Imperial College London

Professor Molly Stevens

Faculty of EngineeringDepartment of Materials

Professor of Biomedical Materials and Regenerative Medicine
 
 
 
//

Contact

 

+44 (0)20 7594 6804m.stevens

 
 
//

Location

 

208Royal School of MinesSouth Kensington Campus

//

Summary

 

Publications

Publication Type
Year
to

495 results found

Kit-Anan W, Mazo M, Wang BX, Leonardo V, Pence I, Gopal S, Gelmi A, Becce M, Chiappini C, Harding SE, Terracciano C, Stevens Met al., 2021, Multiplexing physical stimulation on single human induced pluripotent stem cell-derived cardiomyocytes for phenotype modulation, Biofabrication, Vol: 13, Pages: 1-16, ISSN: 1758-5082

Traditional in vitro bioengineering approaches whereby only individual biophysical cues are manipulated at any one time are highly inefficient, falling short when recapitulating the complexity of the cardiac environment. Multiple biophysical cues are present in the native myocardial niche and are essential during development, as well as in maintenance of adult cardiomyocyte (CM) phenotype in both health and disease. This study establishes a novel biofabrication workflow to study and manipulate hiPSC-CMs and to understand how these cells respond to a multiplexed biophysical environment, namely microscopic topography (3D shape resembling that of adult CM) and substrate stiffness, at a single cell level. Silicon masters were fabricated and developed to generate pillars of the desired 3D shapes, which would be used to mould the designed microwell arrays into a hydrogel. Polyacrylamide was modified with the incorporation of acrylic acid to provide a carboxylic group conjugation site for adhesion motifs, without comprising its capacity to modulate the stiffness. In this manner, individual parameters can be finely tuned independently within the hydrogel: the dimension of 3D shaped microwell and its stiffness. The design allows the platform to isolate single hiPSC-CMs to study solely biophysical cues in an absence of cell-cell physical interaction. Under physiologic-like physical conditions (3D shape resembling that of adult CM and 9.83 kPa substrate stiffness), isolated single hiPSC-CMs exhibit increased Cx-43 density, cell Peer reviewed version of the manuscript published in final form at Biofabrication (2020). membrane stiffness and calcium transient amplitude; co-expression of the subpopulation-related MYL2- MYL7 proteins; while displaying higher anisotropism in comparison to pathologic-like conditions (flat surface and 112 kPa substrate stiffness). This demonstrates that supplying a physiological or pathological microenvironment to an isolated single hiPSC-CM in absen

Journal article

Pinna A, Baghbaderani MT, Hernandez VV, Naruphontjirakul P, Li S, McFarlane T, Hachim D, Stevens MM, Porter AE, Jones JRet al., 2021, Nanoceria provides antioxidant and osteogenic properties to mesoporous silica nanoparticles for osteoporosis treatment, Acta Biomaterialia, Vol: 122, Pages: 365-376, ISSN: 1742-7061

Osteoporosis, a chronic metabolic bone disease, is the most common cause of fractures. Drugs for treating osteoporosis generally inhibit osteoclast (OC) activity, but are rarely aimed at encouraging new bone growth and often cause severe systemic side effects. Reactive oxygen species (ROS) are one of the key triggers of osteoporosis, by inducing osteoblast (OB) and osteocyte apoptosis and promoting osteoclastogenesis. Here we tested the capability of the ROS-scavenger nanoceria encapsulated within mesoporous silica nanoparticles (Ce@MSNs) to treat osteoporosis using a pre-osteoblast MC3T3-E1 cell monoculture in stressed and normal conditions. Ce@MSNs (diameter of 80 ± 10 nm) were synthesised following a scalable two-step process involving sol-gel and wet impregnation methods. The Ce@MSNs at concentration of 100 μg mL−1 induced a significant reduction in oxidative stress produced by t-butyl hydroperoxide and did not alter cell viability significantly. Confocal microscopy showed that MSNs and Ce@MsNs were internalised into the cytoplasm of the pre-osteoblasts after 24 h but were not in the nucleus, avoiding any DNA and RNA modifications. Ce@MSNs provoked mineralisation of the pre-osteoablasts without osteogenic supplements, which did not occur when the cells were exposed to MSN without nanoceria. In a co-culture system of MC3T3-E1 and RAW264.7 macrophages, the Ce@MSNs exhibited antioxidant capability and stimulated cell proliferation and osteogenic responses without adding osteogenic supplements to the culture. The work brings forward an effective platform based for facile synthesis of Ce@MSNs to interact with both OBs and OCs for treatment of osteoporosis.

Journal article

Lin Y, Penna M, Spicer CD, Higgins SG, Gelmi A, Kim N, Wang S-T, Wojciechowski JP, Pashuck ET, Yarovsky I, Stevens MMet al., 2021, High-throughput peptide derivatization toward supramolecular diversification in microtiter plates, ACS Nano, Vol: 15, Pages: 4034-4044, ISSN: 1936-0851

The evolution of life on earth eventually leads to the emergence of species with increased complexity and diversity. Similarly, evolutionary chemical space exploration in the laboratory is a key step to pursue the structural and functional diversity of supramolecular systems. Here, we present a powerful tool that enables rapid peptide diversification and employ it to expand the chemical space for supramolecular functions. Central to this strategy is the exploitation of palladium-catalyzed Suzuki-Miyaura cross-coupling reactions to direct combinatorial synthesis of peptide arrays in microtiter plates under an open atmosphere. Taking advantage of this in situ library design, our results unambiguously deliver a fertile platform for creating a set of intriguing peptide functions including green fluorescent protein-like peptide emitters with chemically encoded emission colors, hierarchical self-assembly into nano-objects, and macroscopic hydrogels. This work also offers opportunities for quickly surveying the diversified peptide arrays and thereby identifying the structural factors that modulate peptide properties.

Journal article

Puetzer JL, Ma T, Sallent I, Gelmi A, Stevens Met al., 2021, Driving hierarchical collagen fiber formation for functional tendon, ligament and meniscus replacement, Biomaterials, Vol: 269, Pages: 1-10, ISSN: 0142-9612

Hierarchical collagen fibers are the primary source of strength in musculoskeletal tendons, ligaments, and menisci. It has remained a challenge to develop these large fibers in engineered replacements or in vivo after injury. The objective of this study was to investigate the ability of restrained cell-seeded high density collagen gels to drive hierarchical fiber formation for multiple musculoskeletal tissues. We found boundary conditions applied to high density collagen gels were capable of driving tenocytes, ligament fibroblasts, and meniscal fibrochondrocytes to develop native-sized hierarchical collagen fibers 20–40 μm in diameter. The fibers organize similar to bovine juvenile collagen with native fibril banding patterns and hierarchical fiber bundles 50–350 μm in diameter by 6 weeks. Mirroring fiber organization, tensile properties of restrained samples improved significantly with time, reaching ~1 MPa. Additionally, tendon, ligament, and meniscal cells produced significantly different sized fibers, different degrees of crimp, and different GAG concentrations, which corresponded with respective juvenile tissue. To our knowledge, these are some of the largest, most organized fibers produced to date in vitro. Further, cells produced tissue specific hierarchical fibers, suggesting this system is a promising tool to better understand cellular regulation of fiber formation to better stimulate it in vivo after injury.

Journal article

Li C, Ouyang L, Armstrong J, Stevens Met al., 2021, Advances in the fabrication of biomaterials for gradient tissue engineering, Trends in Biotechnology, Vol: 39, Pages: 150-164, ISSN: 0167-7799

Natural tissues and organs exhibit an array of spatial gradients, from the polar-ized neural tube during embryonic development to the osteochondral interfacepresent at articulating joints. The strong structure–function relationships inthese heterogeneous tissues have sparked intensive research into the develop-ment of methods that can replicate physiological gradients in engineered tis-sues. In this Review, we consider different gradients present in natural tissuesand discuss their critical importance in functional tissue engineering. Using thisbasis, we consolidate the existing fabrication methods into four categories: addi-tive manufacturing, component redistribution, controlled phase changes, andpostmodification. We have illustrated this with recent examples, highlightedprominent trends in thefield, and outlined a set of criteria and perspectives forgradient fabrication.

Journal article

Solanki A, Lali F, Autefage H, Agarwal S, Nommeots-Nomm A, Metcalfe A, Stevens M, Jones Jet al., 2021, Bioactive glasses and electrospun composites that release cobalt to stimulate the HIF pathway for wound healing applications, Biomaterials Research, Vol: 25, ISSN: 2055-7124

BackgroundBioactive glasses are traditionally associated with bonding to bone through a hydroxycarbonate apatite (HCA) surface layer but the release of active ions is more important for bone regeneration. They are now being used to deliver ions for soft tissue applications, particularly wound healing. Cobalt is known to simulate hypoxia and provoke angiogenesis. The aim here was to develop new bioactive glass compositions designed to be scaffold materials to locally deliver pro-angiogenic cobalt ions, at a controlled rate, without forming an HCA layer, for wound healing applications.MethodsNew melt-derived bioactive glass compositions were designed that had the same network connectivity (mean number of bridging covalent bonds between silica tetrahedra), and therefore similar biodegradation rate, as the original 45S5 Bioglass. The amount of magnesium and cobalt in the glass was varied, with the aim of reducing or removing calcium and phosphate from the compositions. Electrospun poly(ε-caprolactone)/bioactive glass composites were also produced. Glasses were tested for ion release in dissolution studies and their influence on Hypoxia-Inducible Factor 1-alpha (HIF-1α) and expression of Vascular Endothelial Growth Factor (VEGF) from fibroblast cells was investigated.ResultsDissolution tests showed the silica rich layer differed depending on the amount of MgO in the glass, which influenced the delivery of cobalt. The electrospun composites delivered a more sustained ion release relative to glass particles alone. Exposing fibroblasts to conditioned media from these composites did not cause a detrimental effect on metabolic activity but glasses containing cobalt did stabilise HIF-1α and provoked a significantly higher expression of VEGF (not seen in Co-free controls).ConclusionsThe composite fibres containing new bioactive glass compositions delivered cobalt ions at a sustained rate, which could be mediated by the magnesium content of the glass. The dis

Journal article

Horgan C, Bergholt M, Nagelkerke A, Thin MZ, Pence IJ, Kauscher U, Kalber TL, Stuckey D, Stevens Met al., 2021, Integrated photodynamic Raman theranostic system for cancer diagnosis, treatment, and post-treatment molecular monitoring, Theranostics, Vol: 11, Pages: 2006-2019, ISSN: 1838-7640

Theranostics, the combination of diagnosis and therapy, has long held promise as a means to achieving personalised precision cancer treatments. However, despite its potential, theranostics has yet to realise significant clinical translation, largely due the complexity and overriding toxicity concerns of existing theranostic nanoparticle strategies.Methods: Here, we present an alternative nanoparticle-free theranostic approach based on simultaneous Raman spectroscopy and photodynamic therapy (PDT) in an integrated clinical platform for cancer theranostics.Results: We detail the compatibility of Raman spectroscopy and PDT for cancer theranostics, whereby Raman spectroscopic diagnosis can be performed on PDT photosensitiser-positive cells and tissues without inadvertent photosensitiser activation/photobleaching or impaired diagnostic capacity. We further demonstrate that our theranostic platform enables in vivo tumour diagnosis, treatment, and post-treatment molecular monitoring in real-time.Conclusion: This system thus achieves effective theranostic performance, providing a promising new avenue towards the clinical realisation of theranostics.

Journal article

Sero JE, Stevens MM, 2021, Nanoneedle-Based Materials for Intracellular Studies, BIO-NANOMEDICINE FOR CANCER THERAPY, Editors: Fontana, Santos, Publisher: SPRINGER INTERNATIONAL PUBLISHING AG, Pages: 191-219, ISBN: 978-3-030-58173-2

Book chapter

Maynard S, Gelmi A, Skaalure S, Pence I, Lee-Reeves C, Sero J, Whittaker T, Stevens Met al., 2020, Nanoscale molecular quantification of stem cell-hydrogel interactions, ACS Nano, Vol: 14, Pages: 17321-17332, ISSN: 1936-0851

A common approach to tailoring synthetic hydrogels for regenerative medicine applications involves incorporating RGD cell adhesion peptides, yet assessing the cellular response to engineered microenvironments at the nanoscale remains challenging. To date, no study has demonstrated how RGD concentration in hydrogels affects the presentation of individual cell surface receptors. Here we studied the interaction between human mesenchymal stem cells (hMSCs) and RGD-functionalized poly(ethylene glycol) hydrogels, by correlating macro- and nanoscale single-cell interfacial quantification techniques. We quantified RGD unbinding forces on a synthetic hydrogel using single cell atomic force spectroscopy, revealing that short-term binding of hMSCs was sensitive to RGD concentration. We also performed direct stochastic optical reconstruction microscopy (dSTORM) to quantify the molecular interactions between integrin α5β1 and a biomaterial, unexpectedly revealing that increased integrin clustering at the hydrogel-cell interface correlated with fewer available RGD binding sites. Our complementary, quantitative approach uncovered mechanistic insights into specific stem cell-hydrogel interactions, where dSTORM provides nanoscale sensitivity to RGD-dependent differences in cell surface localization of integrin α5β1. Our findings reveal that it is possible to precisely determine how peptide-functionalized hydrogels interact with cells at the molecular scale, thus providing a basis to fine-tune the spatial presentation of bioactive ligands.

Journal article

Brinker CJ, Buriak JM, Chan WCW, Chhowalla M, Glotzer SC, Gogotsi Y, Hammond PT, Hersam MC, Javey A, Kagan CR, Kataoka K, Khademhosseini A, Kim I-D, Kotov NA, Lee S-T, Lee YH, Li Y, Liz-Marzan LM, Millstone JE, Mulvaney P, Nel AE, Nordlander P, Parak WJ, Penner RM, Rogach AL, Schaak RE, Sood AK, Stevens MM, Wee ATS, Weil T, Wilson CG, Weiss PSet al., 2020, Growing Contributions of Nano in 2020, Publisher: AMER CHEMICAL SOC

Other

Potter M, Najer A, Kloeckner A, Zhang S, Holme MN, Nele V, Che J, Penders J, Saunders C, Doutch JJ, Edwards A, Ces O, Stevens Met al., 2020, Controlled dendrimersome nanoreactor system for localised hypochlorite-induced killing of bacteria, ACS Nano, Vol: 14, Pages: 17333-17353, ISSN: 1936-0851

Antibiotic resistance is a serious global health problem necessitating new bactericidal approaches such as nanomedicines. Dendrimersomes (DSs) have recently become a valuable alternative nanocarrier to polymersomes and liposomes due to their molecular definition and synthetic versatility. Despite this, their biomedical application is still in its infancy. Inspired by the localized antimicrobial function of neutrophil phagosomes and the versatility of DSs, a simple three-component DS-based nanoreactor with broad-spectrum bactericidal activity is presented. This was achieved by encapsulation of glucose oxidase (GOX) and myeloperoxidase (MPO) within DSs (GOX-MPO-DSs), self-assembled from an amphiphilic Janus dendrimer, that possesses a semipermeable membrane. By external addition of glucose to GOX-MPO-DS, the production of hypochlorite (−OCl), a highly potent antimicrobial, by the enzymatic cascade was demonstrated. This cascade nanoreactor yielded a potent bactericidal effect against two important multidrug resistant pathogens, Staphylococcus aureus (S. aureus) and Pseudomonas aeruginosa (P. aeruginosa), not observed for H2O2 producing nanoreactors, GOX-DS. The production of highly reactive species such as –OCl represents a harsh bactericidal approach that could also be cytotoxic to mammalian cells. This necessitates the development of strategies for activating –OCl production in a localized manner in response to a bacterial stimulus. One option of locally releasing sufficient amounts of substrate using a bacterial trigger (released toxins) was demonstrated with lipidic glucose-loaded giant unilamellar vesicles (GUVs), envisioning, e.g., implant surface modification with nanoreactors and GUVs for localized production of bactericidal agents in the presence of bacterial growth.

Journal article

Belessiotis-Richards A, Higgins S, Sansom MSP, Alexander-Katz A, Stevens Met al., 2020, Coarse Grained Simulations Suggest the Epsin N-Terminal Homology Domain Can Sense Membrane Curvature Without its Terminal Amphipathic Helix, ACS Nano, Vol: 14, Pages: 16919-6928, ISSN: 1936-0851

Nanoscale membrane curvature is a common feature in cell biology required for functions such as endocytosis, exocytosis and cell migration. These processes require the cytoskeleton to exert forces on the membrane to deform it. Cytosolic proteins contain specific motifs which bind to the membrane, connecting it to the internal cytoskeletal machinery. These motifs often bind charged phosphatidylinositol phosphate lipids present in the cell membrane which play significant roles in signaling. These lipids are important for membrane deforming processes, such as endocytosis, but much remains unknown about their role in the sensing of membrane nanocurvature by protein domains. Using coarse-grained molecular dynamics simulations, we investigated the interaction of a model curvature active protein domain, the epsin N-terminal homology domain (ENTH), with curved lipid membranes. The combination of anionic lipids (phosphatidylinositol 4,5-bisphosphate and phosphatidylserine) within the membrane, protein backbone flexibility, and structural changes within the domain were found to affect the domain's ability to sense, bind, and localize with nanoscale precision at curved membrane regions. The findings suggest that the ENTH domain can sense membrane curvature without the presence of its terminal amphipathic α helix <i>via</i> another structural region we have denoted as H3, re-emphasizing the critical relationship between nanoscale membrane curvature and protein function.

Journal article

J C, Najer A, Blakney A, McKay P, Bellahcene M, Winter C, Sintou A, Tang J, Keane TJ, Schneider M, Shattock R, Sattler S, Stevens Met al., 2020, Neutrophils enable local and non-invasive liposome delivery to inflamed skeletal muscle and ischemic heart, Advanced Materials, Vol: 32, Pages: 1-10, ISSN: 0935-9648

Uncontrolled inflammation is a major pathological factor underlying a range of diseases including autoimmune conditions, cardiovascular disease, and cancer. Improving localized delivery of immunosuppressive drugs to inflamed tissue in a non‐invasive manner offers significant promise to reduce severe side effects caused by systemic administration. Here, a neutrophil‐mediated delivery system able to transport drug‐loaded nanocarriers to inflamed tissue by exploiting the inherent ability of neutrophils to migrate to inflammatory tissue is reported. This hybrid system (neutrophils loaded with liposomes ex vivo) efficiently migrates in vitro following an inflammatory chemokine gradient. Furthermore, the triggered release of loaded liposomes and reuptake by target macrophages is studied. The migratory behavior of liposome‐loaded neutrophils is confirmed in vivo by demonstrating the delivery of drug‐loaded liposomes to an inflamed skeletal muscle in mice. A single low‐dose injection of the hybrid system locally reduces inflammatory cytokine levels. Biodistribution of liposome‐loaded neutrophils in a human‐disease‐relevant myocardial ischemia reperfusion injury mouse model after i.v. injection confirms the ability of injected neutrophils to carry loaded liposomes to inflammation sites. This strategy shows the potential of nanocarrier‐loaded neutrophils as a universal platform to deliver anti‐inflammatory drugs to promote tissue regeneration in inflammatory diseases.

Journal article

Armstrong JPK, Keane TJ, Roques AC, Stephen Patrick P, Mooney CM, Kuan W-L, Pisupati V, Oreffo ROC, Stuckey D, Watt FM, Forbes SJ, Barker RA, Stevens Met al., 2020, A blueprint for translational regenerative medicine, Science Translational Medicine, Vol: 12, ISSN: 1946-6234

The last few decades have produced a large number of proof-of-concept studies in regenerative medicine.However, the route to clinical adoption is fraught with technical and translational obstacles that frequentlyconsign promising academic solutions to the so-called “valley of death.” This review is intended to serve as ablueprint for translational regenerative medicine: we suggest principles to help guide cell and materialselection, present key in vivo imaging modalities and argue that the host immune response should beconsidered throughout therapeutic development. Finally, we suggest a pathway to navigate the oftencomplex regulatory and manufacturing landscape of translational regenerative medicine.

Journal article

Hogset H, Horgan C, Bergholt M, Armstrong J, Torraca V, Chen Q, Keane TJ, Bugeon L, Dallman M, Mostowy S, Stevens Met al., 2020, In vivo biomolecular imaging of zebrafish embryos using confocal Raman spectroscopy, Nature Communications, Vol: 11, Pages: 1-12, ISSN: 2041-1723

Zebrafish embryos provide a unique opportunity to visualize complex biological processes, yet conventional imaging modalities are unable to access intricate biomolecular information without compromising the integrity of the embryos. Here, we report the use of confocal Raman spectroscopic imaging for the visualization and multivariate analysis of biomolecular information extracted from unlabeled zebrafish embryos. We outline broad applications of this method in: (i) visualizing the biomolecular distribution of whole embryos in three dimensions, (ii) resolving anatomical features at subcellular spatial resolution, (iii) biomolecular profiling and discrimination of wild type and ΔRD1 mutant Mycobacterium marinum strains in a zebrafish embryo model of tuberculosis and (iv) in vivotemporal monitoring of the wound response in living zebrafish embryos.Overall, this study demonstrates the application of confocal Raman spectroscopic imaging for the comparative bimolecular analysis in fully intact and living zebrafish embryos.

Journal article

Finbloom JA, Sousa F, Stevens MM, Desai TAet al., 2020, Engineering the drug carrier biointerface to overcome biological barriers to drug delivery, Advanced Drug Delivery Reviews, Vol: 167, Pages: 89-108, ISSN: 0169-409X

Micro and nanoscale drug carriers must navigate through a plethora of dynamic biological systems prior to reaching their tissue or disease targets. The biological obstacles to drug delivery come in many forms and include tissue barriers, mucus and bacterial biofilm hydrogels, the immune system, and cellular uptake and intracellular trafficking. The biointerface of drug carriers influences how these carriers navigate and overcome biological barriers for successful drug delivery. In this review, we examine how key material design parameters lead to dynamic biointerfaces and improved drug delivery across biological barriers. We provide a brief overview of approaches used to engineer key physicochemical properties of drug carriers, such as morphology, surface chemistry, and topography, as well as the development of dynamic responsive materials for barrier navigation. We then discuss essential biological barriers and how biointerface engineering can enable drug carriers to better navigate and overcome these barriers to drug delivery.

Journal article

Higgins S, Lo Fiego A, Patrick I, Creamer A, Stevens Met al., 2020, Organic bioelectronics: using highly conjugated polymers to interface with biomolecules, cells and tissues in the human body, Advanced Materials Technologies, Vol: 5, Pages: 1-35, ISSN: 2365-709X

Conjugated polymers exhibit interesting material and optoelectronic properties that makethem well-suited to the development of biointerfaces. Their biologically relevant mechanicalcharacteristics, ability to be chemically modified, and mixed electronic and ionic chargetransport are captured within the diverse field of organic bioelectronics. Conjugated polymershave been used in wide range of device architectures, and cell and tissue scaffolds. Thesedevices enable biosensing of many biomolecules, such as metabolites, nucleic acids and more.Devices can be used to both stimulate and sense the behavior of cells and tissues. Similarly,tissue interfaces permit interaction with complex organs, aiding both fundamental biologicalunderstanding and providing new opportunities for stimulating regenerative behaviors andbioelectronic based therapeutics. Applications of these materials are broad, and muchcontinues to be uncovered about their fundamental properties. This report covers the currentunderstanding of the fundamentals of conjugated polymer biointerfaces and their interactionswith biomolecules, cells and tissues in the human body. An overview of current materials anddevices is presented, along with highlighted major in vivo and in vitro applications. Finally,open research questions and opportunities are discussed.

Journal article

Nele V, Wojciechowski J, Armstrong J, Stevens Met al., 2020, Tailoring gelation mechanisms for advanced hydrogel applications, Advanced Functional Materials, Vol: 30, Pages: 1-22, ISSN: 1616-301X

Hydrogels are one of the most commonly explored classes of biomaterials. Their chemical and structural versatility has enabled their use across a wide range of applications, including tissue engineering, drug delivery, and cell culture. Hydrogels form upon a sol–gel transition, which can be elicited by different triggers designed to enable precise control over hydrogelation kinetics and hydrogel structure. The chosen hydrogelation trigger and chemistry can have a profound effect on the success of the targeted application. In this Progress Report, a critical overview of recent advances in hydrogel design is presented, with a focus on the available strategies used to trigger the formation of hydrogel networks (e.g., temperature, light, ultrasound). These triggers are presented within a new classification system, and their suitability for six key hydrogel‐based applications is assessed. This Progress Report is intended to guide trigger selection for new hydrogel applications and inspire the rational design of new hydrogelation trigger mechanisms.

Journal article

Ritzau-Reid K, Spicer C, Gelmi A, Grigsby CL, Ponder Jr J, Bemmer V, Creamer A, Vilar R, Serio A, Stevens Met al., 2020, An electroactive oligo-EDOT platform for neural tissue engineering, Advanced Functional Materials, Vol: 30, Pages: 1-11, ISSN: 1616-301X

The unique electrochemical properties of the conductive polymer poly(3,4-ethylenedioxythiophene):polystyrene sulfonate (PEDOT:PSS) make it an attractive material for use in neural tissue engineering applications. However, inadequate mechanical properties, and difficulties in processing and lack of biodegradability have hindered progress in this field. Here, we have improved the functionality of PEDOT:PSS for neural tissue engineering by incorporating 3,4-ethylenedioxythiophene (EDOT) oligomers, synthesised using a novel end-capping strategy, into block co-polymers. By exploiting end-functionalised oligoEDOT constructs as macroinitiators for the polymerization of poly(caprolactone) (PCL), we produce a block co-polymer that is electroactive, processable, and bio-compatible. By combining these properties, we were able to produce electroactive fibrous mats for neuronal culture via solution electrospinning and melt electrospinning writing (MEW). Importantly, we also show that neurite length and branching of neural stem cells can be enhanced on our materials under electrical stimulation, demonstrating the promise of these scaffolds for neural tissue engineering.

Journal article

Zwi Dantsis L, Winter CW, Kauscher U, Ferrini A, Wang B, Whittaker T, Hood SR, Terracciano C, Stevens Met al., 2020, Highly purified extracellular vesicles from human cardiomyocytes demonstrate preferential uptake by human endothelial cells, Nanoscale, Vol: 12, Pages: 19844-19854, ISSN: 2040-3364

Extracellular vesicles (EVs) represent a promising cell-free alternative for treatment of cardiovascular diseases. Nevertheless, the lack of standardised and reproducible isolation methods capable of recovering pure, intact EVs presents a significant obstacle. Additionally, there is significant interest in investigating the interactions of EVs with different cardiac cell types. Here we established a robust technique for the production and isolation of EVs harvested from an enriched (>97% purity) population of human induced pluripotent stem cell (iPSC)-derived cardiomyocytes (CMs) with size exclusion chromatography. Utilizing an advanced fluorescence labelling strategy, we then investigated the interplay of the CM-EVs with the three major cellular components of the myocardium (fibroblasts, cardiomyocytes and endothelial cells) and identified that cardiac endothelial cells show preferential uptake of these EVs. Overall, our findings provide a great opportunity to overcome the translational hurdles associated with the isolation of intact, non-aggregated human iPSC-CM EVs at high purity. Furthermore, understanding in detail the interaction of the secreted EVs with their surrounding cells in the heart may open promising new avenues in the field of EV engineering for targeted delivery in cardiac regeneration.

Journal article

Majid QA, Fricker ATR, Gregory DA, Davidenko N, Hernandez Cruz O, Jabbour RJ, Owen TJ, Basnett P, Lukasiewicz B, Stevens M, Best S, Cameron R, Sinha S, Harding SE, Roy Iet al., 2020, Natural biomaterials for cardiac tissue engineering: a highly biocompatible solution., Frontiers in Cardiovascular Medicine, Vol: 7, Pages: 1-32, ISSN: 2297-055X

Cardiovascular diseases (CVD) constitute a major fraction of the current major global diseases and lead to about 30% of the deaths, i.e., 17.9 million deaths per year. CVD include coronary artery disease (CAD), myocardial infarction (MI), arrhythmias, heart failure, heart valve diseases, congenital heart disease, and cardiomyopathy. Cardiac Tissue Engineering (CTE) aims to address these conditions, the overall goal being the efficient regeneration of diseased cardiac tissue using an ideal combination of biomaterials and cells. Various cells have thus far been utilized in pre-clinical studies for CTE. These include adult stem cell populations (mesenchymal stem cells) and pluripotent stem cells (including autologous human induced pluripotent stem cells or allogenic human embryonic stem cells) with the latter undergoing differentiation to form functional cardiac cells. The ideal biomaterial for cardiac tissue engineering needs to have suitable material properties with the ability to support efficient attachment, growth, and differentiation of the cardiac cells, leading to the formation of functional cardiac tissue. In this review, we have focused on the use of biomaterials of natural origin for CTE. Natural biomaterials are generally known to be highly biocompatible and in addition are sustainable in nature. We have focused on those that have been widely explored in CTE and describe the original work and the current state of art. These include fibrinogen (in the context of Engineered Heart Tissue, EHT), collagen, alginate, silk, and Polyhydroxyalkanoates (PHAs). Amongst these, fibrinogen, collagen, alginate, and silk are isolated from natural sources whereas PHAs are produced via bacterial fermentation. Overall, these biomaterials have proven to be highly promising, displaying robust biocompatibility and, when combined with cells, an ability to enhance post-MI cardiac function in pre-clinical models. As such, CTE has great potential for future clinical solutions and he

Journal article

Ouyang L, Armstrong J, Lin Y, Wojciechowski J, Lee-Reeves C, Hachim D, Zhou K, Burdick JA, Stevens Met al., 2020, Expanding and optimizing 3D bioprinting capabilities using complementary network bioinks, Science Advances, Vol: 6, Pages: 1-13, ISSN: 2375-2548

A major challenge in 3D bioprinting is the limited number of bioinks that fulfill the physiochemical requirements of printing, while also providing a desirable environment for encapsulated cells.Here, we address this limitation by temporarily stabilizing bioinks with a complementary thermo-reversible gelatin network. This strategy enablesthe effective printing of biomaterials that would typically not meet printing requirements, with instrument parameters and structural output largely independent of the base biomaterial. This approach is demonstrated across a library of photo-crosslinkable bioinks derived from natural and synthetic polymers, including gelatin, hyaluronic acid, chondroitin sulfate, dextran, alginate, chitosan, heparin,and poly(ethylene glycol). A range of complex and heterogeneous structures are printed, including soft hydrogel constructs supporting the 3D culture of astrocytes. This highly generalizable methodology expands the palette of available bioinks, allowing the biofabrication of constructs optimized to meet the biological requirements of cell culture and tissue engineering.

Journal article

Massi L, Najer A, Chapman R, Spicer C, Nele V, Che J, Booth MA, Doutch JJ, Stevens Met al., 2020, Tuneable peptide cross-linked nanogels for enzyme-triggered protein delivery, Journal of Materials Chemistry B, Vol: 8, Pages: 8894-8907, ISSN: 2050-750X

Many diseases are associated with the dysregulated activity of enzymes, such as matrixmetalloproteinases (MMPs). This dysregulation can be leveraged in drug delivery to achieve disease- orsite-specific cargo release. Self-assembled polymeric nanoparticles are versatile drug carrier materialsdue to the accessible diversity of polymer chemistry. However, efficient loading of sensitive cargo, suchas proteins, and introducing functional enzyme-responsive behaviour remain challenging. Herein,peptide-crosslinked, temperature-sensitive nanogels for protein delivery were designed to respond toMMP-7, which is overexpressed in many pathologies including cancer and inflammatory diseases. Theincorporation of N-cyclopropylacrylamide (NCPAM) into N-isopropylacrylamide (NIPAM)-basedcopolymers enabled us to tune the polymer lower critical solution temperature from 33 to 44 1C,allowing the encapsulation of protein cargo and nanogel-crosslinking at slightly elevated temperatures.This approach resulted in nanogels that were held together by MMP-sensitive peptides for enzymespecificprotein delivery. We employed a combination of cryogenic transmission electron microscopy(cryo-TEM), dynamic light scattering (DLS), small angle neutron scattering (SANS), and fluorescencecorrelation spectroscopy (FCS) to precisely decipher the morphology, self-assembly mechanism,enzyme-responsiveness, and model protein loading/release properties of our nanogel platform. Simplevariation of the peptide linker sequence and combining multiple different crosslinkers will enable us toadjust our platform to target specific diseases in the future.

Journal article

Whittaker T, Nagelkerke A, Nele V, Kauscher U, Stevens Met al., 2020, Experimental artefacts can lead to misattribution of bioactivity from soluble mesenchymal stem cell paracrine factors to extracellular vesicles, Journal of Extracellular Vesicles, Vol: 9, ISSN: 2001-3078

It has been demonstrated that some commonly used Extracellular Vesicle (EV) isolation techniques can lead to substantial contamination with non-EV factors. Whilst it has been established that this impacts the identification of biomarkers, the impact on apparent EV bioactivity has not been explored. Extracellular vesicles have been implicated as critical mediators of therapeutic human mesenchymal stem cell (hMSC) paracrine signalling. Isolated hMSC-EVs have been used to treat multiple in vitro and in vivo models of tissue damage. However, the relative contributions of EVs and non-EV factors have not been directly compared. The dependence of hMSC paracrine signalling on EVs was first established by ultrafiltration of hMSC-conditioned medium to deplete EVs, which led to a loss of signalling activity. Here, we show that this method also causes depletion of non-EV factors, and that when this is prevented proangiogenic signalling activity is fully restored in vitro. Subsequently, we used size-exclusion chromatography (SEC) to separate EVs and soluble proteins to directly and quantitatively compare their relative contributions to signalling. Non-EV factors were found to be necessary and sufficient for the stimulation of angiogenesis and wound healing in vitro. EVs in isolation were found to be capable of potentiating signalling only when isolated by a low-purity method, or when used at comparatively high concentrations. These results indicate a potential for contaminating soluble factors to artefactually increase the apparent bioactivity of EV isolates and could have implications for future studies on the biological roles of EVs.

Journal article

Li S, Tallia F, Mohammed AA, Stevens MM, Jones JRet al., 2020, Scaffold channel size influences stem cell differentiation pathway in 3-D printed silica hybrid scaffolds for cartilage regeneration, Biomaterials Science, Vol: 8, Pages: 4458-4466, ISSN: 2047-4830

We report that 3-D printed scaffold channel size can direct bone marrow derived stem cell differentiation. Treatment of articular cartilage trauma injuries, such as microfracture surgery, have limited success because durability is limited as fibrocartilage forms. A scaffold-assisted approach, combining microfracture with biomaterials has potential if the scaffold can promote articular cartilage production and share load with cartilage. Here, we investigated human bone marrow derived stromal cell (hBMSC) differentiation in vitro in 3-D printed silica/poly(tetrahydrofuran)/poly(ε-caprolactone) hybrid scaffolds with specific channel sizes. Channel widths of ∼230 μm (210 ± 22 μm mean strut size, 42.4 ± 3.9% porosity) provoked hBMSC differentiation down a chondrogenic path, with collagen Type II matrix prevalent, indicative of hyaline cartilage. When pores were larger (∼500 μm, 229 ± 29 μm mean strut size, 63.8 ± 1.6% porosity) collagen Type I was dominant, indicating fibrocartilage. There was less matrix and voids in smaller channels (∼100 μm, 218 ± 28 μm mean strut size, 31.2 ± 2.9% porosity). Our findings suggest that a 200–250 μm pore channel width, in combination with the surface chemistry and stiffness of the scaffold, is optimal for cell–cell interactions to promote chondrogenic differentiation and enable the chondrocytes to maintain their phenotype.

Journal article

Gurnani P, Blakney AK, Yeow J, Bouton CR, Shattock RJ, Stevens MM, Alexander Cet al., 2020, An improved synthesis of poly(amidoamine)s for complexation with self-amplifying RNA and effective transfection, Polymer Chemistry, Vol: 11, Pages: 5861-5869, ISSN: 1759-9954

Cationic polymers are widely used as materials to condense nucleic acids for gene-based therapies. These have been developed to mainly deliver DNA and RNA for cancer therapies but the ongoing COVID-19 pandemic has demonstrated an urgent need for new DNA and RNA vaccines. Given this, suitable manufacturing conditions for such cationic polymers which can protect the nucleic acid in the formulation and delivery stages but release the cargo in the correct cellular compartment effectively and safely are required. A number of polymers based on poly(amidoamine)s fit these criteria but their syntheses can be time-consuming, inefficient and poorly reproducible, precluding their adoption as manufacturable vaccine excipients. Here we report an improved synthesis of poly(cystamine bisacrylamide-co-4-amino-1-butanol), abbreviated as pABOL, via modifications in concentration, reaction time and reaction conditions. Optimisation of monomer contents and stoichiometries, solvents, diluents and temperature, combined with the application of microwaves, enabled the preparation of vaccine candidate pABOL materials in 4 h compared to 48 h reported for previous syntheses. These procedures were highly reproducible in multiple repeat syntheses. Transfection experiments with a model RNA showed that polymers of formulation with appropriate molar masses and mass distributions were as effective in model cell lines as polymers derived from the unoptimised syntheses which have been shown to have high efficacy as RNA vaccine formulation candidates.

Journal article

Budd J, Miller BS, Manning EM, Lampos V, Zhuang M, Edelstein M, Rees G, Emery VC, Stevens MM, Keegan N, Short MJ, Pillay D, Manley E, Cox IJ, Heymann D, Johnson AM, McKendry RAet al., 2020, Digital technologies in the public-health response to COVID-19, Nature Medicine, Vol: 26, Pages: 1183-1192, ISSN: 1078-8956

Digital technologies are being harnessed to support the public-health response to COVID-19 worldwide, including population surveillance, case identification, contact tracing and evaluation of interventions on the basis of mobility data and communication with the public. These rapid responses leverage billions of mobile phones, large online datasets, connected devices, relatively low-cost computing resources and advances in machine learning and natural language processing. This Review aims to capture the breadth of digital innovations for the public-health response to COVID-19 worldwide and their limitations, and barriers to their implementation, including legal, ethical and privacy barriers, as well as organizational and workforce barriers. The future of public health is likely to become increasingly digital, and we review the need for the alignment of international strategies for the regulation, evaluation and use of digital technologies to strengthen pandemic management, and future preparedness for COVID-19 and other infectious diseases.

Journal article

Ahadian S, Finbloom JA, Mofidfar M, Emir Diltemiz S, Nasrollahi F, Davoodi E, Hosseini V, Mylonaki I, Sangabathuni S, Montazerian H, Fetah K, Nasiri R, Remzi Dokmeci M, Stevens M, Desai TA, Khademhosseini Aet al., 2020, Micro and nanoscale technologies in oral drug delivery, Advanced Drug Delivery Reviews, Vol: 157, Pages: 37-62, ISSN: 0169-409X

Oral administration is a pillar of the pharmaceutical industry and yet it remains challenging to administer hydrophilic therapeutics by the oral route. Smart and controlled oral drug delivery could bypass the physiological barriers that limit the oral delivery of these therapeutics. Micro- and nanoscale technologies, with an unprecedented ability to create, control, and measure micro- or nanoenvironments, have found tremendous applications in biology and medicine. In particular, significant advances have been made in using these technologies for oral drug delivery. In this review, we briefly describe biological barriers to oral drug delivery and micro and nanoscale fabrication technologies. Micro and nanoscale drug carriers fabricated using these technologies, including bioadhesives, microparticles, micropatches, and nanoparticles, are described. Other applications of micro and nanoscale technologies are discussed, including the fabrication of devices and tissue engineering models to precisely control or assess oral drug delivery in vivo and in vitro, respectively. Strategies to advance translation of micro and nanotechnologies into clinical trials for oral drug delivery are mentioned. Finally, challenges and future prospects on further integration of micro and nanoscale technologies with oral drug delivery systems are highlighted.

Journal article

Ouyang L, Armstrong J, Salmeron-Sanchez M, Stevens Met al., 2020, Assembly of living building blocks to engineer complex tissues, Advanced Functional Materials, Vol: 30, Pages: 1-22, ISSN: 1616-301X

The great demand for tissue and organ grafts, compounded by an aging demographic and a shortage of available donors, has driven the development of bioengineering approaches that can generate biomimetic tissues in vitro. Despite the considerable progress in conventional scaffold‐based tissue engineering, the recreation of physiological complexity has remained a challenge. Bottom‐up tissue engineering strategies have opened up a new avenue for the modular assembly of living building blocks into customized tissue architectures. This Progress Report overviews the recent progress and trends in the fabrication and assembly of living building blocks, with a key highlight on emerging bioprinting technologies that can be used for modular assembly and complexity in tissue engineering. By summarizing the work to date, providing new classifications of different living building blocks, highlighting state‐of‐the‐art research and trends, and offering personal perspectives on future opportunities, this Progress Report aims to aid and inspire other researchers working in the field of modular tissue engineering.

Journal article

Watts C, Hanham S, Armstrong J, Ahmad M, Stevens M, Klein Net al., 2020, Microwave dielectric sensing of free-flowing, single, living cells in aqueous suspension, IEEE Journal of Electromagnetics, RF and Microwaves in Medicine and Biology, Vol: 4, Pages: 97-208, ISSN: 2469-7249

Dielectric measurements offer the possibility of highly sensitive detection of physical cell properties, and are of interest for clinical applications due to their non-destructive nature and the lack of need for cell labelling. Here we report sensitive measurements on single, living, free-flowing cells (not electrostatically or dielectrophoretically trapped, cultured or fixed directly on sensing elements) in aqueous medium at ~9.8 GHz taken using a coupled dielectric-split ring resonator assembly. Inductive coupling between the two resonators enabled separation of microfluidic chips from RF connectors and allowed for time-resolved continuous-wave measurements on flowing single cells via the coaxial ports of a dielectric-loaded microwave cavity. Analysis via an equivalent circuit model showed that the novel resonator assembly maintained the permittivity-dependent sensitivity of a split ring resonator while operating at quality factors >1000 with lossy aqueous media (typically ~1900). Using a microfluidic channel with a 300 x 300 μm cross section, at a water-loaded resonant amplitude of ~-22 dB at 0 dBm input power level, shifts in amplitude due to individual cells passing through the sensing region of up to -0.0015 dB were observed. Correlations between averaged amplitude shifts and cell size as well as material properties demonstrate the diagnostic potential of this technique.

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: limit=30&id=00398347&person=true&page=4&respub-action=search.html