Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 103
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Lab Chip ; 24(3): 537-548, 2024 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-38168806

RESUMEN

The human body is made up of approximately 40 trillion cells in close contact, with the cellular density of individual tissues varying from 1 million to 1 billion cells per cubic centimetre. Interactions between different cell types (termed heterotypic) are thus common in vivo. Communication between cells can take the form of direct cell-cell contact mediated by plasma membrane proteins or through paracrine signalling mediated through the release, diffusion, and receipt of soluble factors. There is currently no systematic method to investigate the relative contributions of these mechanisms to cell behaviour. In this paper, we detail the conception, development and validation of a microfluidic device that allows cell-cell contact and paracrine signalling in defined areas and over a variety of biologically relevant length scales, referred to as the interactome-device or 'I-device'. Importantly, by intrinsic device design features, cells in different regions in the device are exposed to four different interaction types, including a) no heterotypic cell interaction, b) only paracrine signalling, c) only cell-cell direct contact, or d) both forms of interaction (paracrine and cell-cell direct contact) together. The device design was validated by both mathematical modelling and experiments. Perfused stem cell culture over the medium term and the formation of direct contact between cells in the culture chambers was confirmed. The I-device offers significant flexibility, being able to be applied to any combination of adherent cells to determine the relative contributions of different communication mechanisms to cellular outcomes.


Asunto(s)
Comunicación Celular , Técnicas de Cultivo de Célula , Humanos , Técnicas de Cocultivo , Comunicación Paracrina , Dispositivos Laboratorio en un Chip
2.
EMBO J ; 42(13): e112095, 2023 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-37226896

RESUMEN

The unique nerve terminal targeting of botulinum neurotoxin type A (BoNT/A) is due to its capacity to bind two receptors on the neuronal plasma membrane: polysialoganglioside (PSG) and synaptic vesicle glycoprotein 2 (SV2). Whether and how PSGs and SV2 may coordinate other proteins for BoNT/A recruitment and internalization remains unknown. Here, we demonstrate that the targeted endocytosis of BoNT/A into synaptic vesicles (SVs) requires a tripartite surface nanocluster. Live-cell super-resolution imaging and electron microscopy of catalytically inactivated BoNT/A wildtype and receptor-binding-deficient mutants in cultured hippocampal neurons demonstrated that BoNT/A must bind coincidentally to a PSG and SV2 to target synaptic vesicles. We reveal that BoNT/A simultaneously interacts with a preassembled PSG-synaptotagmin-1 (Syt1) complex and SV2 on the neuronal plasma membrane, facilitating Syt1-SV2 nanoclustering that controls endocytic sorting of the toxin into synaptic vesicles. Syt1 CRISPRi knockdown suppressed BoNT/A- and BoNT/E-induced neurointoxication as quantified by SNAP-25 cleavage, suggesting that this tripartite nanocluster may be a unifying entry point for selected botulinum neurotoxins that hijack this for synaptic vesicle targeting.


Asunto(s)
Toxinas Botulínicas Tipo A , Toxinas Botulínicas Tipo A/metabolismo , Membrana Celular/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Vesículas Sinápticas/metabolismo , Animales , Ratas
3.
APL Bioeng ; 6(3): 030402, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35996390
4.
Sci Rep ; 11(1): 2462, 2021 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-33510250

RESUMEN

Biological computation requires in vivo control of molecular behavior to progress development of autonomous devices. miRNA switches represent excellent, easily engineerable synthetic biology tools to achieve user-defined gene regulation. Here we present the construction of a synthetic network to implement detoxification functionality. We employed a modular design strategy by engineering toxin-induced control of an enzyme scavenger. Our miRNA switch results show moderate synthetic expression control over a biologically active detoxification enzyme molecule, using an established design protocol. However, following a new design approach, we demonstrated an evolutionarily designed miRNA switch to more effectively activate enzyme activity than synthetically designed versions, allowing markedly improved extrinsic user-defined control with a toxin as inducer. Our straightforward new design approach is simple to implement and uses easily accessible web-based databases and prediction tools. The ability to exert control of toxicity demonstrates potential for modular detoxification systems that provide a pathway to new therapeutic and biocomputing applications.


Asunto(s)
Enzimas/metabolismo , MicroARNs/genética , Biosíntesis de Proteínas/genética , Toxinas Biológicas/toxicidad , Secuencia de Bases , Citocromo P-450 CYP1A2/metabolismo , Activación Enzimática/efectos de los fármacos , Silenciador del Gen , Células HEK293 , Humanos , MicroARNs/química , MicroARNs/metabolismo , Conformación de Ácido Nucleico , Teofilina/farmacología , Factores de Tiempo
5.
J Tissue Eng ; 11: 2041731420954712, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33178409

RESUMEN

Using microspherical scaffolds as building blocks to repair bone defects of specific size and shape has been proposed as a tissue engineering strategy. Here, phosphate glass (PG) microcarriers doped with 5 mol % TiO2 and either 0 mol % CoO (CoO 0%) or 2 mol % CoO (CoO 2%) were investigated for their ability to support osteogenic and vascular responses of human mesenchymal stem cells (hMSCs). Together with standard culture techniques, cell-material interactions were studied using a novel perfusion microfluidic bioreactor that enabled cell culture on microspheres, along with automated processing and screening of culture variables. While titanium doping was found to support hMSCs expansion and differentiation, as well as endothelial cell-derived vessel formation, additional doping with cobalt did not improve the functionality of the microspheres. Furthermore, the microfluidic bioreactor enabled screening of culture parameters for cell culture on microspheres that could be potentially translated to a scaled-up system for tissue-engineered bone manufacturing.

6.
Biomater Sci ; 8(20): 5677-5689, 2020 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-32915185

RESUMEN

Mimicking the complex hierarchical architecture of the 'osteon', the functional unit of cortical bone, from the bottom-up offers the possibility of generating mature bone tissue in tissue engineered bone substitutes. In this work, a modular 'bottom-up' approach has been developed to assemble bone niche-mimicking nanocomposite scaffolds composed of aligned electrospun nanofibers of poly(lactic-co-glycolic acid) (PLGA) encapsulating aligned rod-shape nano-sized hydroxyapatite (nHA). By encoding axial orientation of the nHA within these aligned nanocomposite fibers, significant improvements in mechanical properties, surface roughness, hydrophilicity and in vitro simulated body fluid (SBF) mineral deposition were achieved. Moreover, these hierarchical scaffolds induced robust formation of bone hydroxyapatite and osteoblastic maturation of human bone marrow-derived mesenchymal stem cells (hBMSCs) in growth media that was absent of any soluble osteogenic differentiation factors. The results of this investigation confirm that these tailored, aligned nanocomposite fibers, in the absence of media-bone inductive factors, offer the requisite biophysical and biochemical cues to hBMSCs to promote and support their differentiation into mature osteoblast cells and form early bone-like tissue in vitro.


Asunto(s)
Durapatita , Células Madre Mesenquimatosas , Biomimética , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Señales (Psicología) , Glicoles , Humanos , Ácido Láctico , Osteogénesis , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Ingeniería de Tejidos , Andamios del Tejido
7.
Sci Adv ; 6(2): eaaw2746, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31934619

RESUMEN

Creating complex multicellular kidney organoids from pluripotent stem cells shows great promise. Further improvements in differentiation outcomes, patterning, and maturation of specific cell types are, however, intrinsically limited by standard tissue culture approaches. We describe a novel full factorial microbioreactor array-based methodology to achieve rapid interrogation and optimization of this complex multicellular differentiation process in a facile manner. We successfully recapitulate early kidney tissue patterning events, exploring more than 1000 unique conditions in an unbiased and quantitative manner, and define new media combinations that achieve near-pure renal cell type specification. Single-cell resolution identification of distinct renal cell types within multilayered kidney organoids, coupled with multivariate analysis, defined the definitive roles of Wnt, fibroblast growth factor, and bone morphogenetic protein signaling in their specification, exposed retinoic acid as a minimal effector of nephron patterning, and highlighted critical contributions of induced paracrine factors on cell specification and patterning.


Asunto(s)
Riñón/crecimiento & desarrollo , Organoides/crecimiento & desarrollo , Comunicación Paracrina , Perfusión , Células Madre Pluripotentes/citología , Reactores Biológicos , Humanos , Riñón/efectos de los fármacos , Análisis Multivariante , Nefronas/citología , Nefronas/efectos de los fármacos , Organoides/efectos de los fármacos , Comunicación Paracrina/efectos de los fármacos , Células Madre Pluripotentes/efectos de los fármacos , Células Madre Pluripotentes/metabolismo , Tretinoina/farmacología , Vía de Señalización Wnt/efectos de los fármacos
8.
ACS Nano ; 13(10): 11129-11143, 2019 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-31580055

RESUMEN

The ability of mesenchymal stem cells to sense nanoscale variations in extracellular matrix (ECM) compositions in their local microenvironment is crucial to their survival and their fate; however, the underlying molecular mechanisms defining how such fates are temporally modulated remain poorly understood. In this work, we have utilized self-assembled block copolymer surfaces to present nanodomains of an adhesive peptide found in many ECM proteins at different lateral spacings (from 30 to 60 nm) and studied the temporal response (2 h to 14 days) of human mesenchymal stem cells (hMSCs) using a panel of real-time localization and activity biosensors. Our findings revealed that within the first 4 to 24 h postadhesion and spreading, hMSCs on smaller nanodomain spacings recruit more activated FAK and Src proteins to produce larger, longer-lived, and increased numbers of focal adhesions (FAs). The adhesions formed on smaller nanospacings rapidly recruit higher amounts of nonmuscle myosin IIA and vinculin and experience tension forces (by >5 pN/FA) significantly higher than those observed on larger nanodomain spacings. The transmission of higher levels of tension into the cytoskeleton at short times was accompanied by higher Rac1, cytosolic ß-catenin, and nuclear localization of YAP/TAZ and RUNX2, which together biased the commitment of hMSCs to an osteogenic fate. This investigation provides mechanistic insights to confirm that smaller lateral spacings of adhesive nanodomains alter hMSC mechanosensing and biases mechanotransduction at short times via differential coupling of FAK/Src/Rac1/myosin IIA/YAP/TAZ signaling pathways to support longer-term changes in stem cell differentiation and state.


Asunto(s)
Adipogénesis/genética , Linaje de la Célula/genética , Células Madre Mesenquimatosas/efectos de los fármacos , Osteogénesis/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Adipogénesis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula/efectos de los fármacos , Microambiente Celular/efectos de los fármacos , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Citoesqueleto/efectos de los fármacos , Citoesqueleto/genética , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/genética , Adhesiones Focales/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Humanos , Células Madre Mesenquimatosas/citología , Osteogénesis/efectos de los fármacos , Polímeros/química , Polímeros/farmacología , Transducción de Señal/efectos de los fármacos , Factores de Transcripción/genética , Proteínas Señalizadoras YAP , beta Catenina/genética , Proteína de Unión al GTP rac1/genética
9.
Biomaterials ; 223: 119430, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31493696

RESUMEN

Cells are capable of sensing the differences in elastic and viscous properties (i.e., the 'viscoelasticity') of their tissue microenvironment and responding accordingly by changing their transcriptional activity and modifying their behaviors. When designing viscoelastic materials to mimic the mechanical properties of native tissue niches, it is important to consider the timescales over which cells probe their microenvironment, as the response of a viscoelastic material to an imposed stress or strain is timescale dependent. Although the timescale of cellular mechano-sensing is currently unknown, hydrogel substrates with tunable viscoelastic spectra can allow one to probe the cellular response to timescale dependent mechanical properties. Here, we report on a cytocompatible and viscoelastic hydrogel culture system with reversible boronate ester cross-links, formed from pendant boronic acid and vicinal diol moieties, where the equilibrium kinetics of esterification were leveraged to tune the viscoelastic spectrum. We found that viscoelasticity increased as a function of the boronic acid and vicinal diol concentration, and also increased with decreasing cross-linker concentration, where the maximal loss tangent achieved with this system was 0.55 at 0.1 rad s-1. Additionally, we found that the cis-vicinal diols configuration altered the viscoelastic spectra, where a tan δ peak occurred at ~1 rad s-1 for hydrogels functionalized with boronic acid, while an additional peak formed at ≥10 rad s-1 for hydrogels functionalized with both boronic acid and cis-vic-diols. In experiments with NIH-3T3 fibroblasts cultured on these hydrogels, the projected cell area and nuclear area, focal adhesion tension, and subcellular localization of YAP/TAZ were all found to be lower for cells cultured on the viscoelastic hydrogels compared to elastic hydrogels with a similar storage modulus. Despite these differences, there was not a statistically significant relationship between the frequency dependent viscoelastic material properties characterized in this study and cellular morphologies, focal adhesion tension, or the subcellular localization of YAP. While these results demonstrate that mechanotransduction pathways are affected by viscoelasticity, they also suggest that these mechanotransduction pathways are not particularly sensitive to the frequency dependent viscoelastic material properties from 0.1 to 10 rad s-1.


Asunto(s)
Materiales Biocompatibles/química , Ácidos Borónicos/química , Ésteres/química , Hidrogeles/química , Mecanotransducción Celular , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Proteínas de Ciclo Celular/metabolismo , Elasticidad , Transferencia Resonante de Energía de Fluorescencia , Cinética , Ratones , Células 3T3 NIH , Reología , Transactivadores/metabolismo , Viscosidad , Proteínas Señalizadoras YAP
10.
Biomaterials ; 210: 105-115, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31060868

RESUMEN

Human articular cartilage is a complex multi-zonal tissue in which cells displaying three chondrocyte phenotypes (persistent, transient and hypertrophic) are supported and maintained by distinctly different (zonal) combinations of extracellular matrix (ECM) molecules. Articular cartilage has limited regenerative capacity, even though adjacent to the medullary cavity, an easily accessible reservoir of multipotent progenitor cells capable of eliciting repair, (human) mesenchymal stromal/stem cells (hMSCs). A greater understanding of the impacts of the extracellular cues provided in each zone of articular cartilage on hMSCs thus offers the potential to develop new scaffolds that can effect multi-zonal cartilage generation. In this work, we have systematically surveyed combinatorial mixtures of peptide sequences derived from ECM and cell adhesion molecules (CAMs) found to be present in cartilage and bone tissues, at a range of concentrations and ratios, to assess their ability to modulate hMSC fate. We show that directed differentiation of hMSCs towards persistent, transient and hypertrophic chondrogenic phenotypes is possible via the controlled presentation of specific peptide combinations on self-assembled polymeric coatings displaying hexagonally-packed nanodomains. These biomimetic substrates highlight that a high level of spatial and compositional control over biochemical cues is required by hMSCs in order to specify different cellular sub-phenotypes.


Asunto(s)
Cartílago Articular/metabolismo , Diferenciación Celular/efectos de los fármacos , Condrogénesis/efectos de los fármacos , Células Madre Mesenquimatosas/citología , Nanopartículas/química , Péptidos/farmacología , Adhesión Celular/efectos de los fármacos , Recuento de Células , Linaje de la Célula/efectos de los fármacos , Células Cultivadas , Matriz Extracelular/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos , Fenotipo
11.
Biomacromolecules ; 19(3): 721-730, 2018 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-29437383

RESUMEN

Capturing cell-secreted extracellular matrix (ECM) proteins through cooperative binding with high specificity and affinity is an important function of native tissue matrices during both tissue homeostasis and repair. However, while synthetic hydrogels, such as those based on poly(ethylene glycol) (PEG), are often proposed as ideal materials to deliver human mesenchymal stem cells (hMSCs) to sites of injury to enable tissue repair, they do not have this capability-a capability that would enable cells to actively remodel their local extracellular microenvironment and potentially provide the required feedback control for more effective tissue genesis. In this work, we detail a methodology that engenders poly(ethylene glycol) (PEG)-based two-dimensional substrates and three-dimensional porous hydrogels with the ability to capture desired extracellular matrix (ECM) proteins with high specificity. This "encoded" ECM protein capture is achieved by decorating the PEG-based materials with protein binding peptides (PBPs) synthesized to be specific in their binding of fibronectin, laminin, and collagen I, which are not only the most omnipresent ECM proteins in human tissues but, as we confirmed, are also secreted to differing extents by hMSCs under in vitro maintenance conditions. By encapsulating hMSCs into these PBP-functionalized hydrogels, and culturing them in protein-free maintenance media, we demonstrate that these PBPs not only actively recruit targeted ECM proteins as they are secreted from hMSCs but also retain them to much higher levels compared to nonfunctionalized gels. This novel approach thus enables the fabrication of encoded surfaces and hydrogels that capture cell-secreted proteins, with high specificity and affinity, in a programmable manner, ready for applications in many bioengineering applications, including bioactive surface coatings, bioassays, stem cell culture, tissue engineering, and regenerative medicine.


Asunto(s)
Proteínas de la Matriz Extracelular , Hidrogeles/química , Células Madre Mesenquimatosas/metabolismo , Péptidos/química , Polietilenglicoles/química , Proteínas de la Matriz Extracelular/química , Proteínas de la Matriz Extracelular/aislamiento & purificación , Proteínas de la Matriz Extracelular/metabolismo , Humanos , Células Madre Mesenquimatosas/química , Células Madre Mesenquimatosas/citología
12.
Nat Commun ; 9(1): 257, 2018 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-29343687

RESUMEN

Mechanotransduction is a strong driver of mesenchymal stem cell (MSC) fate. In vitro, variations in matrix mechanics invoke changes in MSC proliferation, migration and differentiation. However, when incorporating MSCs within injectable, inherently soft hydrogels, this dominance over MSC response substantially limits our ability to couple the ease of application of hydrogels with efficiently directed MSC differentiation, especially in the case of bone generation. Here, we identify differential miRNA expression in response to varying hydrogel stiffness and RhoA activity. We show that modulation of miR-100-5p and miR-143-3p can be used to bias MSC fate and provide mechanistic insight by demonstrating convergence on mTOR signalling. By modulating these mechanosensitive miRNAs, we can enhance osteogenesis in a soft 3D hydrogel. The outcomes of this study provide new understanding of the mechanisms regulating MSC mechanotransduction and differentiation, but also a novel strategy with which to drive MSC fate and significantly impact MSC-based tissue-engineering applications.


Asunto(s)
Diferenciación Celular/genética , Proliferación Celular/genética , Células Madre Mesenquimatosas/metabolismo , MicroARNs/genética , Células Cultivadas , Regulación de la Expresión Génica , Humanos , Hidrogeles/metabolismo , Mecanotransducción Celular , Células Madre Mesenquimatosas/citología , Microscopía Confocal , Osteogénesis/genética , Transducción de Señal/genética , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Ingeniería de Tejidos/métodos
13.
Nat Protoc ; 12(12): 2590-2622, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29189775

RESUMEN

Our understanding of endocytic pathway dynamics is restricted by the diffraction limit of light microscopy. Although super-resolution techniques can overcome this issue, highly crowded cellular environments, such as nerve terminals, can also dramatically limit the tracking of multiple endocytic vesicles such as synaptic vesicles (SVs), which in turn restricts the analytical dissection of their discrete diffusional and transport states. We recently introduced a pulse-chase technique for subdiffractional tracking of internalized molecules (sdTIM) that allows the visualization of fluorescently tagged molecules trapped in individual signaling endosomes and SVs in presynapses or axons with 30- to 50-nm localization precision. We originally developed this approach for tracking single molecules of botulinum neurotoxin type A, which undergoes activity-dependent internalization and retrograde transport in autophagosomes. This method was then adapted to localize the signaling endosomes containing cholera toxin subunit-B that undergo retrograde transport in axons and to track SVs in the crowded environment of hippocampal presynapses. We describe (i) the construction of a custom-made microfluidic device that enables control over neuronal orientation; (ii) the 3D printing of a perfusion system for sdTIM experiments performed on glass-bottom dishes; (iii) the dissection, culturing and transfection of hippocampal neurons in microfluidic devices; and (iv) guidance on how to perform the pulse-chase experiments and data analysis. In addition, we describe the use of single-molecule-tracking analytical tools to reveal the average and the heterogeneous single-molecule mobility behaviors. We also discuss alternative reagents and equipment that can, in principle, be used for sdTIM experiments and describe how to adapt sdTIM to image nanocluster formation and/or tubulation in early endosomes during sorting events. The procedures described in this protocol take ∼1 week.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Endocitosis , Dispositivos Laboratorio en un Chip , Neuronas/citología , Imagen Óptica/métodos , Animales , Técnicas de Cultivo de Célula/instrumentación , Supervivencia Celular , Células Cultivadas , Diseño de Equipo , Hipocampo/citología , Imagen Óptica/instrumentación , Perfusión/instrumentación , Perfusión/métodos , Impresión Tridimensional , Ratas , Ratas Sprague-Dawley , Transfección/instrumentación , Transfección/métodos
14.
Macromol Rapid Commun ; 38(8)2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28221701

RESUMEN

Advanced polymerization methodologies, such as reversible addition-fragmentation transfer (RAFT), allow unprecedented control over star polymer composition, topology, and functionality. However, using RAFT to produce high throughput (HTP) combinatorial star polymer libraries remains, to date, impracticable due to several technical limitations. Herein, the methodology "rapid one-pot sequential aqueous RAFT" or "rosa-RAFT," in which well-defined homo-, copolymer, and mikto-arm star polymers can be prepared in very low to medium reaction volumes (50 µL to 2 mL) via an "arm-first" approach in air within minutes, is reported. Due to the high conversion of a variety of acrylamide/acrylate monomers achieved during each successive short reaction step (each taking 3 min), the requirement for intermediary purification is avoided, drastically facilitating and accelerating the star synthesis process. The presented methodology enables RAFT to be applied to HTP polymeric bio/nanomaterials discovery pipelines, in which hundreds of complex polymeric formulations can be rapidly produced, screened, and scaled up for assessment in a wide range of applications.


Asunto(s)
Materiales Biocompatibles/síntesis química , Técnicas Químicas Combinatorias/métodos , Polimerizacion , Polímeros/síntesis química , Acrilamida/química , Acrilatos/química , Materiales Biocompatibles/química , Modelos Químicos , Estructura Molecular , Nanoestructuras/química , Polímeros/química , Reproducibilidad de los Resultados
15.
Stem Cells Transl Med ; 6(4): 1178-1190, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28205415

RESUMEN

Cost-effective expansion of human mesenchymal stem/stromal cells (hMSCs) remains a key challenge for their widespread clinical deployment. Fibroblast growth factor-2 (FGF-2) is a key hMSC mitogen often supplemented to increase hMSC growth rates. However, hMSCs also produce endogenous FGF-2, which critically interacts with cell surface heparan sulfate (HS). We assessed the interplay of FGF-2 with a heparan sulfate variant (HS8) engineered to bind FGF-2 and potentiate its activity. Bone marrow-derived hMSCs were screened in perfused microbioreactor arrays (MBAs), showing that HS8 (50 µg/ml) increased hMSC proliferation and cell number after 3 days, with an effect equivalent to FGF-2 (50 ng/ml). In combination, the effects of HS8 and FGF-2 were additive. Differential cell responses, from upstream to downstream culture chambers under constant flow of media in the MBA, provided insights into modulation of FGF-2 transport by HS8. HS8 treatment induced proliferation mainly in the downstream chambers, suggesting a requirement for endogenous FGF-2 accumulation, whereas responses to FGF-2 occurred primarily in the upstream chambers. Adding HS8 along with FGF-2, however, maximized the range of FGF-2 effectiveness. Measurements of FGF-2 in static cultures then revealed that this was because HS8 caused increased endogenous FGF-2 production and liberated FGF-2 from the cell surface into the supernatant. HS8 also sustained levels of supplemented FGF-2 available over 3 days. These results suggest HS8 enhances hMSC proliferation and expansion by leveraging endogenous FGF-2 production and maximizing the effect of supplemented FGF-2. This is an exciting strategy for cost-effective expansion of hMSCs. Stem Cells Translational Medicine 2017;6:1178-1190.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos/metabolismo , Heparitina Sulfato/farmacología , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Humanos , Microfluídica , Transducción de Señal/efectos de los fármacos
16.
J Mater Chem B ; 5(12): 2263-2272, 2017 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-32263617

RESUMEN

P(TMC-co-LLA) elastomers have shown great potential for various biomaterial and tissue engineering applications. This study systematically investigated properties key to such applications. Three P(TMC-co-LLA) copolymers with 9 to 32 mol% TMC were synthesised and processed into 3D fibrous scaffolds using solution electrospinning. A range of fiber diameters (0.5-5.9 µm) and pore sizes (3.5-19.8 µm) were achieved simply by adjusting the voltage, collector distance and feed rate during electrospinning. The morphological features of the electrospun scaffolds were affected by the copolymer composition such that the average fiber diameters decreased in the order of P(TMC20-co-LLA80) > P(TMC32-co-LLA68) > P(TMC9-co-LLA91), which suggests inherent properties of the copolymers influence the electrospinning process. In addition, the specific parameter combinations applied during electrospinning did not affect the thermal properties of the scaffolds, however, it was confirmed that rapid solidification of the fibers occurred during electrospinning which lowered the inherent crystallinity and caused deviations of the thermodynamic state from equilibrium. Mechanical testing revealed that the Young's modulus and ultimate tensile strength were dependent on the morphology of the fibrous scaffolds, while in contrast, the ductility and toughness were strongly composition dependent with P(TMC20-co-LLA80) scaffolds displaying lower ductility and toughness than P(TMC32-co-LLA68) scaffolds.

18.
Nat Commun ; 7: 12976, 2016 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-27687129

RESUMEN

Axonal retrograde transport of signalling endosomes from the nerve terminal to the soma underpins survival. As each signalling endosome carries a quantal amount of activated receptors, we hypothesized that it is the frequency of endosomes reaching the soma that determines the scale of the trophic signal. Here we show that upregulating synaptic activity markedly increased the flux of plasma membrane-derived retrograde endosomes (labelled using cholera toxin subunit-B: CTB) in hippocampal neurons cultured in microfluidic devices, and live Drosophila larval motor neurons. Electron and super-resolution microscopy analyses revealed that the fast-moving sub-diffraction-limited CTB carriers contained the TrkB neurotrophin receptor, transiently activated by synaptic activity in a BDNF-independent manner. Pharmacological and genetic inhibition of TrkB activation selectively prevented the coupling between synaptic activity and the retrograde flux of signalling endosomes. TrkB activity therefore controls the encoding of synaptic activity experienced by nerve terminals, digitalized as the flux of retrogradely transported signalling endosomes.

19.
Sci Rep ; 6: 24637, 2016 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-27097795

RESUMEN

Inducing cardiomyocyte proliferation in post-mitotic adult heart tissue is attracting significant attention as a therapeutic strategy to regenerate the heart after injury. Model animal screens have identified several candidate signalling pathways, however, it remains unclear as to what extent these pathways can be exploited, either individually or in combination, in the human system. The advent of human cardiac cells from directed differentiation of human pluripotent stem cells (hPSCs) now provides the ability to interrogate human cardiac biology in vitro, but it remains difficult with existing culture formats to simply and rapidly elucidate signalling pathway penetrance and interplay. To facilitate high-throughput combinatorial screening of candidate biologicals or factors driving relevant molecular pathways, we developed a high-density microbioreactor array (HDMA)--a microfluidic cell culture array containing 8100 culture chambers. We used HDMAs to combinatorially screen Wnt, Hedgehog, IGF and FGF pathway agonists. The Wnt activator CHIR99021 was identified as the most potent molecular inducer of human cardiomyocyte proliferation, inducing cell cycle activity marked by Ki67, and an increase in cardiomyocyte numbers compared to controls. The combination of human cardiomyocytes with the HDMA provides a versatile and rapid tool for stratifying combinations of factors for heart regeneration.


Asunto(s)
Diferenciación Celular , Descubrimiento de Drogas , Ensayos Analíticos de Alto Rendimiento , Células Madre Pluripotentes Inducidas/citología , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Proliferación Celular , Células Cultivadas , Descubrimiento de Drogas/instrumentación , Descubrimiento de Drogas/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , Ensayos Analíticos de Alto Rendimiento/normas , Humanos , Miocitos Cardíacos/efectos de los fármacos , Reproducibilidad de los Resultados , Transducción de Señal/efectos de los fármacos
20.
Sci Rep ; 6: 21820, 2016 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-26911660

RESUMEN

We introduce "sense, track and separate" approach for the removal of Hg(2+) ion from aqueous media using highly ordered and magnetic mesoporous ferrosilicate nanocages functionalised with rhodamine fluorophore derivative. These functionalised materials offer both fluorescent and magnetic properties in a single system which help not only to selectively sense the Hg(2+) ions with a high precision but also adsorb and separate a significant amount of Hg(2+) ion in aqueous media. We demonstrate that the magnetic affinity of these materials, generated from the ultrafine γ-Fe2O3 nanoparticles present inside the nanochannels of the support, can efficiently be used as a fluorescent tag to sense the Hg(2+) ions present in NIH3T3 fibroblasts live cells and to track the movement of the cells by external magnetic field monitored using confocal fluorescence microscopy. This simple approach of introducing multiple functions in the magnetic mesoporous materials raise the prospect of creating new advanced functional materials by fusing organic, inorganic and biomolecules to create advanced hybrid nanoporous materials which have a potential use not only for sensing and the separation of toxic metal ions but also for cell tracking in bio-separation and the drug delivery.


Asunto(s)
Mercurio/análisis , Microscopía Fluorescente , Nanoestructuras/química , Espectrometría de Fluorescencia , Animales , Colorantes Fluorescentes/química , Iones/química , Nanopartículas de Magnetita/química , Metales/química , Ratones , Células 3T3 NIH , Porosidad , Rodaminas/química , Silanos/química , Difracción de Rayos X
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...