Imperial College London

DrYuvalElani

Faculty of EngineeringDepartment of Chemical Engineering

Senior Lecturer
 
 
 
//

Contact

 

+44 (0)20 7594 1208y.elani Website CV

 
 
//

Location

 

413ACE ExtensionSouth Kensington Campus

//

Summary

 

Publications

Publication Type
Year
to

61 results found

Karamdad K, Hindley J, Friddin MS, Bolognesi G, Law RV, Brooks NJ, Ces O, Elani Yet al., 2018, Engineering thermoresponsive phase separated vesicles formed via emulsion phase transfer as a content-release platform, Chemical Science, Vol: 9, Pages: 4851-4858, ISSN: 2041-6520

Giant unilamellar vesicles (GUVs) are a well-established tool for the study of membrane biophysics and are increasingly used as artificial cell models and functional units in biotechnology. This trend is driven by the development of emulsion-based generation methods such as Emulsion Phase Transfer (EPT), which facilitates the encapsulation of almost any water-soluble compounds (including biomolecules) regardless of size or charge, is compatible with droplet microfluidics, and allows GUVs with asymmetric bilayers to be assembled. However, the ability to control the composition of membranes formed via EPT remains an open question; this is key as composition gives rise to an array of biophysical phenomena which can be used to add functionality to membranes. Here, we evaluate the use of GUVs constructed via this method as a platform for phase behaviour studies and take advantage of composition-dependent features to engineer thermally-responsive GUVs. For the first time, we generate ternary GUVs (DOPC/DPPC/cholesterol) using EPT, and by compensating for the lower cholesterol incorporation efficiencies, show that these possess the full range of phase behaviour displayed by electroformed GUVs. As a demonstration of the fine control afforded by this approach, we demonstrate release of dye and peptide cargo when ternary GUVs are heated through the immiscibility transition temperature, and show that release temperature can be tuned by changing vesicle composition. We show that GUVs can be individually addressed and release triggered using a laser beam. Our findings validate EPT as a suitable method for generating phase separated vesicles and provide a valuable proof-of-concept for engineering content release functionality into individually addressable vesicles, which could have a host of applications in the development of smart synthetic biosystems.

Journal article

Hindley JW, Elani Y, McGilvery CM, Ali S, Bevan CL, Law R, Ces Oet al., 2018, Light-triggered enzymatic reactions in nested vesicle reactors, Nature Communications, Vol: 9, Pages: 1-6, ISSN: 2041-1723

Cell-sized vesicles have tremendous potential both as miniaturised pL reaction vessels and in bottom-up synthetic biology as chassis for artificial cells. In both these areas the introduction of light-responsive modules affords increased functionality, for example, to initiate enzymatic reactions in the vesicle interior with spatiotemporal control. Here we report a system composed of nested vesicles where the inner compartments act as phototransducers, responding to ultraviolet irradiation through diacetylene polymerisation-induced pore formation to initiate enzymatic reactions. The controlled release and hydrolysis of a fluorogenic β-galactosidase substrate in the external compartment is demonstrated, where the rate of reaction can be modulated by varying ultraviolet exposure time. Such cell-like nested microreactor structures could be utilised in fields from biocatalysis through to drug delivery.

Journal article

Elani Y, Trantidou T, Wylie D, Dekker L, Polizzi K, Law R, Ces Oet al., 2018, Constructing vesicle-based artificial cells with embedded living cells as organelle-like modules, Scientific Reports, Vol: 8, Pages: 1-8, ISSN: 2045-2322

There is increasing interest in constructing artificial cells by functionalising lipid vesicles with biological and synthetic machinery. Due to their reduced complexity and lack of evolved biochemical pathways, the capabilities of artificial cells are limited in comparison to their biological counterparts. We show that encapsulating living cells in vesicles provides a means for artificial cells to leverage cellular biochemistry, with the encapsulated cells serving organelle-like functions as living modules inside a larger synthetic cell assembly. Using microfluidic technologies to construct such hybrid cellular bionic systems, we demonstrate that the vesicle host and the encapsulated cell operate in concert. The external architecture of the vesicle shields the cell from toxic surroundings, while the cell acts as a bioreactor module that processes encapsulated feedstock which is further processed by a synthetic enzymatic metabolism co-encapsulated in the vesicle.

Journal article

Thomas JM, Friddin MS, Ces O, Elani Yet al., 2017, Programming membrane permeability using integrated membrane pores and blockers as molecular regulators, Chemical Communications, Vol: 53, Pages: 12282-12285, ISSN: 1359-7345

We report a bottom-up synthetic biology approach to engineering vesicles with programmable permeabilities. Exploiting the concentration-dependent relationship between constitutively active pores (alpha-hemolysin) and blockers allows blockers to behave as molecular regulators for tuning permeability, enabling us to systematically modulate cargo release kinetics without changing the lipid fabric of the system.

Journal article

de Bruin A, Friddin MS, Elani Y, Brooks N, Law R, Seddon J, Ces Oet al., 2017, A transparent 3D printed device for assembling droplet hydrogel bilayers (DHBs), RSC Advances, Vol: 7, Pages: 47796-47800, ISSN: 2046-2069

We report a new approach for assembling droplet hydrogel bilayers (DHBs) using a transparent 3D printed device. We characterise the transparency of our platform, confirm bilayer formation using electrical measurements and show that single-channel recordings can be obtained using our reusable rapid prototyped device. This method significantly reduces the cost and infrastructure required to develop devices for DHB assembly and downstream study.

Journal article

Trantidou T, Friddin M, Elani Y, Brooks NJ, Law RV, Seddon JM, Ces Oet al., 2017, Engineering compartmentalized biomimetic micro- and nanocontainers, ACS Nano, Vol: 11, Pages: 6549-6565, ISSN: 1936-086X

Compartmentalization of biological content and function is a key architectural feature in biology, where membrane bound micro- and nanocompartments are used for performing a host of highly specialized and tightly regulated biological functions. The benefit of compartmentalization as a design principle is behind its ubiquity in cells and has led to it being a central engineering theme in construction of artificial cell-like systems. In this review, we discuss the attractions of designing compartmentalized membrane-bound constructs and review a range of biomimetic membrane architectures that span length scales, focusing on lipid-based structures but also addressing polymer-based and hybrid approaches. These include nested vesicles, multicompartment vesicles, large-scale vesicle networks, as well as droplet interface bilayers, and double-emulsion multiphase systems (multisomes). We outline key examples of how such structures have been functionalized with biological and synthetic machinery, for example, to manufacture and deliver drugs and metabolic compounds, to replicate intracellular signaling cascades, and to demonstrate collective behaviors as minimal tissue constructs. Particular emphasis is placed on the applications of these architectures and the state-of-the-art microfluidic engineering required to fabricate, functionalize, and precisely assemble them. Finally, we outline the future directions of these technologies and highlight how they could be applied to engineer the next generation of cell models, therapeutic agents, and microreactors, together with the diverse applications in the emerging field of bottom-up synthetic biology.

Journal article

Salehi-Reyhani A, Ces O, Elani Y, 2017, Artificial cell mimics as simplified models for the study of cell biology, Experimental Biology and Medicine, Vol: 242, Pages: 1309-1317, ISSN: 1535-3702

Living cells are hugely complex chemical systems composed of a milieu of distinct chemical species (including DNA, proteins, lipids, and metabolites) interconnected with one another through a vast web of interactions: this complexity renders the study of cell biology in a quantitative and systematic manner a difficult task. There has been an increasing drive towards the utilization of artificial cells as cell mimics to alleviate this, a development that has been aided by recent advances in artificial cell construction. Cell mimics are simplified cell-like structures, composed from the bottom-up with precisely defined and tunable compositions. They allow specific facets of cell biology to be studied in isolation, in a simplified environment where control of variables can be achieved without interference from a living and responsive cell. This mini-review outlines the core principles of this approach and surveys recent key investigations that use cell mimics to address a wide range of biological questions. It will also place the field in the context of emerging trends, discuss the associated limitations, and outline future directions of the field.

Journal article

Trantidou T, Elani Y, Parsons E, Ces Oet al., 2017, Hydrophilic surface modification of PDMS for droplet microfluidics using a simple, quick, and robust method via PVA deposition, Microsystems and Nanoengineering, Vol: 3, ISSN: 2055-7434

Polydimethylsiloxane (PDMS) is a dominant material in the fabrication of microfluidic devices to generate water-in-oil droplets, particularly lipid-stabilized droplets, because of its highly hydrophobic nature. However, its key property of hydrophobicity has hindered its use in the microfluidic generation of oil-in-water droplets, which requires channels to have hydrophilic surface properties. In this article, we developed, optimized, and characterized a method to produce PDMS with a hydrophilic surface via the deposition of polyvinyl alcohol following plasma treatment and demonstrated its suitability for droplet generation. The proposed method is simple, quick, effective, and low cost and is versatile with respect to surfactants, with droplets being successfully generated using both anionic surfactants and more biologically relevant phospholipids. This method also allows the device to be selectively patterned with both hydrophilic and hydrophobic regions, leading to the generation of double emulsions and inverted double emulsions.

Journal article

Friddin MS, Bolognesi G, Elani Y, Brooks N, Law R, Seddon J, Neil M, ces Oet al., 2016, Optically assembled droplet interface bilayer (OptiDIB) networks from cell-sized microdroplets, Soft Matter, Vol: 12, Pages: 7731-7734, ISSN: 1744-6848

We report a new platform technology to systematically assemble droplet interface bilayer (DIB) networks in user-defined 3D architectures from cell-sized droplets using optical tweezers. Our OptiDIB platform is the first demonstration of optical trapping to precisely construct 3D DIB networks, paving the way for the development of a new generation of modular bio-systems.

Journal article

Friddin MS, Bolognesi G, Elani Y, Brooks N, Law R, Seddon J, Neil M, Ces Oet al., 2016, The optical assembly of bilayer networks from cell-sized droplets for synthetic biology, Systems and Synthetic Biology

Conference paper

Elani Y, 2016, Construction of membrane-bound artificial cells using microfluidics: a new frontier in bottom-up synthetic biology, Biochemical Society Transactions, Vol: 44, Pages: 723-730, ISSN: 1470-8752

The quest to construct artificial cells from the bottom-up using simple building blocks has received much attention over recent decades and is one of the grand challenges in synthetic biology. Cell mimics that are encapsulated by lipid membranes are a particularly powerful class of artificial cells due to their biocompatibility and the ability to reconstitute biological machinery within them. One of the key obstacles in the field centres on the following: how can membrane-based artificial cells be generated in a controlled way and in high-throughput? In particular, how can they be constructed to have precisely defined parameters including size, biomolecular composition and spatial organization? Microfluidic generation strategies have proved instrumental in addressing these questions. This article will outline some of the major principles underpinning membrane-based artificial cells and their construction using microfluidics, and will detail some recent landmarks that have been achieved.

Journal article

Friddin MS, Bolognesi G, Elani Y, Brooks N, Law R, Seddon J, Neil M, Ces Oet al., 2016, Optical tweezers to assemble 2D and 3D droplet interface bilayer networks from cell-sized droplets, EMBL Microfluidics

Conference paper

Elani Y, Solvas XC, Edel JB, Law RV, Ces Oet al., 2016, Microfluidic generation of encapsulated droplet interface bilayer networks (multisomes) and their use as cell-like reactors., Chemical Communications, Vol: 52, Pages: 5961-5964, ISSN: 1364-548X

Compartmentalised structures based on droplet interface bilayers (DIBs), including multisomes and compartmentalised vesicles, are seen by many as the next generation of biomimetic soft matter devices. Herein, we outline a microfluidic approach for the construction of miniaturised multisomes of pL volumes in high-throughput and demonstrate their potential as vehicles for in situ chemical synthesis.

Journal article

Ces O, Elani Y, Karamdad K, Friddin MS, Barter LMC, Bolognesi G, Law RV, Chan CL, Brooks NJ, Seddon JMet al., 2016, Novel microfluidic technologies for the bottom-up construction of artificial cells

This talk will outline novel microfluidic strategies for biomembrane engineering that are capable of fabricating vesicles [1], droplet interface bilayer networks [2], multisomes [3] and artificial tissues [4] where parameters such as membrane asymmetry, membrane curvature, compartment connectivity and individual compartment contents can be controlled. Various bulk methods, such as extrusion, gentle hydration and electroformation, have been synonymous with the formation of lipid vesicles over recent years. However these strategies suffer from significant shortcomings associated with these processes including limited control of vesicle structural parameters such as size, lamellarity, membrane composition and internal contents. To address this technological bottleneck we have developed novel microfluidic platforms to form lipid vesicles in high-Throughput with full control over the composition of both the inner and outer leaflet of the membrane thereby enabling the manufacture of symmetric and asymmetric vesicles. This is achieved by manufacturing microfluidic channels with a step junction, produced by double-layer photolithography, which facilitates the transfer of a W/O emulsion across an oil-water phase boundary and the self-Assembly of a phospholipid bilayer. These platforms are being used to explore the role of asymmetry in biological systems [1] and study the engineering rules that regulate membrane mediated protein-protein interactions [5]. In addition, these technologies are enabling the construction of biological machines capable of acting as micro-reactors [6], environmental sensors and smart delivery vehicles [5] as well as complex multi-compartment artificial cells where the contents and connectivity of each compartment can be controlled. These compartments are separated by biological functional membranes that can facilitate transport between the compartments themselves and between the compartments and external environment. This approach has led to the deve

Conference paper

Friddin MS, Bolognesi G, Elani Y, Brooks NJ, Law RV, Seddon JM, Neil MAA, Ces Oet al., 2016, Light-driven drag and drop assembly of micron-scale bilayer networks for synthetic biology, Pages: 545-546

We have developed a new method to assemble single- or multi-layered networks of droplet interface bilayers (DIBs) from cell-sized droplets using a single beam optical trap (optical tweezers). The novelty of our approach is the ability to directly trap the microdroplets with the laser and manipulate them in 3D to construct DIB networks of user-defined architectures. Our method does not require a complex optical setup, is versatile, contactless, benefits from both high spatial and temporal resolution, and could set a new paradigm for the assembly of smart, synthetic biosystems.

Conference paper

Trantidou T, Elani Y, Ces O, 2016, Versatile strategies for the microfluidic generation of lipid-stabilised double emulsions, Pages: 1595-1596

Lipid-stabilised double emulsions have recently gained much importance in translational healthcare as potential micro-bioreactors for the synthesis of high-end materials, in situ drug delivery, and as templates for artificial cells in synthetic biology. Whilst microfluidic generation of surfactantstabilised systems is well established, lipid-stabilised systems are notoriously more cumbersome to produce, since they require specific surface chemistries and many surface modification techniques are incompatible with lipids. This paper reports a simple, robust and versatile method for the microfluidic generation of stable and monodisperse double emulsions using biologically relevant phospholipids.

Conference paper

Carreras P, Elani Y, Law RV, Brooks NJ, Seddon JM, Ces Oet al., 2015, A microfluidic platform for size-dependent generation of droplet interface bilayer networks on rails, Biomicrofluidics, Vol: 9, ISSN: 1932-1058

Dropletinterface bilayer (DIB) networks are emerging as a cornerstone technology for the bottom up construction of cell-like and tissue-like structures and bio-devices. They are an exciting and versatile model-membrane platform, seeing increasing use in the disciplines of synthetic biology, chemical biology, and membrane biophysics. DIBs are formed when lipid-coated water-in-oil droplets are brought together—oil is excluded from the interface, resulting in a bilayer. Perhaps the greatest feature of the DIB platform is the ability to generate bilayer networks by connecting multiple droplets together, which can in turn be used in applications ranging from tissue mimics, multicellular models, and bio-devices. For such applications, the construction and release of DIB networks of defined size and composition on-demand is crucial. We have developed a droplet-based microfluidic method for the generation of different sized DIB networks (300–1500 pl droplets) on-chip. We do this by employing a droplet-on-rails strategy where droplets are guided down designated paths of a chip with the aid of microfabricated grooves or “rails,” and droplets of set sizes are selectively directed to specific rails using auxiliary flows. In this way we can uniquely produce parallel bilayer networks of defined sizes. By trapping several droplets in a rail, extended DIB networks containing up to 20 sequential bilayers could be constructed. The trapped DIB arrays can be composed of different lipid types and can be released on-demand and regenerated within seconds. We show that chemical signals can be propagated across the bio-network by transplanting enzymatic reaction cascades for inter-droplet communication.

Journal article

Elani Y, Law RV, Ces O, 2015, Erratum: Vesicle-based artificial cells: Recent developments and prospects for drug delivery (Therapeutic Delivery (2015) 6: 5 (541-543)), Therapeutic Delivery, Vol: 6, ISSN: 2041-5990

Journal article

Elani Y, Law RV, Ces O, 2015, Vesicle-based artificial cells: recent developments and prospects for drug delivery, THERAPEUTIC DELIVERY, Vol: 6, Pages: 541-543, ISSN: 2041-5990

Journal article

Elani Y, 2015, Development of Microfluidic Technologies for the Construction of Multi-Compartment Vesicles and their Applications as Artificial Cells

Thesis dissertation

Elani Y, Law R, Ces O, 2015, Vesicle-based artificial cells: recent developments and prospects for drug delivery, Therapeutic delivery, ISSN: 2041-6008

Journal article

Elani Y, Law RV, Ces O, 2015, Protein synthesis in artificial cells: using compartmentalisation for spatial organisation in vesicle bioreactors, PHYSICAL CHEMISTRY CHEMICAL PHYSICS, Vol: 17, Pages: 15534-15537, ISSN: 1463-9076

Journal article

Elani Y, Purushothaman S, Booth PJ, Seddon JM, Brooks NJ, Law RV, Ces Oet al., 2015, Measurements of the effect of membrane asymmetry on the mechanical properties of lipid bilayers, CHEMICAL COMMUNICATIONS, Vol: 51, Pages: 6976-6979, ISSN: 1359-7345

Journal article

Ces O, Elani Y, Law R, 2014, Vesicle-based artificial cells as chemical microreactors with spatially segregated reaction pathways, Nature Communications, Vol: 5, Pages: 1-5, ISSN: 2041-1723

In the discipline of bottom-up synthetic biology, vesicles define the boundaries of artificial cells and are increasingly being used as biochemical microreactors operating in physiological environments. As the field matures, there is a need to compartmentalize processes in different spatial localities within vesicles, and for these processes to interact with one another. Here we address this by designing and constructing multi-compartment vesicles within which an engineered multi-step enzymatic pathway is carried out. The individual steps are isolated in distinct compartments, and their products traverse into adjacent compartments with the aid of transmembrane protein pores, initiating subsequent steps. Thus, an engineered signalling cascade is recreated in an artificial cellular system. Importantly, by allowing different steps of a chemical pathway to be separated in space, this platform bridges the gap between table-top chemistry and chemistry that is performed within vesicles.

Journal article

Carreras P, Elani Y, Law R, Brooks N, Seddon J, Ceset al., 2014, A droplet trapping microfluidic device for the study of mass-transport across droplet interface bilayers, MicroTAS 2014

Conference paper

Elani Y, Law R, Ces O, 2014, Microfluidic generation of networked droplet collections and lipid membrane constructs, 18th International Conference on Miniaturized Systems for Chemistry and Life Sciences, Pages: 677-679, ISSN: 1556-5904

We report on microfluidic strategies to generate several multi-compartment membrane-basedstructures, including droplet interface bilayer networks and multi-compartment vesicles. Thesedevelopments allow the current status quo— where microdroplets are used as isolated vessels— to bechanged. By linking droplets together with lipid membranes, higher order systems can be generated, withparticular ramifications for bottom-up synthetic biology and for functional droplet-based microreactorsand biodevices.

Conference paper

Carreras P, Law RV, Brooks NJ, Seddon JM, Ces Oet al., 2014, Microfluidic generation of droplet interface bilayer networks incorporating real-time size sorting in linear and non-linear configurations, Biomicrofluidics, Vol: 8, ISSN: 1932-1058

In this study, a novel droplet based microfluidic method for the generation of different sized droplet interface bilayers is reported. A microfluidic platform was designed, which allows the generation and packing of picoliter lipid coated water droplets. Droplets were generated by hydrodynamic focusing coupled with selective transport along grooves according to their size. A trapping structure at the end of the groove and a fine control of the flow pressures allowed for the droplets to be successfully trapped and aligned on demand. This technology facilitates the fine control of droplet size production as well as the generation of extended networks from a variety of lipids including 1,2-diphytanoyl-sn-glycero-3- phosphocholine and 1,2-dioleoyl-sn-glycero-3-phosphocholine in linear and non- linear configurations, which is vital to the application of Droplet Interface Bilayers to biological network construction on-chip.

Journal article

Elani Y, Gee A, Law RV, Ces Oet al., 2013, Manufacturing vesicles with internal bilayer partitions: a novel unit for synthetic biology, 9th European-Biophysical-Societies-Association Congress, Publisher: SPRINGER, Pages: S55-S55, ISSN: 0175-7571

Conference paper

Elani Y, Gee A, Law RV, Ces Oet al., 2013, Engineering multi-compartment vesicle networks, CHEMICAL SCIENCE, Vol: 4, Pages: 3332-3338, ISSN: 2041-6520

Journal article

This data is extracted from the Web of Science and reproduced under a licence from Thomson Reuters. You may not copy or re-distribute this data in whole or in part without the written consent of the Science business of Thomson Reuters.

Request URL: http://wlsprd.imperial.ac.uk:80/respub/WEB-INF/jsp/search-html.jsp Request URI: /respub/WEB-INF/jsp/search-html.jsp Query String: id=00665983&limit=30&person=true&page=2&respub-action=search.html