Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Brain Behav Immun ; 109: 1-22, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36584795

RESUMEN

Multiple sclerosis (MS) is a chronic autoimmune disease of the central nervous system (CNS) characterized by focal inflammatory lesions and prominent demyelination. Even though the currently available therapies are effective in treating the initial stages of disease, they are unable to halt or reverse disease progression into the chronic progressive stage. Thus far, no repair-inducing treatments are available for progressive MS patients. Hence, there is an urgent need for the development of new therapeutic strategies either targeting the destructive immunological demyelination or boosting endogenous repair mechanisms. Using in vitro, ex vivo, and in vivo models, we demonstrate that selective inhibition of phosphodiesterase 4 (PDE4), a family of enzymes that hydrolyzes and inactivates cyclic adenosine monophosphate (cAMP), reduces inflammation and promotes myelin repair. More specifically, we segregated the myelination-promoting and anti-inflammatory effects into a PDE4D- and PDE4B-dependent process respectively. We show that inhibition of PDE4D boosts oligodendrocyte progenitor cells (OPC) differentiation and enhances (re)myelination of both murine OPCs and human iPSC-derived OPCs. In addition, PDE4D inhibition promotes in vivo remyelination in the cuprizone model, which is accompanied by improved spatial memory and reduced visual evoked potential latency times. We further identified that PDE4B-specific inhibition exerts anti-inflammatory effects since it lowers in vitro monocytic nitric oxide (NO) production and improves in vivo neurological scores during the early phase of experimental autoimmune encephalomyelitis (EAE). In contrast to the pan PDE4 inhibitor roflumilast, the therapeutic dose of both the PDE4B-specific inhibitor A33 and the PDE4D-specific inhibitor Gebr32a did not trigger emesis-like side effects in rodents. Finally, we report distinct PDE4D isoform expression patterns in human area postrema neurons and human oligodendroglia lineage cells. Using the CRISPR-Cas9 system, we confirmed that pde4d1/2 and pde4d6 are the key targets to induce OPC differentiation. Collectively, these data demonstrate that gene specific PDE4 inhibitors have potential as novel therapeutic agents for targeting the distinct disease processes of MS.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Esclerosis Múltiple , Inhibidores de Fosfodiesterasa 4 , Humanos , Ratones , Animales , Vaina de Mielina/metabolismo , Esclerosis Múltiple/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/farmacología , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/uso terapéutico , Potenciales Evocados Visuales , Oligodendroglía/metabolismo , Encefalomielitis Autoinmune Experimental/metabolismo , Diferenciación Celular , Inhibidores de Fosfodiesterasa 4/farmacología , Inhibidores de Fosfodiesterasa 4/uso terapéutico , Antiinflamatorios/farmacología , Ratones Endogámicos C57BL
2.
Chem Rev ; 120(19): 10547-10607, 2020 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-32407108

RESUMEN

Bioprinting techniques have been flourishing in the field of biofabrication with pronounced and exponential developments in the past years. Novel biomaterial inks used for the formation of bioinks have been developed, allowing the manufacturing of in vitro models and implants tested preclinically with a certain degree of success. Furthermore, incredible advances in cell biology, namely, in pluripotent stem cells, have also contributed to the latest milestones where more relevant tissues or organ-like constructs with a certain degree of functionality can already be obtained. These incredible strides have been possible with a multitude of multidisciplinary teams around the world, working to make bioprinted tissues and organs more relevant and functional. Yet, there is still a long way to go until these biofabricated constructs will be able to reach the clinics. In this review, we summarize the main bioprinting activities linking them to tissue and organ development and physiology. Most bioprinting approaches focus on mimicking fully matured tissues. Future bioprinting strategies might pursue earlier developmental stages of tissues and organs. The continuous convergence of the experts in the fields of material sciences, cell biology, engineering, and many other disciplines will gradually allow us to overcome the barriers identified on the demanding path toward manufacturing and adoption of tissue and organ replacements.


Asunto(s)
Bioimpresión , Modelos Biológicos , Impresión Tridimensional , Ingeniería de Tejidos , Humanos
3.
Biomacromolecules ; 21(6): 2208-2217, 2020 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-32243138

RESUMEN

Supramolecular and dynamic biomaterials hold promise to recapitulate the time-dependent properties and stimuli-responsiveness of the native extracellular matrix (ECM). Host-guest chemistry is one of the most widely studied supramolecular bonds, yet the binding characteristics of host-guest complexes (ß-CD/adamantane) in relevant biomaterials have mostly focused on singular host-guest interactions or nondiscrete multivalent pendent polymers. The stepwise synergistic effect of multivalent host-guest interactions for the formation of dynamic biomaterials remains relatively unreported. In this work, we study how a series of multivalent adamantane (guest) cross-linkers affect the overall binding affinity and ability to form supramolecular networks with alginate-CD (Alg-CD). These binding constants of the multivalent cross-linkers were determined via NMR titrations and showed increases in binding constants occurring with multivalent constructs. The higher multivalent cross-linkers enabled hydrogel formation; furthermore, an increase in binding and gelation was observed with the inclusion of a phenyl spacer to the cross-linker. A preliminary screen shows that only cross-linking Alg-CD with an 8-arm-multivalent guest results in robust gel formation. These cytocompatible hydrogels highlight the importance of multivalent design for dynamically cross-linked hydrogels. These materials hold promise for development toward cell- and small molecule-delivery platforms and allow discrete and fine-tuning of network properties.


Asunto(s)
Materiales Biocompatibles , Hidrogeles , Alginatos , Polímeros
4.
Nanomedicine ; 10(7): 1559-69, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24792217

RESUMEN

Interactions between Schwann cells (SCs) and scaffolds are important for tissue development during nerve regeneration, because SCs physiologically assist in directing the growth of regenerating axons. In this study, we prepared electrospun scaffolds combining poly (3-hydroxybutyrate) (PHB) and poly (3-hydroxybutyrate-co-3-hydroxyvalerate) (PHBV) functionalized with either collagen I, H-Gly-Arg-Gly-Asp-Ser-OH (GRGDS), H-Tyr-Ile-Gly-Ser-Arg-NH2 (YIGSR), or H-Arg-Asn-Ile-Ala-Glu-Ile-Ile-Lys-Asp-Ile-OH (p20) neuromimetic peptides to mimic naturally occurring ECM motifs for nerve regeneration. Cells cultured on fibrous mats presenting these biomolecules showed a significant increase in metabolic activity and proliferation while exhibiting unidirectional orientation along the orientation of the fibers. Real-time PCR showed cells cultured on peptide-modified scaffolds had a significantly higher neurotrophin expression compared to those on untreated nanofibers. Our study suggests that biofunctionalized aligned PHB/PHBV nanofibrous scaffolds may elicit essential cues for SCs activity and could serve as a potential scaffold for nerve regeneration. From the clinical editor: Nanotechnology-based functionalized scaffolds represent one of the most promising approaches in peripheral nerve recovery, as well as spinal cord recovery. In this study, bio-functionalized and aligned PHB/PHBV nanofibrous scaffolds were found to elicit essential cues for Schwann cell activity, therefore could serve as a potential scaffold for nerve regeneration.


Asunto(s)
Nanofibras , Péptidos/química , Polihidroxialcanoatos/química , Células de Schwann/citología , Andamios del Tejido , Ensayo de Inmunoadsorción Enzimática , Humanos , Microscopía Electrónica de Rastreo , Prohibitinas , Espectroscopía Infrarroja por Transformada de Fourier
5.
Biomater Sci ; 12(6): 1371-1404, 2024 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-38363090

RESUMEN

Peripheral nerve damage results in the loss of sensorimotor and autonomic functions, which is a significant burden to patients. Furthermore, nerve injuries greater than the limiting gap length require surgical repair. Although autografts are the preferred clinical choice, their usage is impeded by their limited availability, dimensional mismatch, and the sacrifice of another functional donor nerve. Accordingly, nerve guidance conduits, which are tubular scaffolds engineered to provide a biomimetic environment for nerve regeneration, have emerged as alternatives to autografts. Consequently, a few nerve guidance conduits have received clinical approval for the repair of short-mid nerve gaps but failed to regenerate limiting gap damage, which represents the bottleneck of this technology. Thus, it is still necessary to optimize the morphology and constituent materials of conduits. This review summarizes the recent advances in nerve conduit technology. Several manufacturing techniques and conduit designs are discussed, with emphasis on the structural improvement of simple hollow tubes, additive manufacturing techniques, and decellularized grafts. The main objective of this review is to provide a critical overview of nerve guidance conduit technology to support regeneration in long nerve defects, promote future developments, and speed up its clinical translation as a reliable alternative to autografts.


Asunto(s)
Materiales Biocompatibles , Traumatismos de los Nervios Periféricos , Humanos , Nervios Periféricos , Andamios del Tejido , Traumatismos de los Nervios Periféricos/cirugía , Regeneración Nerviosa
6.
MethodsX ; 10: 102139, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37025651

RESUMEN

[This corrects the article DOI: 10.1016/j.mex.2019.10.018.].

7.
Acta Biomater ; 165: 111-124, 2023 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-36283613

RESUMEN

Bone fractures are one of the most common traumatic large-organ injuries and although many fractures can heal on their own, 2-12% of fractures are slow healing or do not heal (non-unions). Autologous grafts are currently used for treatment of non-unions but are associated with limited healthy bone tissue. Tissue engineered cell-based products have promise for an alternative treatment method. It was previously demonstrated that cartilaginous microspheroids of periosteum-derived cells could be assembled into scaffold-free constructs and heal murine critically-sized long bone defects (non-unions). However, the handleability of such scaffold-free implants can be compromised when scaling-up. In this work, cartilaginous spheroids were combined with melt electrowritten (MEW) meshes to create an engineered cell-based implant, able to induce in vivo bone formation. MEW polycaprolactone meshes were tailored to contain pores (116 ± 28 µm) of a size that captured microspheroids (180 ± 15 µm). Periosteum-derived microspheroids pre-cultured for 4 days, were seeded on MEW meshes and gene expression analysis demonstrated up-regulation of chondrogenic (SOX9, COL2) and prehypertrophic (VEGF) gene markers after 14 days, creating a biohybrid sheet. When implanted subcutaneously (4 weeks), the biohybrid sheets mineralized (23 ± 3% MV/TV) and formed bone and bone marrow. Bone formation was also observed when implanted in a murine critically-sized long bone defect, though a high variation between samples was detected. The high versatility of this biofabrication approach lies in the possibility to tailor the scaffolds to shape and dimensions corresponding to the large bone defects and the individual patient using robust bone forming building blocks. These strategies are instrumental in the development of personalized regenerative therapies with predictive clinical outcomes. STATEMENT OF SIGNIFICANCE: Successful treatments for healing of large long bone defects are still limited and 2-12% of fractures do not heal properly. We combined a novel biofabrication technique: melt electrowriting (MEW), with robust biology: bone forming cartilaginous spheroids to create biohybrid sheets able to form bone upon implantation. MEW enabled the fabrication of scaffolds with micrometer-sized fibers in defined patterns which allowed the capturing of and merging with cartilaginous spheroids which had the potency to mature into bone via the developmental process of endochondral ossification. The present study contributes to the rapidly growing field of "Biofabrication with Spheroid and Organoid Materials'' and demonstrates design considerations that are of great importance for biofabrication of functional tissues through the assembly of cellular spheroids.


Asunto(s)
Cartílago , Fracturas Óseas , Humanos , Ratones , Animales , Ingeniería de Tejidos/métodos , Osteogénesis , Cicatrización de Heridas , Periostio , Andamios del Tejido
8.
Macromol Biosci ; 23(1): e2200387, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36222273

RESUMEN

Sensory innervation of the skin is essential for its function, homeostasis, and wound healing mechanisms. Thus, to adequately model the cellular microenvironment and function of native skin, in vitro human skin equivalents (hSE) containing a sensory neuron population began to be researched. In this work, a fully human 3D platform of hSE innervated by induced pluripotent stem cell-derived nociceptor neurospheres (hNNs), mimicking the native mode of innervation, is established. Both the hSE and nociceptor population exhibit morphological and phenotypical characteristics resembling their native counterparts, such as epidermal and dermal layer formation and nociceptor marker exhibition, respectively. In the co-culture platform, neurites develop from the hNNs and navigate in 3D to innervate the hSE from a distance. To probe both skin and nociceptor functionality, a clinically available capsaicin patch (Qutenza) is applied directly over the hSE section and neuron reaction is analyzed. Application of the patch causes an exposure time-dependent neurite regression and degeneration. In platforms absent of hSE, axonal degeneration is further increased, highlighting the role of the skin construct as a barrier. In sum, an in vitro tool of functional innervated skin with high interest for preclinical research is established.


Asunto(s)
Células Receptoras Sensoriales , Piel , Humanos , Cicatrización de Heridas , Neuritas , Células Cultivadas
9.
Bioact Mater ; 21: 209-222, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36101857

RESUMEN

The dual role of macrophages in the healing process depends on macrophage ability to polarize into phenotypes that can propagate inflammation or exert anti-inflammatory and tissue-remodeling functions. Controlling scaffold geometry has been proposed as a strategy to influence macrophage behavior and favor the positive host response to implants. Here, we fabricated Polycaprolactone (PCL) scaffolds by Melt Electrowriting (MEW) to investigate the ability of scaffold architecture to modulate macrophage polarization. Primary human macrophages unpolarized (M0) or polarized into M1, M2a, and M2c phenotypes were cultured on PCL films and MEW scaffolds with pore geometries (square, triangle, and rhombus grid) characterized by different angles. M0, M2a, and M2c macrophages wrapped along the fibers, while M1 macrophages formed clusters with rounded cells. Cell bridges were formed only for angles up to 90°. No relevant differences were found among PCL films and 3D scaffolds in terms of surface markers. CD206 and CD163 were highly expressed by M2a and M2c macrophages, with M2a macrophages presenting also high levels of CD86. M1 macrophages expressed moderate levels of all markers. The rhombus architecture promoted an increased release by M2a macrophages of IL10, IL13, and sCD163 compared to PCL films. The proangiogenic factor IL18 was also upregulated by the rhombus configuration in M0 and M2a macrophages compared to PCL films. The interesting findings obtained for the rhombus architecture represent a starting point for the design of scaffolds able to modulate macrophage phenotype, prompting investigations addressed to verify their ability to facilitate the healing process in vivo.

10.
Eur J Pharm Sci ; 185: 106441, 2023 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-37004962

RESUMEN

Phosphodiesterase 4 (PDE4) inhibitors have been extensively researched for their anti-inflammatory and neuroregenerative properties. Despite the known neuroplastic and myelin regenerative properties of nonselective PDE4 inhibitors on the central nervous system, the direct impact on peripheral remyelination and subsequent neuroregeneration has not yet been investigated. Therefore, to examine the possible therapeutic effect of PDE4 inhibition on peripheral glia, we assessed the differentiation of primary rat Schwann cells exposed in vitro to the PDE4 inhibitor roflumilast. To further investigate the differentiation promoting effects of roflumilast, we developed a 3D model of rat Schwann cell myelination that closely resembles the in vivo situation. Using these in vitro models, we demonstrated that pan-PDE4 inhibition using roflumilast significantly promoted differentiation of Schwann cells towards a myelinating phenotype, as indicated by the upregulation of myelin proteins, including MBP and MAG. Additionally, we created a unique regenerative model comprised of a 3D co-culture of rat Schwann cells and human iPSC-derived neurons. Schwann cells treated with roflumilast enhanced axonal outgrowth of iPSC-derived nociceptive neurons, which was accompanied by an accelerated myelination speed, thereby showing not only phenotypic but also functional changes of roflumilast-treated Schwann cells. Taken together, the PDE4 inhibitor roflumilast possesses a therapeutic benefit to stimulate Schwann cell differentiation and, subsequently myelination, as demonstrated in the biologically relevant in vitro platform used in this study. These results can aid in the development of novel PDE4 inhibition-based therapies in the advancement of peripheral regenerative medicine.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4 , Inhibidores de Fosfodiesterasa 4 , Ratas , Animales , Humanos , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/metabolismo , Inhibidores de Fosfodiesterasa 4/farmacología , Inhibidores de Fosfodiesterasa 4/metabolismo , Células de Schwann/metabolismo , Vaina de Mielina/genética
11.
Adv Healthc Mater ; 12(19): e2203021, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37057819

RESUMEN

Cartilage tissue presents low self-repair capability and lesions often undergo irreversible progression. Structures obtained by tissue engineering, such as those based in extrusion bioprinting of constructs loaded with stem cell spheroids may offer valuable alternatives for research and therapeutic purposes. Human mesenchymal stromal cell (hMSC) spheroids can be chondrogenically differentiated faster and more efficiently than single cells. This approach allows obtaining larger tissues in a rapid, controlled and reproducible way. However, it is challenging to control tissue architecture, construct stability, and cell viability during maturation. Herein, this work reports a reproducible bioprinting process followed by a successful post-bioprinting chondrogenic differentiation procedure using large quantities of hMSC spheroids encapsulated in a xanthan gum-alginate hydrogel. Multi-layered constructs are bioprinted, ionically crosslinked, and post chondrogenically differentiated for 28 days. The expression of glycosaminoglycan, collagen II and IV are observed. After 56 days in culture, the bioprinted constructs are still stable and show satisfactory cell metabolic activity with profuse extracellular matrix production. These results show a promising procedure to obtain 3D models for cartilage research and ultimately, an in vitro proof-of-concept of their potential use as stable chondral tissue implants.


Asunto(s)
Bioimpresión , Ingeniería de Tejidos , Humanos , Ingeniería de Tejidos/métodos , Bioimpresión/métodos , Cartílago , Diferenciación Celular , Células Madre , Impresión Tridimensional , Andamios del Tejido/química
12.
Nanotechnology ; 23(27): 275102, 2012 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-22710035

RESUMEN

The F11 hybridoma, a dorsal root ganglion-derived cell line, was used to investigate the response of nociceptive sensory neurons to nanotopographical guidance cues. This established this cell line as a model of peripheral sensory neuron growth for tissue scaffold design. Cells were seeded on substrates of cyclic olefin copolymer (COC) films imprinted via nanoimprint lithography (NIL) with a grating pattern of nano-scale grooves and ridges. Different ridge widths were employed to alter the focal adhesion formation, thereby changing the cell/substrate interaction. Differentiation was stimulated with forskolin in culture medium consisting of either 1 or 10% fetal bovine serum (FBS). Per medium condition, similar neurite alignment was achieved over the four day period, with the 1% serum condition exhibiting longer, more aligned neurites. Immunostaining for focal adhesions found the 1% FBS condition to also have fewer, less developed focal adhesions. The robust response of the F11 to guidance cues further builds on the utility of this cell line as a sensory neuron model, representing a useful tool to explore the design of regenerative guidance tissue scaffolds.


Asunto(s)
Hibridomas/citología , Hibridomas/fisiología , Impresión Molecular/métodos , Células Receptoras Sensoriales/citología , Células Receptoras Sensoriales/fisiología , Ingeniería de Tejidos/métodos , Andamios del Tejido , Animales , Adhesión Celular , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular , Línea Celular , Separación Celular/métodos , Supervivencia Celular , Ratones , Ratas
13.
ACS Appl Mater Interfaces ; 14(28): 31567-31585, 2022 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-35815638

RESUMEN

Nerves and blood vessels are present in most organs and are indispensable for their function and homeostasis. Within these organs, neurovascular (NV) tissue forms congruent patterns and establishes vital interactions. Several human pathologies, including diabetes type II, produce NV disruptions with serious consequences that are complicated to study using animal models. Complex in vitro organ platforms, with neural and vascular supply, allow the investigation of such interactions, whether in a normal or pathological context, in an affordable, simple, and direct manner. To date, a few in vitro models contain NV tissue, and most strategies report models with nonbiomimetic representations of the native environment. To this end, we have established here an NV platform that contains mature vasculature and neural tissue, composed of human microvascular endothelial cells (HMVECs), induced pluripotent stem cell (iPSCs)-derived sensory neurons, and primary rat Schwann cells (SCs) within a fibrin-embedded polymeric scaffold. First, we show that SCs can induce the formation of and stabilize vascular networks to the same degree as the traditional and more thoroughly studied human dermal fibroblasts (HDFs). We also show that through SC prepatterning, we are able to control vessel orientation. Using our NV platform, we demonstrate the concomitant formation of three-dimensional neural and vascular tissue, and the influence of different medium formulations and cell types on the NV tissue outcome. Finally, we propose a protocol to form mature NV tissue, via the integration of independent neural and vascular constituents. The platform described here provides a versatile and advanced model for in vitro research of the NV axis.


Asunto(s)
Células Madre Pluripotentes Inducidas , Tejido Nervioso , Animales , Biomimética , Células Endoteliales/metabolismo , Humanos , Ratas , Células de Schwann/metabolismo
14.
Biomater Adv ; 143: 213183, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36371971

RESUMEN

Despite tissue engineering advances, current nerve guidance conduits (NGCs) are still failing in repairing critical-sized defects. This study aims, therefore, at tackling large nerve gaps (2 cm) by designing NGCs possessing refined physicochemical properties enhancing the activity of Schwann cells (SCs) that support nerve regeneration over long distances. As such, a combinatorial strategy adopting novel plasma-induced surface chemistry and architectural heterogeneity was considered. A mechanically suitable copolymer (Polyactive®) was electrospun to produce nanofibrous NGCs mimicking the extracellular matrix. An innovative seamless double-layered architecture consisting of an inner wall comprised of bundles of aligned fibers with intercalated random fibers and an outer wall fully composed of random fibers was conceived to synergistically provide cell guidance cues and sufficient nutrient inflow. NGCs were subjected to argon plasma treatments using a dielectric barrier discharge (DBD) and a plasma jet (PJ). Surface chemical changes were examined by advanced X-ray photoelectron spectroscopy (XPS) micro-mappings. The DBD homogeneously increased the surface oxygen content from 17 % to 28 % on the inner wall. The PJ created a gradient chemistry throughout the inner wall with an oxygen content gradually increasing from 21 % to 30 %. In vitro studies revealed enhanced primary SC adhesion, elongation and proliferation on plasma-treated NGCs. A cell gradient was observed on the PJ-treated NGCs thus underlining the favorable oxygen gradient in promoting cell chemotaxis. A gradual change from circular to highly elongated SC morphologies mimicking the bands of Büngner was visualized along the gradient. Overall, plasma-treated NGCs are promising candidates paving the way towards critical nerve gap repair.


Asunto(s)
Regeneración Tisular Dirigida , Regeneración Tisular Dirigida/métodos , Andamios del Tejido/química , Regeneración Nerviosa/fisiología , Ingeniería de Tejidos/métodos , Polímeros/química , Oxígeno
15.
ACS Mater Lett ; 4(4): 701-706, 2022 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-36568348

RESUMEN

Smart polymeric biomaterials have been the focus of many recent biomedical studies, especially those with adaptability to defects and potential to be implanted in the human body. Herein we report a versatile and straightforward method to convert non-thermoresponsive hydrogels into thermoresponsive systems with shape memory ability. As a proof of concept, a thermoresponsive polyurethane mesh was embedded within a methacrylated chitosan (CHTMA), gelatin (GELMA), laminarin (LAMMA) or hyaluronic acid (HAMA) hydrogel network, which afforded hydrogel composites with shape memory ability. With this system, we achieved good to excellent shape fixity ratios (50-90%) and excellent shape recovery ratios (∼100%, almost instantaneously) at body temperature (37 °C). Cytocompatibility tests demonstrated good viability either with cells on top or encapsulated during all shape memory processes. This straightforward approach opens a broad range of possibilities to convey shape memory properties to virtually any synthetic or natural-based hydrogel for several biological and nonbiological applications.

16.
Biomater Sci ; 10(17): 4740-4755, 2022 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-35861034

RESUMEN

Few synthetic hydrogels can mimic both the viscoelasticity and supramolecular fibrous structure found in the naturally occurring extracellular matrix (ECM). Furthermore, the ability to control the viscoelasticity of fibrous supramolecular hydrogel networks to influence cell culture remains a challenge. Here, we show that modular mixing of supramolecular architectures with slow and fast exchange dynamics can provide a suitable environment for multiple cell types and influence cellular aggregation. We employed modular mixing of two synthetic benzene-1,3,5-tricarboxamide (BTA) architectures: a small molecule water-soluble BTA with slow exchange dynamics and a telechelic polymeric BTA-PEG-BTA with fast exchange dynamics. Copolymerisation of these two supramolecular architectures was observed, and all tested formulations formed stable hydrogels in water and cell culture media. We found that rational tuning of mechanical and viscoelastic properties is possible by mixing BTA with BTA-PEG-BTA. These hydrogels showed high viability for both chondrocyte (ATDC5) and human dermal fibroblast (HDF) encapsulation (>80%) and supported neuronal outgrowth (PC12 and dorsal root ganglion, DRG). Furthermore, ATDC5s and human mesenchymal stem cells (hMSCs) were able to form spheroids within these viscoelastic hydrogels, with control over cell aggregation modulated by the dynamic properties of the material. Overall, this study shows that modular mixing of supramolecular architectures enables tunable fibrous hydrogels, creating a biomimetic environment for cell encapsulation. These materials are suitable for the formation and culture of spheroids in 3D, critical for upscaling tissue engineering approaches towards cell densities relevant for physiological tissues.


Asunto(s)
Biomimética , Hidrogeles , Benzamidas , Benceno , Humanos , Hidrogeles/química , Agua
17.
ACS Appl Mater Interfaces ; 14(25): 28628-28638, 2022 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-35715217

RESUMEN

Tissue-engineered constructs are currently limited by the lack of vascularization necessary for the survival and integration of implanted tissues. Hydrogen sulfide (H2S), an endogenous signaling gas (gasotransmitter), has been recently reported as a promising alternative to growth factors to mediate and promote angiogenesis in low concentrations. Yet, sustained delivery of H2S remains a challenge. Herein, we have developed angiogenic scaffolds by covalent attachment of an H2S donor to a polycaprolactone (PCL) electrospun scaffold. These scaffolds were engineered to include azide functional groups (on 1, 5, or 10% of the PCL end groups) and were modified using a straightforward click reaction with an alkyne-functionalized N-thiocarboxyanhydride (alkynyl-NTA). This created H2S-releasing scaffolds that rely on NTA ring-opening in water followed by conversion of released carbonyl sulfide into H2S. These functionalized scaffolds showed dose-dependent release of H2S based on the amount of NTA functionality within the scaffold. The NTA-functionalized fibrous scaffolds supported human umbilical vein endothelial cell (HUVEC) proliferation, formed more confluent endothelial monolayers, and facilitated the formation of tight cell-cell junctions to a greater extent than unfunctionalized scaffolds. Covalent conjugation of H2S donors to scaffolds not only promotes HUVEC proliferation in vitro, but also increases neovascularization in ovo, as observed in the chick chorioallantoic membrane assay. NTA-functionalized scaffolds provide localized control over vascularization through the sustained delivery of a powerful endogenous angiogenic agent, which should be further explored to promote angiogenesis in tissue engineering.


Asunto(s)
Sulfuro de Hidrógeno , Animales , Membrana Corioalantoides , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Sulfuro de Hidrógeno/metabolismo , Sulfuro de Hidrógeno/farmacología , Neovascularización Fisiológica , Ingeniería de Tejidos , Andamios del Tejido
18.
Trends Endocrinol Metab ; 32(8): 623-638, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34127366

RESUMEN

Nerves and blood vessels (BVs) establish extensive arborized networks to innervate tissues and deliver oxygen/metabolic support. Developmental cues direct the formation of these intricate and often overlapping patterns, which reflect close interactions within the peripheral neurovascular system. Besides the mutual dependence to survive and function, nerves and BVs share several receptors and ligands, as well as principles of differentiation, growth and pathfinding. Neurovascular (NV) interactions are maintained in adult life and are essential for certain regenerative mechanisms, such as wound healing. In pathological situations (e.g., type 2 diabetes mellitus), the NV system can be severely perturbed and become dysfunctional. Unwanted neural growth and vascularization are also associated with the progression of some pathologies, such as cancer and endometriosis. In this review, we describe the fundamental NV interactions in development, highlighting the similarities between both networks and wiring mechanisms. We also describe the NV contribution to regenerative processes and potential pathological dysfunctions. Finally, we provide an overview of current in vitro models used to replicate and investigate the NV ecosystem, addressing present limitations and future perspectives.


Asunto(s)
Vasos Sanguíneos/fisiología , Nervios Periféricos/fisiología , Diferenciación Celular , Diabetes Mellitus Tipo 2 , Endometriosis , Femenino , Humanos , Neoplasias , Cicatrización de Heridas
19.
ACS Appl Mater Interfaces ; 13(25): 29293-29302, 2021 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-34128651

RESUMEN

Biological recognition sites are very useful for biomedical purposes and, more specifically, for polymeric scaffolds. However, synthetic polymers are not capable of providing specific biological recognition sites. To solve this inconvenience, functionalization of biological moieties is typically performed, oftentimes via peptide binding. In this sense, the main task is capturing the biological complexity of a protein. This study proposes a possible alternative solution to this challenge. Our approach is based on the combination of molecular imprinting (MI) and electrospinning processes. We propose here an alternative MI approach with polymeric structures, instead of using cross-linkers and monomers as conventionally performed. Different PCL-protein scaffolds were produced via electrospinning before performing MI. Gelatin, collagen, and elastin were used as proteins. Results evidenced that the MI process conducted with PCL electrospun membranes was carried out with ionic interactions between the desired molecules and the recognition sites formed. In addition, it has been proved that MI was more efficient when using gelatin as a template. This approach opens a new stage in the development of recognition sites in scaffolds obtained with synthetic polymers and their application for biomedical purposes.


Asunto(s)
Técnicas Electroquímicas/métodos , Impresión Molecular/métodos , Poliésteres/química , Proteínas/química , Gelatina/química , Nanofibras/química
20.
J Biomed Mater Res A ; 109(9): 1600-1612, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33665968

RESUMEN

The creation of skeletal muscle tissue in vitro is a major topic of interest today in the field of biomedical research, due to the lack of treatments for muscle loss due to traumatic accidents or disease. For this reason, the intrinsic properties of nanofibrillar structures to promote cell adhesion, proliferation, and cell alignment presents an attractive tool for regenerative medicine to recreate organized tissues such as muscle. Electrospinning is one of the processing techniques often used for the fabrication of these nanofibrous structures and the combination of synthetic and natural polymers is often required to achieve optimal mechanical and physiochemical properties. Here, polycaprolactone (PCL) is selected as a synthetic polymer used for the fabrication of scaffolds, and the effect of protein addition on the final scaffolds' properties is studied. Collagen and gelatin were the proteins selected and two different concentrations were analyzed (2 and 4 wt/vol%). Different PCL/protein systems were prepared, and a structural, mechanical and functional characterization was performed. The influence of fiber alignment on the properties of the final scaffolds was assessed through morphological, mechanical and biological evaluations. A bioreactor was used to promote cell proliferation and differentiation within the scaffolds. The results revealed that protein addition produced a decrease in the fiber size of the membranes, an increase in their hydrophilicity, and a softening of their mechanical properties. The biological study showed the ability of the selected systems to harbor cells, allow their growth and, potentially, develop musculoskeletal tissues.


Asunto(s)
Colágeno/farmacología , Gelatina/farmacología , Músculo Esquelético/fisiología , Poliésteres/farmacología , Ingeniería de Tejidos , Andamios del Tejido/química , Animales , Colágeno/ultraestructura , Módulo de Elasticidad , Peces , Gelatina/ultraestructura , Músculo Esquelético/efectos de los fármacos , Nanofibras/química , Nanofibras/ultraestructura , Estrés Mecánico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA