can be difficult to purify, deliberately engineer and reconstitute within synthetic cells which therefore limits the realization of multifaceted functions of modern cytoskeletons in synthetic cells. Here, we review recent progress in the development of synthetic cytoskeletons made from deoxyribonucleic acid (DNA) as a complementary strategy. In particular, we explore the capabilities and limitations of DNA cytoskeletons to mimic functions of natural cystoskeletons like reversible assembly, cargo transport, force generation, mechanical support and guided polymerization. With recent examples, we showcase the power of rationally designed DNA cytoskeletons for bottom-up assembled synthetic cells as fully engineerable entities. Nevertheless, the realization of dynamic instability, self-replication and genetic encoding as well as contractile force generating motors remains a fruitful challenge for the complete integration of multifunctional DNA-based cytoskeletons into synthetic cells.
Collapse
Affiliation(s)
- Kevin Jahnke
- Biophysical Engineering Group, Max Planck Institute for Medical Research, Jahnstraße 29, 69120 Heidelberg, Germany
- Department of Physics and Astronomy, Heidelberg University, 69120 Heidelberg, Germany
| | - Kerstin Göpfrich
- Biophysical Engineering Group, Max Planck Institute for Medical Research, Jahnstraße 29, 69120 Heidelberg, Germany
- Center for Molecular Biology (ZMBH), Heidelberg University, Im Neuenheimer Feld 329, 69120 Heidelberg, Germany
| |
Collapse
|
42
|
Piunti C, Cimetta E. Microfluidic approaches for producing lipid-based nanoparticles for drug delivery applications. BIOPHYSICS REVIEWS 2023; 4:031304. [PMID: 38505779 PMCID: PMC10903496 DOI: 10.1063/5.0150345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 08/15/2023] [Indexed: 03/21/2024]
Abstract
The importance of drug delivery for disease treatment is supported by a vast literature and increasing ongoing clinical studies. Several categories of nano-based drug delivery systems have been considered in recent years, among which lipid-based nanomedicines, both artificial and cell-derived, remain the most approved. The best artificial systems in terms of biocompatibility and low toxicity are liposomes, as they are composed of phospholipids and cholesterol, the main components of cell membranes. Extracellular vesicles-biological nanoparticles released from cells-while resembling liposomes in size, shape, and structure, have a more complex composition with up to hundreds of different types of lipids, proteins, and carbohydrates in their membranes, as well as an internal cargo. Although nanoparticle technologies have revolutionized drug delivery by enabling passive and active targeting, increased stability, improved solubilization capacity, and reduced dose and adverse effects, the clinical translation remains challenging due to manufacturing limitations such as laborious and time-consuming procedures and high batch-to-batch variability. A sea change occurred when microfluidic strategies were employed, offering advantages in terms of precise particle handling, simplified workflows, higher sensitivity and specificity, and good reproducibility and stability over bulk methods. This review examines scientific advances in the microfluidics-mediated production of lipid-based nanoparticles for therapeutic applications. We will discuss the preparation of liposomes using both hydrodynamic focusing of microfluidic flow and mixing by herringbone and staggered baffle micromixers. Then, an overview on microfluidic approaches for producing extracellular vesicles and extracellular vesicles-mimetics for therapeutic applications will describe microfluidic extrusion, surface engineering, sonication, electroporation, nanoporation, and mixing. Finally, we will outline the challenges, opportunities, and future directions of microfluidic investigation of lipid-based nanoparticles in the clinic.
Collapse
|
43
|
He C, He X, Zhang Y, Han X, Yang Y, Shen Y, Wang T, Wu Q, Yang Y, Xu W, Bai J, Wang Z. Development of a Microfluidic Formatted Ultrasound-Controlled Monodisperse Lipid Vesicles' Hydrogel Dressing Combined with Ultrasound for Transdermal Drug Delivery System. Macromol Biosci 2023; 23:e2300049. [PMID: 37178331 DOI: 10.1002/mabi.202300049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 05/09/2023] [Indexed: 05/15/2023]
Abstract
Transdermal drug delivery system (TDDS) has attracted much attention in the pharmaceutical technology area. However, the current methods are difficult to ensure penetration efficiency, controllability, and safety in the dermis, so its widespread clinical use has been limited. This work proposes an ultrasound-controlled monodisperse lipid vesicles (U-CMLVs) hydrogel dressing, which combines with ultrasound to form TDDS. Using microfluidic technology, prepare size controllable U-CMLVs with high drug encapsulation efficiency and quantitative encapsulation of ultrasonic response materials, and even uniform mix them with hydrogel to prepare the required thickness of dressings. The high encapsulation efficiency can ensure sufficient dosage of the drugs and further realize the control of ultrasonic response through quantitative encapsulation of ultrasound-responsive materials. Using high frequency (5 MHz, 0.4 W cm-2 ) and low frequency (60 kHz, 1 W cm-2 ) ultrasound to control the movement and rupture of U-CMLVs, the contents not only penetrate the stratum corneum into the epidermis but also break through the bottleneck of penetration efficiency, and deep into the dermis. These findings provide the groundwork for deep, controllable, efficient, and safe drug delivery through TDDS and lay a foundation for further expanding its application.
Collapse
Affiliation(s)
- Chengdian He
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
- Chongqing Key Laboratory of Biomedical Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Xiong He
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
- Chongqing Key Laboratory of Biomedical Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Yi Zhang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
- Chongqing Key Laboratory of Biomedical Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Xiaofeng Han
- Department of Dermatology, Chongqing First People's Hospital and Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400011, China
| | - Yujun Yang
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical Laboratory Microfluidics and SPRi Engineering Research Center Chongqing Medical University, Chongqing, 400016, China
| | - Yong Shen
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
- Chongqing Key Laboratory of Biomedical Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Teng Wang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
- Chongqing Key Laboratory of Biomedical Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Qing Wu
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
- Chongqing Key Laboratory of Biomedical Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Yukun Yang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
- Chongqing Key Laboratory of Biomedical Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Wei Xu
- Department of Dermatology, Chongqing First People's Hospital and Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400011, China
| | - Jin Bai
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
- Chongqing Key Laboratory of Biomedical Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Zhenyu Wang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
- Chongqing Key Laboratory of Biomedical Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| |
Collapse
|
44
|
Pilkington CP, Contini C, Barritt JD, Simpson PA, Seddon JM, Elani Y. A microfluidic platform for the controlled synthesis of architecturally complex liquid crystalline nanoparticles. Sci Rep 2023; 13:12684. [PMID: 37542147 PMCID: PMC10403506 DOI: 10.1038/s41598-023-39205-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 07/21/2023] [Indexed: 08/06/2023] Open
Abstract
Soft-matter nanoparticles are of great interest for their applications in biotechnology, therapeutic delivery, and in vivo imaging. Underpinning this is their biocompatibility, potential for selective targeting, attractive pharmacokinetic properties, and amenability to downstream functionalisation. Morphological diversity inherent to soft-matter particles can give rise to enhanced functionality. However, this diversity remains untapped in clinical and industrial settings, and only the simplest of particle architectures [spherical lipid vesicles and lipid/polymer nanoparticles (LNPs)] have been routinely exploited. This is partially due to a lack of appropriate methods for their synthesis. To address this, we have designed a scalable microfluidic hydrodynamic focusing (MHF) technology for the controllable, rapid, and continuous production of lyotropic liquid crystalline (LLC) nanoparticles (both cubosomes and hexosomes), colloidal dispersions of higher-order lipid assemblies with intricate internal structures of 3-D and 2-D symmetry. These particles have been proposed as the next generation of soft-matter nano-carriers, with unique fusogenic and physical properties. Crucially, unlike alternative approaches, our microfluidic method gives control over LLC size, a feature we go on to exploit in a fusogenic study with model cell membranes, where a dependency of fusion on particle diameter is evident. We believe our platform has the potential to serve as a tool for future studies involving non-lamellar soft nanoparticles, and anticipate it allowing for the rapid prototyping of LLC particles of diverse functionality, paving the way toward their eventual wide uptake at an industrial level.
Collapse
Affiliation(s)
- Colin P Pilkington
- Department of Chemistry, Molecular Science Research Hub, Imperial College London, 82 Wood Lane, London, W12 0BZ, UK.
- Department of Chemical Engineering, Imperial College London, Exhibition Road, London, SW7 2AZ, UK.
| | - Claudia Contini
- Department of Chemical Engineering, Imperial College London, Exhibition Road, London, SW7 2AZ, UK
| | - Joseph D Barritt
- Department of Life Sciences, Imperial College London, Exhibition Road, London, SW7 2AZ, UK
| | - Paul A Simpson
- Department of Life Sciences, Centre for Structural Biology, Imperial College London, Exhibition Road, London, SW7 2AZ, UK
| | - John M Seddon
- Department of Chemistry, Molecular Science Research Hub, Imperial College London, 82 Wood Lane, London, W12 0BZ, UK
| | - Yuval Elani
- Department of Chemical Engineering, Imperial College London, Exhibition Road, London, SW7 2AZ, UK.
| |
Collapse
|
45
|
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
|
46
|
Zahid AA, Chakraborty A, Luo W, Coyle A, Paul A. Tailoring the Inherent Properties of Biobased Nanoparticles for Nanomedicine. ACS Biomater Sci Eng 2023. [PMID: 37378614 DOI: 10.1021/acsbiomaterials.3c00364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/29/2023]
Abstract
Biobased nanoparticles are at the leading edge of the rapidly developing field of nanomedicine and biotherapeutics. Their unique size, shape, and biophysical properties make them attractive tools for biomedical research, including vaccination, targeted drug delivery, and immune therapy. These nanoparticles are engineered to present native cell receptors and proteins on their surfaces, providing a biomimicking camouflage for therapeutic cargo to evade rapid degradation, immune rejection, inflammation, and clearance. Despite showing promising clinical relevance, commercial implementation of these biobased nanoparticles is yet to be fully realized. In this perspective, we discuss advanced biobased nanoparticle designs used in medical applications, such as cell membrane nanoparticles, exosomes, and synthetic lipid-derived nanoparticles, and highlight their benefits and potential challenges. Moreover, we critically assess the future of preparing such particles using artificial intelligence and machine learning. These advanced computational tools will be able to predict the functional composition and behavior of the proteins and cell receptors present on the nanoparticle surfaces. With more advancement in designing new biobased nanoparticles, this field of research could play a key role in dictating the future rational design of drug transporters, thereby ultimately improving overall therapeutic outcomes.
Collapse
Affiliation(s)
- Alap Ali Zahid
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, Ontario N6A 5B9, Canada
| | - Aishik Chakraborty
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, Ontario N6A 5B9, Canada
| | - Wei Luo
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, Ontario N6A 5B9, Canada
| | - Ali Coyle
- School of Biomedical Engineering, The University of Western Ontario, London, Ontario N6A 5B9, Canada
| | - Arghya Paul
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, Ontario N6A 5B9, Canada
- School of Biomedical Engineering, The University of Western Ontario, London, Ontario N6A 5B9, Canada
- Department of Chemistry, The Centre for Advanced Materials and Biomaterials Research, The University of Western Ontario, London, Ontario N6A 5B9, Canada
| |
Collapse
|
47
|
Noba K, Yoshimoto S, Tanaka Y, Yokoyama T, Matsuura T, Hori K. Simple Method for the Creation of a Bacteria-Sized Unilamellar Liposome with Different Proteins Localized to the Respective Sides of the Membrane. ACS Synth Biol 2023; 12:1437-1446. [PMID: 37155350 DOI: 10.1021/acssynbio.2c00564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Artificial cells are membrane vesicles mimicking cellular functions. To date, giant unilamellar vesicles made from a single lipid membrane with a diameter of 10 μm or more have been used to create artificial cells. However, the creation of artificial cells that mimic the membrane structure and size of bacteria has been limited due to technical restrictions of conventional liposome preparation methods. Here, we created bacteria-sized large unilamellar vesicles (LUVs) with proteins localized asymmetrically to the lipid bilayer. Liposomes containing benzylguanine-modified phospholipids were prepared by combining the conventional water-in-oil emulsion method and the extruder method, and green fluorescent protein fused with SNAP-tag was localized to the inner leaflet of the lipid bilayer. Biotinylated lipid molecules were then inserted externally, and the outer leaflet was modified with streptavidin. The resulting liposomes had a size distribution in the range of 500-2000 nm with a peak at 841 nm (the coefficient of variation was 10.3%), which was similar to that of spherical bacterial cells. Fluorescence microscopy, quantitative evaluation using flow cytometry, and western blotting proved the intended localization of different proteins on the lipid membrane. Cryogenic electron microscopy and quantitative evaluation by α-hemolysin insertion revealed that most of the created liposomes were unilamellar. Our simple method for the preparation of bacteria-sized LUVs with asymmetrically localized proteins will contribute to the creation of artificial bacterial cells for investigating functions and the significance of their surface structure and size.
Collapse
Affiliation(s)
- Kosaku Noba
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, Aichi 464-8603, Japan
| | - Shogo Yoshimoto
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, Aichi 464-8603, Japan
| | - Yoshikazu Tanaka
- Graduate School of Life Sciences, Tohoku University, 2-1-1 Katahira, Aoba-Ku, Sendai 980-8577, Japan
| | - Takeshi Yokoyama
- Graduate School of Life Sciences, Tohoku University, 2-1-1 Katahira, Aoba-Ku, Sendai 980-8577, Japan
| | - Tomoaki Matsuura
- Earth-Life Science Institute, Tokyo Institute of Technology, Ookayama 2-12-1-i7E-307, Meguro-Ku, Tokyo 152-8550, Japan
| | - Katsutoshi Hori
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, Aichi 464-8603, Japan
| |
Collapse
|
48
|
Liu Y, Lin G, Medina-Sánchez M, Guix M, Makarov D, Jin D. Responsive Magnetic Nanocomposites for Intelligent Shape-Morphing Microrobots. ACS NANO 2023; 17:8899-8917. [PMID: 37141496 DOI: 10.1021/acsnano.3c01609] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
With the development of advanced biomedical theragnosis and bioengineering tools, smart and soft responsive microstructures and nanostructures have emerged. These structures can transform their body shape on demand and convert external power into mechanical actions. Here, we survey the key advances in the design of responsive polymer-particle nanocomposites that led to the development of smart shape-morphing microscale robotic devices. We overview the technological roadmap of the field and highlight the emerging opportunities in programming magnetically responsive nanomaterials in polymeric matrixes, as magnetic materials offer a rich spectrum of properties that can be encoded with various magnetization information. The use of magnetic fields as a tether-free control can easily penetrate biological tissues. With the advances in nanotechnology and manufacturing techniques, microrobotic devices can be realized with the desired magnetic reconfigurability. We emphasize that future fabrication techniques will be the key to bridging the gaps between integrating sophisticated functionalities of nanoscale materials and reducing the complexity and footprints of microscale intelligent robots.
Collapse
Affiliation(s)
- Yuan Liu
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen, 518055 Guangdong Province, P. R. China
| | - Gungun Lin
- Institute for Biomedical Materials and Devices, School of Mathematical and Physical Sciences, Faculty of Science, University of Technology Sydney, 15 Broadway, Ultimo, NSW 2007, Australia
| | - Mariana Medina-Sánchez
- Micro- and NanoBiomedical Engineering Group (MNBE), Institute for Integrative Nanosciences, Leibniz Institute for Solid State and Materials Research (IFW), 01069 Dresden, Germany
- Chair of Micro- and NanoSystems, Center for Molecular Bioengineering (B CUBE), Dresden University of Technology, 01062 Dresden, Germany
| | - Maria Guix
- Universitat de Barcelona, Departament de Ciència dels Materials i Química Física, Institut de Química Teòrica i Computacional Barcelona, 08028 Barcelona, Spain
| | - Denys Makarov
- Helmholtz-Zentrum Dresden-Rossendorf e.V., Institute of Ion Beam Physics and Materials Research, Bautzner Landstrasse 400, 01328 Dresden, Germany
| | - Dayong Jin
- Institute for Biomedical Materials and Devices, School of Mathematical and Physical Sciences, Faculty of Science, University of Technology Sydney, 15 Broadway, Ultimo, NSW 2007, Australia
| |
Collapse
|
49
|
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
|
50
|
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
|