Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Soft Matter ; 2024 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-38841883

RESUMEN

Correction for 'Flax fibre reinforced alginate poloxamer hydrogel: assessment of mechanical and 4D printing potential' by Charles de Kergariou et al., Soft Matter, 2024, 20, 4021-4034, https://doi.org/10.1039/D4SM00135D.

2.
Int J Oral Sci ; 16(1): 37, 2024 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-38734663

RESUMEN

Emerging regenerative cell therapies for alveolar bone loss have begun to explore the use of cell laden hydrogels for minimally invasive surgery to treat small and spatially complex maxilla-oral defects. However, the oral cavity presents a unique and challenging environment for in vivo bone tissue engineering, exhibiting both hard and soft periodontal tissue as well as acting as key biocenosis for many distinct microbial communities that interact with both the external environment and internal body systems, which will impact on cell fate and subsequent treatment efficacy. Herein, we design and bioprint a facile 3D in vitro model of a human dentine interface to probe the effect of the dentine surface on human mesenchymal stem cells (hMSCs) encapsulated in a microporous hydrogel bioink. We demonstrate that the dentine substrate induces osteogenic differentiation of encapsulated hMSCs, and that both dentine and ß-tricalcium phosphate substrates stimulate extracellular matrix production and maturation at the gel-media interface, which is distal to the gel-substrate interface. Our findings demonstrate the potential for long-range effects on stem cells by mineralized surfaces during bone tissue engineering and provide a framework for the rapid development of 3D dentine-bone interface models.


Asunto(s)
Diferenciación Celular , Dentina , Células Madre Mesenquimatosas , Osteogénesis , Ingeniería de Tejidos , Humanos , Osteogénesis/fisiología , Ingeniería de Tejidos/métodos , Fosfatos de Calcio , Hidrogeles , Técnicas In Vitro , Bioimpresión , Andamios del Tejido , Propiedades de Superficie , Matriz Extracelular , Células Cultivadas
3.
Soft Matter ; 20(19): 4021-4034, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38695256

RESUMEN

The mechanical and printing performance of a new biomaterial, flax fibre-reinforced alginate-poloxamer based hydrogel, for load-bearing and 4D printing biomedical applications is described in this study. The-self suspendable ability of the material was evaluated by optimising the printing parameters and conducting a collapse test. 1% of the flax fibre weight fraction was sufficient to obtain an optimum hydrogel composite from a mechanical perspective. The collapse test showed that the addition of flax fibres allowed a consistent print without support over longer distances (8 and 10 mm) than the unreinforced hydrogel. The addition of 1% of flax fibres increased the viscosity by 39% and 129% at strain rates of 1 rad s-1 and 5 rad s-1, respectively, compared to the unreinforced hydrogel. The distributions of fibre size and orientation inside the material were also evaluated to identify the internal morphology of the material. The difference of coefficients of moisture expansion between the printing direction (1.29 × 10-1) and the transverse direction (6.03 × 10-1) showed potential for hygromorphic actuation in 4D printing. The actuation authority was demonstrated by printing a [0°; 90°] stacking sequence and rosette-like structures, which were then actuated using humidity gradients. Adding fibres to the hydrogel improved the repeatability of the actuation, while lowering the actuation authority from 0.11 mm-1 to 0.08 mm-1. Overall, this study highlighted the structural and actuation-related benefits of adding flax fibres to hydrogels.

4.
STAR Protoc ; 5(1): 102899, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38367231

RESUMEN

Surgical treatment of pediatric congenital heart disease with tissue grafts is a lifesaving intervention. Decellularization to reduce immunogenicity of tissue grafts is an increasingly popular alternative to glutaraldehyde fixation. Here, we present a protocol to decellularize porcine right ventricular outflow tracts using a 3D printed flow chamber. We describe steps for 3D printing the flow rig, preparing porcine tissue, and using the flow rig to utilize shear forces for decellularization. We then detail procedures for characterizing the acellular scaffold. For complete details on the use and execution of this protocol, please refer to Vafaee et al.1.


Asunto(s)
Ventrículos Cardíacos , Impresión Tridimensional , Porcinos , Humanos , Niño , Animales , Ventrículos Cardíacos/diagnóstico por imagen
5.
J Mater Chem B ; 11(46): 11006-11023, 2023 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-37953707

RESUMEN

Neuronal tissue engineering has immense potential for treating neurological disorders and facilitating nerve regeneration. Conducting polymers (CPs) have emerged as a promising class of materials owing to their unique electrical conductivity and biocompatibility. CPs, such as poly(3,4-ethylenedioxythiophene) (PEDOT), poly(3-hexylthiophene) (P3HT), polypyrrole (PPy), and polyaniline (PANi), have been extensively explored for their ability to provide electrical cues to neural cells. These polymers are widely used in various forms, including porous scaffolds, hydrogels, and nanofibers, and offer an ideal platform for promoting cell adhesion, differentiation, and axonal outgrowth. CP-based scaffolds can also serve as drug delivery systems, enabling localized and controlled release of neurotrophic factors and therapeutic agents to enhance neural regeneration and repair. CP-based scaffolds have demonstrated improved neural regeneration, both in vitro and in vivo, for treating spinal cord and peripheral nerve injuries. In this review, we discuss synthesis and scaffold processing methods for CPs and their applications in neuronal tissue regeneration. We focused on a detailed literature review of the central and peripheral nervous systems.


Asunto(s)
Polímeros , Ingeniería de Tejidos , Ingeniería de Tejidos/métodos , Polímeros/uso terapéutico , Andamios del Tejido , Pirroles/farmacología , Neuronas
6.
J Am Chem Soc ; 145(41): 22659-22670, 2023 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-37812759

RESUMEN

Lipid nanoparticles (LNPs) are becoming widely adopted as vectors for the delivery of therapeutic payloads but generally lack intrinsic tissue-homing properties. These extracellular vesicle (EV) mimetics can be targeted toward the liver, lung, or spleen via charge modification of their lipid headgroups. Homing to other tissues has only been achieved via covalent surface modification strategies using small-molecule ligands, peptides, or monoclonal antibodies─methods that are challenging to couple with large-scale manufacturing. Herein, we design a novel modular artificial membrane-binding protein (AMBP) platform for the modification of LNPs postformation. The system is composed of two protein modules that can be readily coupled using bioorthogonal chemistry to yield the AMBP. The first is a membrane anchor module comprising a supercharged green fluorescent protein (scGFP) electrostatically conjugated to a dynamic polymer surfactant corona. The second is a functional module containing a cardiac tissue fibronectin homing sequence from the bacterial adhesin CshA. We demonstrate that LNPs modified using the AMBP exhibit a 20-fold increase in uptake by fibronectin-rich C2C12 cells under static conditions and a 10-fold increase under physiologically relevant shear stresses, with no loss of cell viability. Moreover, we show targeted localization of the AMBP-modified LNPs in zebrafish hearts, highlighting their therapeutic potential as a vector for the treatment of cardiac disease and, more generally, as a smart vector.


Asunto(s)
Fibronectinas , Nanopartículas , Animales , Pez Cebra , Liposomas , Nanopartículas/química , ARN Interferente Pequeño/química
7.
J Mater Chem B ; 11(27): 6225-6248, 2023 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-37309580

RESUMEN

Nanomaterial composition, morphology, and mechanical performance are critical parameters for tissue engineering. Within this rapidly expanding space, tubular nanomaterials (TNs), including carbon nanotubes (CNTs), titanium oxide nanotubes (TNTs), halloysite nanotubes (HNTs), silica nanotubes (SiNTs), and hydroxyapatite nanotubes (HANTs) have shown significant potential across a broad range of applications due to their high surface area, versatile surface chemistry, well-defined mechanical properties, excellent biocompatibility, and monodispersity. These include drug delivery vectors, imaging contrast agents, and scaffolds for bone tissue engineering. This review is centered on the recent developments in TN-based biomaterials for structural tissue engineering, with a strong focus on bone tissue regeneration. It includes a detailed literature review on TN-based orthopedic coatings for metallic implants and composite scaffolds to enhance in vivo bone regeneration.


Asunto(s)
Nanotubos de Carbono , Ingeniería de Tejidos , Ingeniería de Tejidos/métodos , Nanotubos de Carbono/química , Huesos , Materiales Biocompatibles/química , Durapatita/química
8.
Drug Deliv Transl Res ; 13(11): 2719-2738, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37301780

RESUMEN

The burden of cancer is increasing globally. Several challenges facing its mainstream treatment approaches have formed the basis for the development of targeted delivery systems to carry and distribute anti-cancer payloads to their defined targets. This site-specific delivery of drug molecules and gene payloads to selectively target druggable biomarkers aimed at inducing cell death while sparing normal cells is the principal goal for cancer therapy. An important advantage of a delivery vector either viral or non-viral is the cumulative ability to penetrate the haphazardly arranged and immunosuppressive tumour microenvironment of solid tumours and or withstand antibody-mediated immune response. Biotechnological approaches incorporating rational protein engineering for the development of targeted delivery systems which may serve as vehicles for packaging and distribution of anti-cancer agents to selectively target and kill cancer cells are highly desired. Over the years, these chemically and genetically modified delivery systems have aimed at distribution and selective accumulation of drug molecules at receptor sites resulting in constant maintenance of high drug bioavailability for effective anti-tumour activity. In this review, we highlighted the state-of-the art viral and non-viral drug and gene delivery systems and those under developments focusing on cancer therapy.


Asunto(s)
Antineoplásicos , Neoplasias , Humanos , Vectores Genéticos , Terapia Genética/métodos , Técnicas de Transferencia de Gen , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Antineoplásicos/uso terapéutico , Microambiente Tumoral
9.
Int J Biol Macromol ; 233: 123511, 2023 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-36773882

RESUMEN

The appreciation of how conventional and fossil-based materials could be harmful to our planet is growing, especially when considering single-use and non-biodegradable plastics manufactured from fossil fuels. Accordingly, tackling climate change and plastic waste pollution entails a more responsible approach to sourcing raw materials and the adoption of less destructive end-of-life pathways. Livestock animals, in particular ruminants, process plant matter using a suite of mechanical, chemical and biological mechanisms through the act of digestion. The manure from these "living bioreactors" is ubiquitous and offers a largely untapped source of lignocellulosic biomass for the development of bio-based and biodegradable materials. In this review, we assess recent studies made into manure-based cellulose materials in terms of their material characteristics and implications for sustainability. Despite the surprisingly diverse body of research, it is apparent that progress towards the commercialisation of manure-derived cellulose materials is hindered by a lack of truly sustainable options and robust data to assess the performance against conventional materials alternatives. Nanocellulose, a natural biopolymer, has been successfully produced by living bioreactors and is presented as a candidate for future developments. Life cycle assessments from non-wood sources are however minimal, but there are some initial indications that manure-derived nanocellulose would offer environmental benefits over traditional wood-derived sources.


Asunto(s)
Celulosa , Estiércol , Animales , Contaminación Ambiental , Plásticos , Reactores Biológicos
10.
Biofabrication ; 15(1)2022 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-36321254

RESUMEN

We describe the development of a high-throughput bioprinted colorectal cancer (CRC) spheroid platform with high levels of automation, information content, and low cell number requirement. This is achieved via the formulation of a hydrogel bioink with a compressive Young's modulus that is commensurate with that of colonic tissue (1-3 kPa), which supports exponential growth of spheroids from a wide range of CRC cell lines. The resulting spheroids display tight cell-cell junctions, bioink matrix-cell interactions and necrotic hypoxic cores. By combining high content light microscopy imaging and processing with rapid multiwell plate bioprinting, dose-response profiles are generated from CRC spheroids challenged with oxaliplatin (OX) and fluorouracil (5FU), as well as radiotherapy. Bioprinted CRC spheroids are shown to exhibit high levels of chemoresistance relative to cell monolayers, and OX was found to be significantly less effective against tumour spheroids than in monolayer culture, when compared to 5FU.


Asunto(s)
Bioimpresión , Neoplasias Colorrectales , Humanos , Esferoides Celulares , Bioimpresión/métodos , Fluorouracilo , Línea Celular , Oxaliplatino
11.
Adv Sci (Weinh) ; 9(32): e2202359, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35988154

RESUMEN

The surface of a carboxylate-enriched octuple mutant of Bacillus subtilis lipase A (8M) is chemically anionized to produce core (8M)-shell (cationic polymer surfactants) bionanoconjugates in protein liquid form, which are termed anion-type biofluids. The resultant lipase biofluids exhibit a 2.5-fold increase in hydrolytic activity when compared with analogous lipase biofluids based on anionic polymer surfactants. In addition, the applicability of the anion-type biofluid using Myoglobin (Mb) that is well studied in anion-type solvent-free liquid proteins is evaluated. Although anionization resulted in the complete unfolding of Mb, the active α-helix level is partially recovered in the anion-type biofluids, and the effect is accentuated in the cation-type Mb biofluids. These highly active anion-type solvent-free liquid enzymes exhibit increased thermal stability and provide a new direction in solvent-free liquid protein research.


Asunto(s)
Lipasa , Tensoactivos , Solventes/química , Lipasa/química , Lipasa/metabolismo , Tensoactivos/química , Hidrólisis , Polímeros/química , Mioglobina/química
12.
Biomaterials ; 285: 121547, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35533445

RESUMEN

Targeting stem cells to cartilage lesions has the potential to enhance engraftment and chondrogenesis. Denatured type II collagen fibrils (gelatin) are exposed in lesions at the surface of osteoarthritic articular cartilage and are therefore ideal target sites. We have designed and investigated chimeric mutants of the three modules of the MMP-2 collagen binding domain (CBD) as potential ligands for stem cell targeting. We expressed full-length CBD for the first time and used it to identify the most important amino acid residues for binding to gelatin. Module 2 of CBD had the highest affinity binding to both Type I and Type II gelatin, whereas module 1 showed specificity for type II gelatin and module 3 for type I gelatin. We went on to generate chimeric forms of CBD consisting of three repeats of module 1 (111), module 2 (222) or module 3 (333). 111 lacked solubility and could not be further characterised. However 222 was found to bind to type II gelatin 14 times better than CBD, suggesting it would be optimal for attachment to cartilage lesions, whilst 333 was found to bind to type I gelatin 12 times better than CBD, suggesting it would be optimal for attachment to lesions in type I collagen-rich tissues. We coated 222 onto the external membrane of Mesenchymal Stem Cells and demonstrated higher attachment of the coated cells to type II gelatin than uncoated cells. We conclude that the three modules of CBD each have specific biological properties that can be exploited for targeting stem cells to cartilage lesions and other pathological sites.


Asunto(s)
Cartílago Articular , Metaloproteinasa 2 de la Matriz , Proteínas Portadoras/metabolismo , Cartílago/metabolismo , Cartílago Articular/metabolismo , Colágeno Tipo I/metabolismo , Gelatina , Metaloproteinasa 2 de la Matriz/metabolismo , Membranas Artificiales , Unión Proteica , Estructura Terciaria de Proteína , Células Madre/metabolismo
13.
ACS Appl Mater Interfaces ; 13(50): 60433-60445, 2021 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-34894651

RESUMEN

Catalytically active materials for the enhancement of personalized protective equipment (PPE) could be advantageous to help alleviate threats posed by neurotoxic organophosphorus compounds (OPs). Accordingly, a chimeric protein comprised of a supercharged green fluorescent protein (scGFP) and phosphotriesterase from Agrobacterium radiobacter (arPTE) was designed to drive the polymer surfactant (S-)-mediated self-assembly of microclusters to produce robust, enzymatically active materials. The chimera scGFP-arPTE was structurally characterized via circular dichroism spectroscopy and synchrotron radiation small-angle X-ray scattering, and its biophysical properties were determined. Significantly, the chimera exhibited greater thermal stability than the native constituent proteins, as well as a higher catalytic turnover number (kcat). Furthermore, scGFP-arPTE was electrostatically complexed with monomeric S-, driving self-assembly into [scGFP-arPTE][S-] nanoclusters, which could be dehydrated and cross-linked to yield enzymatically active [scGFP-arPTE][S-] porous films with a high-order structure. Moreover, these clusters could self-assemble within cotton fibers to generate active composite textiles without the need for the pretreatment of the fabrics. Significantly, the resulting materials maintained the biophysical activities of both constituent proteins and displayed recyclable and persistent activity against the nerve agent simulant paraoxon.


Asunto(s)
Materiales Biocompatibles/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Hidrolasas de Triéster Fosfórico/metabolismo , Polímeros/metabolismo , Tensoactivos/metabolismo , Textiles , Agrobacterium tumefaciens/enzimología , Materiales Biocompatibles/química , Proteínas Fluorescentes Verdes/química , Ensayo de Materiales , Modelos Moleculares , Tamaño de la Partícula , Hidrolasas de Triéster Fosfórico/química , Polímeros/química , Tensoactivos/química
14.
Biomaterials ; 276: 120996, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34280823

RESUMEN

One of the major challenges within the emerging field of injectable stem cell therapies for articular cartilage (AC) repair is the retention of sufficient viable cell numbers at the site of injury. Even when delivered via intra-articular injection, the number of stem cells retained at the target is often low and declines rapidly over time. To address this challenge, an artificial plasma membrane binding nanocomplex was rationally designed to provide human mesenchymal stem cells (hMSCs) with increased adhesion to articular cartilage tissue. The nanocomplex comprises the extracellular matrix (ECM) binding peptide of a placenta growth factor-2 (PlGF-2) fused to a supercharged green fluorescent protein (scGFP), which was electrostatically conjugated to anionic polymer surfactant chains to yield [S-]scGFP_PlGF2. The [S-]scGFP_PlGF2 nanocomplex spontaneously inserts into the plasma membrane of hMSCs, is not cytotoxic, and does not inhibit differentiation. The nanocomplex-modified hMSCs showed a significant increase in affinity for immobilised collagen II, a key ECM protein of cartilage, in both static and dynamic cell adhesion assays. Moreover, the cells adhered strongly to bovine ex vivo articular cartilage explants resulting in high cell numbers. These findings suggest that the re-engineering of hMSC membranes with [S-]scGFP_PlGF2 could improve the efficacy of injectable stem cell-based therapies for the treatment of damaged articular cartilage.


Asunto(s)
Cartílago Articular , Células Madre Mesenquimatosas , Animales , Proteínas Portadoras , Bovinos , Adhesión Celular , Diferenciación Celular , Condrogénesis , Matriz Extracelular , Humanos , Membranas Artificiales , Polímeros , Células Madre , Tensoactivos
15.
Int J Nanomedicine ; 16: 2585-2595, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33833513

RESUMEN

BACKGROUND: In cancer nanomedicine, drugs are transported by nanocarriers through a biological system to produce a therapeutic effect. The efficacy of the treatment is affected by the ability of the nanocarriers to overcome biological transport barriers to reach their target. In this work, we focus on the process of nanocarrier penetration through tumour tissue after extravasation. Visualising the dynamics of nanocarriers in tissue is difficult in vivo, and in vitro assays often do not capture the spatial and physical constraints relevant to model tissue penetration. METHODS: We propose a new simple, low-cost method to observe the transport dynamics of nanoparticles through a tissue-mimetic microfluidic chip. After loading a chip with triplicate conditions of gel type and loading with microparticles, microscopic analysis allows for tracking of fluorescent nanoparticles as they move through hydrogels (Matrigel and Collagen I) with and without cell-sized microparticles. A bespoke image-processing codebase written in MATLAB allows for statistical analysis of this tracking, and time-dependent dynamics can be determined. RESULTS: To demonstrate the method, we show size-dependence of transport mechanics can be observed, with diffusion of fluorescein dye throughout the channel in 8 h, while 20 nm carboxylate FluoSphere diffusion was hindered through both Collagen I and Matrigel™. Statistical measurements of the results are generated through the software package and show the significance of both size and presence of microparticles on penetration depth. CONCLUSION: This provides an easy-to-understand output for the end user to measure nanoparticle tissue penetration, enabling the first steps towards future automated experimentation of transport dynamics for rational nanocarrier design.


Asunto(s)
Geles/química , Microfluídica/métodos , Nanopartículas/administración & dosificación , Nanopartículas/metabolismo , Andamios del Tejido/química , Colágeno/química , Colágeno/metabolismo , Difusión , Humanos , Nanomedicina/métodos , Nanopartículas/química
16.
Stem Cells Transl Med ; 10(6): 855-866, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33660953

RESUMEN

Myocardial infarction (MI) has been the primary cause of death in developed countries, resulting in a major psychological and financial burden for society. Current treatments for acute MI are directed toward rapid restoration of perfusion to limit damage to the myocardium, rather than promoting tissue regeneration and subsequent contractile function recovery. Regenerative cell therapies (CTs), in particular those using multipotent stem cells (SCs), are in the spotlight for treatment post-MI. Unfortunately, the efficacy of CTs is somewhat limited by their poor long-term viability, homing, and engraftment to the myocardium. In response, a range of novel SC-based technologies are in development to provide additional cellular modalities, bringing CTs a step closer to the clinic. In this review, the current landscape of emerging CTs and their augmentation strategies for the treatment post-MI are discussed. In doing so, we highlight recent advances in cell membrane reengineering via genetic modifications, recombinant protein immobilization, and the utilization of soft biomimetic scaffold interfaces.


Asunto(s)
Infarto del Miocardio , Miocardio , Trasplante de Células Madre , Humanos , Células Madre Multipotentes/citología , Infarto del Miocardio/fisiopatología , Miocardio/citología , Regeneración , Trasplante de Células Madre/métodos , Cicatrización de Heridas
17.
Phys Rev Lett ; 126(8): 088102, 2021 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-33709739

RESUMEN

The interaction between proteins and hydration water stabilizes protein structure and promotes functional dynamics, with water translational motions enabling protein flexibility. Engineered solvent-free protein-polymer hybrids have been shown to preserve protein structure, function, and dynamics. Here, we used neutron scattering, protein and polymer perdeuteration, and molecular dynamics simulations to explore how a polymer dynamically replaces water. Even though relaxation rates and vibrational properties are strongly modified in polymer coated compared to hydrated proteins, liquidlike polymer dynamics appear to plasticize the conjugated protein in a qualitatively similar way as do hydration-water translational motions.


Asunto(s)
Polímeros/química , Proteínas/química , Diaminas/química , Glicolatos/química , Enlace de Hidrógeno , Simulación de Dinámica Molecular , Mioglobina/química , Difracción de Neutrones , Polietilenglicoles/química , Conformación Proteica , Termodinámica , Agua/química
18.
ACS Appl Polym Mater ; 3(12): 6070-6077, 2021 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-35983011

RESUMEN

Here, we describe a facile route to the synthesis of enzymatically active highly fabricable plastics, where the enzyme is an intrinsic component of the material. This is facilitated by the formation of an electrostatically stabilized enzyme-polymer surfactant nanoconstruct, which, after lyophilization and melting, affords stable macromolecular dispersions in a wide range of organic solvents. A selection of plastics can then be co-dissolved in the dispersions, which provides a route to bespoke 3D enzyme plastic nanocomposite structures using a wide range of fabrication techniques, including melt electrowriting, casting, and piston-driven 3D printing. The resulting constructs comprising active phosphotriesterase (arPTE) readily detoxify organophosphates with persistent activity over repeated cycles and for long time periods. Moreover, we show that the protein guest molecules, such as arPTE or sfGFP, increase the compressive Young's modulus of the plastics and that the identity of the biomolecule influences the nanomorphology and mechanical properties of the resulting materials. Overall, we demonstrate that these biologically active nanocomposite plastics are compatible with state-of-the-art 3D fabrication techniques and that the methodology could be readily applied to produce robust and on-demand smart nanomaterial structures.

19.
Front Cardiovasc Med ; 7: 598890, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33330660

RESUMEN

Cell therapies are emerging as a new therapeutic frontier for the treatment of ischemic disease. However, femoral occlusions can be challenging environments for effective therapeutic cell delivery. In this study, cell-engineered hybrid scaffolds are implanted around the occluded femoral artery and the therapeutic benefit through the formation of new collateral arteries is investigated. First, it is reported the fabrication of different hybrid "hard-soft" 3D channel-shaped scaffolds comprising either poly(ε-caprolactone) (PCL) or polylactic-co-glycolic acid (PLGA) and electro-spun of gelatin (GL) nanofibers. Both PCL-GL and PLGA-GL scaffolds show anisotropic characteristics in mechanical tests and PLGA displays a greater rigidity and faster degradability in wet conditions. The resulting constructs are engineered using human adventitial pericytes (APCs) and both exhibit excellent biocompatibility. The 3D environment also induces expressional changes in APCs, conferring a more pronounced proangiogenic secretory profile. Bioprinting of alginate-pluronic gel (AG/PL), containing APCs and endothelial cells, completes the hybrid scaffold providing accurate spatial organization of the delivered cells. The scaffolds implantation around the mice occluded femoral artery shows that bioengineered PLGA hybrid scaffold outperforms the PCL counterpart accelerating limb blood flow recovery through the formation arterioles with diameters >50 µm, demonstrating the therapeutic potential in stimulating reparative angiogenesis.

20.
Adv Biosyst ; 4(11): e2000101, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33166084

RESUMEN

The extent to which biologic payloads can be effectively delivered to cells is a limiting factor in the development of new therapies. Limitations arise from the lack of pharmacokinetic stability of biologics in vivo. Encapsulating biologics in a protective delivery vector has the potential to improve delivery profile and enhance performance. Coacervate microdroplets are developed as cell-mimetic materials with established potential for the stabilization of biological molecules, such as proteins and nucleic acids. Here, the development of biodegradable coacervate microvectors (comprising synthetically modified amylose polymers) is presented, for the delivery of biologic payloads to cells. Amylose-based coacervate microdroplets are stable under physiological conditions (e.g., temperature and ionic strength), are noncytotoxic owing to their biopolymeric structure, spontaneously interacted with the cell membrane, and are able to deliver and release proteinaceous payloads beyond the plasma membrane. In particular, myoglobin, an oxygen storage and antioxidant protein, is successfully delivered into human mesenchymal stem cells (hMSCs) within 24 h. Furthermore, coacervate microvectors are implemented for the delivery of human bone morphogenetic protein 2 growth factor, inducing differentiation of hMSCs into osteoprogenitor cells. This study demonstrates the potential of coacervate microdroplets as delivery microvectors for biomedical research and the development of new therapies.


Asunto(s)
Proteína Morfogenética Ósea 2 , Diferenciación Celular/efectos de los fármacos , Sistemas de Liberación de Medicamentos/métodos , Células Madre Mesenquimatosas/metabolismo , Amilosa/química , Biopolímeros/química , Proteína Morfogenética Ósea 2/química , Proteína Morfogenética Ósea 2/farmacocinética , Proteína Morfogenética Ósea 2/farmacología , Células Cultivadas , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA