Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Adv Sci (Weinh) ; 11(11): e2308478, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38113315

RESUMO

Discogenic pain is associated with deep nerve ingrowth in annulus fibrosus tissue (AF) of intervertebral disc (IVD). To model AF nerve ingrowth, primary bovine dorsal root ganglion (DRG) micro-scale tissue units are spatially organised around an AF explant by mild hydrodynamic forces within a collagen matrix. This results in a densely packed multicellular system mimicking the native DRG tissue morphology and a controlled AF-neuron distance. Such a multicellular organisation is essential to evolve populational-level cellular functions and in vivo-like morphologies. Pro-inflammatory cytokine-primed AF demonstrates its neurotrophic and neurotropic effects on nociceptor axons. Both effects are dependent on the AF-neuron distance underpinning the role of recapitulating inter-tissue/organ anatomical proximity when investigating their crosstalk. This is the first in vitro model studying AF nerve ingrowth by engineering mature and large animal tissues in a morphologically and physiologically relevant environment. The new approach can be used to biofabricate multi-tissue/organ models for untangling pathophysiological conditions and develop novel therapies.


Assuntos
Degeneração do Disco Intervertebral , Disco Intervertebral , Animais , Bovinos , Colágeno , Neurônios , Gânglios Espinais
2.
Materials (Basel) ; 16(22)2023 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-38005143

RESUMO

Osteochondral (OC) disorders such as osteoarthritis (OA) damage joint cartilage and subchondral bone tissue. To understand the disease, facilitate drug screening, and advance therapeutic development, in vitro models of OC tissue are essential. This study aims to create a bioprinted OC miniature construct that replicates the cartilage and bone compartments. For this purpose, two hydrogels were selected: one composed of gelatin methacrylate (GelMA) blended with nanosized hydroxyapatite (nHAp) and the other consisting of tyramine-modified hyaluronic acid (THA) to mimic bone and cartilage tissue, respectively. We characterized these hydrogels using rheological testing and assessed their cytotoxicity with live-dead assays. Subsequently, human osteoblasts (hOBs) were encapsulated in GelMA-nHAp, while micropellet chondrocytes were incorporated into THA hydrogels for bioprinting the osteochondral construct. After one week of culture, successful OC tissue generation was confirmed through RT-PCR and histology. Notably, GelMA/nHAp hydrogels exhibited a significantly higher storage modulus (G') compared to GelMA alone. Rheological temperature sweeps and printing tests determined an optimal printing temperature of 20 °C, which remained unaffected by the addition of nHAp. Cell encapsulation did not alter the storage modulus, as demonstrated by amplitude sweep tests, in either GelMA/nHAp or THA hydrogels. Cell viability assays using Ca-AM and EthD-1 staining revealed high cell viability in both GelMA/nHAp and THA hydrogels. Furthermore, RT-PCR and histological analysis confirmed the maintenance of osteogenic and chondrogenic properties in GelMA/nHAp and THA hydrogels, respectively. In conclusion, we have developed GelMA-nHAp and THA hydrogels to simulate bone and cartilage components, optimized 3D printing parameters, and ensured cell viability for bioprinting OC constructs.

3.
Mater Today Bio ; 22: 100775, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37674778

RESUMO

Herein we show an accessible technique based on Faraday waves that assist the rapid assembly of osteoinductive ß-Tricalcium phosphate (ß-TCP) particles as well as human osteoblast pre-assembled in spheroids. The hydrodynamic forces originating at 'seabed' of the assembly chamber can be used to tightly aggregate inorganic and biological entities at packing densities that resemble those of native tissues. Additionally, following a layer-by-layer assembly procedure, centimeter scaled osteoinductive three-dimensional and cellularized constructs have been fabricated. We showed that the intimate connection between biological building blocks is essential in engineering living system able of localized mineral deposition. Our results demonstrate, for the first time, the possibility to obtain three-dimensional cellularized and acellularized anisotropic constructs using Faraday waves.

4.
Mater Today Bio ; 16: 100357, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35880098

RESUMO

The tumor microenvironment (TME), consisting of extracellular matrix, proteins, stromal cells, and a vascular system, is reported to have a key role in cancer progression and prognosis. Thereby, the interaction between the vascular network and tumor mass is an important feature of the TME since the anticancer agents which are delivered to the TME can trigger the vascular response and influence the therapeutic outcome of the treatment. To identify and develop new therapeutic strategies, 3D in vitro models that recapitulate the complexity of the TME are urgently needed. Among them, vascularized tumor models are a promising approach, allowing to target tumor angiogenesis and reduce tumor growth. By using sound patterning, cells can be condensed locally into highly reproducible patterns through the action of mild hydrodynamic forces. Here, we use a soundwave-driven cell assembly approach to create a ring-shaped microcapillary network in fibrin hydrogel. Then, we generate a 3D vascularized tumor model by combining a tumor heterotypic spheroid, consisting of fibroblasts and Malignant Pleural Mesothelioma (MPM) cells, with the surrounding vascular ring. Based on its shape, we name it Saturn-like vascularized Tumor Model (STM). The growth of the microcapillary network is monitored over time by fluorescence imaging. The area covered by the microcapillary network, and its continuous increase in presence of the heterotypic tumor spheroid was monitored. Interestingly, this effect is enhanced when treating the STM with the anticancer agent Cisplatin. Overall, we show the use of sound patterning as a fast and cell-friendly approach to spatially organize and condense cells, to generate a 3D in vitro platform from which simple readouts of drug tests can be extracted by image analysis, with the potential to provide a model system for tailored tumor therapy.

5.
Biomater Sci ; 9(4): 1237-1245, 2021 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-33576754

RESUMO

The uniform and aligned arrangement of tendon cells is a marker of tendon tissue morphology and the embodiment of its biological anisotropy. However, most of the hydrogels used for tendon tissue engineering do not present anisotropic structures. In this work, a magnetically-responsive nanocomposite hydrogel composed of collagen type I (COL I) and aligned iron oxide nanoparticles (IOPs) was investigated for potential application in tendon tissue engineering. COL I with a mixture of remotely aligned IOPs (A/IOPs) and human tendon stem/progenitor cells (COL I-A/IOPs-hTSPCs) was prepared and the alignment of IOPs was induced under a remote magnetic field. Following the gelation of COL I, a stable and anisotropic nanocomposite COL I-A/IOPs hydrogel was formed. In addition, hTSPCs embedded in COL I with random IOPs (COL I-R/IOPs-hTSPCs) and in pure COL I (COL I-hTSPCs) were used as control groups. Cell viability, proliferation, morphology, cell row formation, and alignment of IOPs and hTSPCs were evaluated over time. In addition, a comprehensive gene expression profile of 48 different genes, including tendon-related genes and lineage/cross-linking genes, was obtained by implementing designer quantitative RT-PCR plates. The hTSPCs morphology followed the orientation of the anisotropic COL I-A/IOPs hydrogel with increased row formation in comparison to pristine COL I and COL-R/IOPs. Moreover, higher proliferation rate and significant upregulation of tendon gene markers were measured in comparison to hTSPCs cultivated in the COL I-R/IOPs and COL I. Thus, we suggest that providing the cells with aligned focal contact points, namely the aligned IOPs, is sufficient to provoke an immense effect on the formation of aligned cell rows. Taken together, we report a novel strategy for directing stem cell behavior without the use of exogenous growth factors or pre-aligned COL I fibers, and propose that anisotropic nanocomposite hydrogels hold great potential for tendon tissue engineering applications.


Assuntos
Engenharia Tecidual , Alicerces Teciduais , Anisotropia , Humanos , Nanogéis , Células-Tronco , Tendões
6.
Biofabrication ; 13(1)2020 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-32977317

RESUMO

Morphogenesis, a complex process, ubiquitous in developmental biology and many pathologies, is based on self-patterning of cells. Spatial patterns of cells, organoids, or inorganic particles can be forced on demand using acoustic surface standing waves, such as the Faraday waves. This technology allows tuning of parameters (sound frequency, amplitude, chamber shape) under contactless, fast and mild culture conditions, for morphologically relevant tissue generation. We call this method Sound Induced Morphogenesis (SIM). In this work, we use SIM to achieve tight control over patterning of endothelial cells and mesenchymal stem cells densities within a hydrogel, with the endpoint formation of vascular structures. Here, we first parameterize our system to produce enhanced cell density gradients. Second, we allow for vasculogenesis after SIM patterning control and compare our controlled technology against state-of-the-art microfluidic culture systems, the latter characteristic of pure self-organized patterning and uniform initial density. Our sound-induced cell density patterning and subsequent vasculogenesis requires less cells than the microfluidic chamber. We advocate for the use of SIM for rapid, mild, and reproducible morphogenesis induction and further explorations in the regenerative medicine and cell therapy fields.


Assuntos
Células Endoteliais , Som , Hidrogéis , Morfogênese , Organoides
7.
Artigo em Inglês | MEDLINE | ID: mdl-32373603

RESUMO

3D Bioprinting (3DBP) technologies open many possibilities for the generation of highly complex cellularized constructs. Nano-biomaterials have been largely used in tissue engineering and regenerative medicine (TERM) for different purposes and functions depending on their intrinsic properties and how they have been presented in the biologic environment. Combination of bioprinting and nano-biomaterials paves the way for unexpected opportunities in the biofabrication scenario, by improving critical weakness of these manufacturing processes while enhancing their efficiency by spatially arranging nano-features. 3D organization of cells is fundamental for a successful design and maturation of native tissues. A critical challenge for the production of biological constructs is to support and guide cell growth toward their natural microenvironment, ensuring a harmonious presence of specific biochemical and biophysical cues to direct cell behavior. Also, precise arrays of stimuli need to be designed to induce stem cell differentiation toward specific tissues. Introducing nano-sized bioactive material can direct cell fate, playing a role in the differentiation process and leading to the biofabrication of functional structures. Nano-composite bio-ink can be used to generate cell instructive scaffolds or either directly printed with cells. In addition, the presence of nano-particles within 3D printed constructs can lead to control them through multiple external physical stimuli, representing an additional tool for healthcare applications. Finally, there is an emerging interest to create biological constructs having active properties, such as sensing, motion or shape modification. In this review, we highlight how introducing nano-biomaterials in bioprinting approaches leads to promising strategies for tissue regeneration.

8.
J Colloid Interface Sci ; 501: 185-191, 2017 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-28456102

RESUMO

The development of biocompatible collagen substrates able to conduct electric current along specific pathways represent an appealing issue in tissue engineering, since it is well known that electrical stimuli significantly affects important cell behaviour, such as proliferation, differentiation, directional migration, and, therefore, tissue regeneration. In this work, a cheap and easy approach was proposed to produce collagen-based films exhibiting enhanced electrical conductivity, through the simple manipulation of a weak external magnetic trigger. Paramagnetic iron oxide nanoparticles (NPs) capped by a biocompatible polyethylene-glycol coating were synthetized by a co-precipitation and solvothermic method and sprayed onto a collagen suspension. The system was then subjected to a static external magnetic field in order to conveniently tune NPs organization. Under the action of the external stimulus, NPs were induced to orient along the magnetic field lines, forming long-range aligned micropatterns within the collagen matrix. Drying of the substrate following water evaporation permanently blocked the magnetic architecture produced, thereby preserving NPs organization even after magnetic field removal. Electrical conductivity measurements clearly showed that the presence of such a magnetic framework endowed collagen with marked conductive properties in specific directions. The biocompatibility of the paramagnetic collagen films was also demonstrated by MTT cell cytotoxicity test.

9.
Nanomedicine ; 13(3): 1267-1277, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28115252

RESUMO

This study presents a simple and reproducible method of micropatterning the novel nanocomposite polymer (POSS-PCU) using a sacrificial phosphate glass fiber template for tendon tissue engineering applications. The diameters of the patterned scaffolds produced were dependent on the diameter of the glass fibers (15 µm) used. Scaffolds were tested for their physical properties and reproducibility using various microscopy techniques. For the first time, we show that POSS-PCU supports growth of human tenocytes cells. Furthermore, we show that cellular alignment, their biological function and expression of various tendon related proteins such as scleraxis, collagen I and III, tenascin-C are significantly elevated on the micropatterned polymer surfaces compared to flat samples. This study demonstrated a simple, reproducible method of micropatterning POSS-PCU nanocomposite polymer for novel tendon repair applications, which when provided with physical cues could help mimic the microenvironment of tenocytes cells.


Assuntos
Vidro/química , Nanocompostos/química , Fosfatos/química , Tendões/citologia , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Proliferação de Células , Células Cultivadas , Colágeno/análise , Colágeno/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Tendões/metabolismo , Molhabilidade
10.
Bioact Mater ; 2(4): 208-223, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29744431

RESUMO

The aim of the present study was the in vitro and in vivo analysis of a bi-layered 3D-printed scaffold combining a PLA layer and a biphasic PLA/bioglass G5 layer for regeneration of osteochondral defects in vivo Focus of the in vitro analysis was on the (molecular) weight loss and the morphological and mechanical variations after immersion in SBF. The in vivo study focused on analysis of the tissue reactions and differences in the implant bed vascularization using an established subcutaneous implantation model in CD-1 mice and established histological and histomorphometrical methods. Both scaffold parts kept their structural integrity, while changes in morphology were observed, especially for the PLA/G5 scaffold. Mechanical properties decreased with progressive degradation, while the PLA/G5 scaffolds presented higher compressive modulus than PLA scaffolds. The tissue reaction to PLA included low numbers of BMGCs and minimal vascularization of its implant beds, while the addition of G5 lead to higher numbers of BMGCs and a higher implant bed vascularization. Analysis revealed that the use of a bi-layered scaffold shows the ability to observe distinct in vivo response despite the physical proximity of PLA and PLA/G5 layers. Altogether, the results showed that the addition of G5 enables to reduce scaffold weight loss and to increase mechanical strength. Furthermore, the addition of G5 lead to a higher vascularization of the implant bed required as basis for bone tissue regeneration mediated by higher numbers of BMGCs, while within the PLA parts a significantly lower vascularization was found optimally for chondral regeneration. Thus, this data show that the analyzed bi-layered scaffold may serve as an ideal basis for the regeneration of osteochondral tissue defects. Additionally, the results show that it might be able to reduce the number of experimental animals required as it may be possible to analyze the tissue response to more than one implant in one experimental animal.

11.
Mater Sci Eng C Mater Biol Appl ; 38: 55-62, 2014 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-24656352

RESUMO

Achieving high quality 3D-printed structures requires establishing the right printing conditions. Finding processing conditions that satisfy both the fabrication process and the final required scaffold properties is crucial. This work stresses the importance of studying the outcome of the plasticizing effect of PEG on PLA-based blends used for the fabrication of 3D-direct-printed scaffolds for tissue engineering applications. For this, PLA/PEG blends with 5, 10 and 20% (w/w) of PEG and PLA/PEG/bioactive CaP glass composites were processed in the form of 3D rapid prototyping scaffolds. Surface analysis and differential scanning calorimetry revealed a rearrangement of polymer chains and a topography, wettability and elastic modulus increase of the studied surfaces as PEG was incorporated. Moreover, addition of 10 and 20% PEG led to non-uniform 3D structures with lower mechanical properties. In vitro degradation studies showed that the inclusion of PEG significantly accelerated the degradation rate of the material. Results indicated that the presence of PEG not only improves PLA processing but also leads to relevant surface, geometrical and structural changes including modulation of the degradation rate of PLA-based 3D printed scaffolds.


Assuntos
Ácido Láctico/química , Polietilenoglicóis/química , Polímeros/química , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Interferometria , Fenômenos Mecânicos , Microscopia de Força Atômica , Microscopia Eletrônica de Varredura , Poliésteres , Porosidade , Temperatura
12.
Acta Biomater ; 10(2): 613-22, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24211731

RESUMO

Recent studies have pointed towards a decisive role of inflammation in triggering tissue repair and regeneration, while at the same time it is accepted that an exacerbated inflammatory response may lead to rejection of an implant. Within this context, understanding and having the capacity to regulate the inflammatory response elicited by 3-D scaffolds aimed for tissue regeneration is crucial. This work reports on the analysis of the cytokine profile of human monocytes/macrophages in contact with biodegradable 3-D scaffolds with different surface properties, architecture and controlled pore geometry, fabricated by 3-D printing technology. Fabrication processes were optimized to create four different 3-D platforms based on polylactic acid (PLA), PLA/calcium phosphate glass or chitosan. Cytokine secretion and cell morphology of human peripheral blood monocytes allowed to differentiate on the different matrices were analyzed. While all scaffolds supported monocyte/macrophage adhesion and stimulated cytokine production, striking differences between PLA-based and chitosan scaffolds were found, with chitosan eliciting increased secretion of tumor necrosis factor (TNF)-α, while PLA-based scaffolds induced higher production of interleukin (IL)-6, IL-12/23 and IL-10. Even though the material itself induced the biggest differences, the scaffold geometry also impacted on TNF-α and IL-12/23 production, with chitosan scaffolds having larger pores and wider angles leading to a higher secretion of these pro-inflammatory cytokines. These findings strengthen the appropriateness of these 3-D platforms to study modulation of macrophage responses by specific parameters (chemistry, topography, scaffold architecture).


Assuntos
Quitosana/farmacologia , Inflamação/patologia , Ácido Láctico/farmacologia , Macrófagos/citologia , Monócitos/citologia , Polímeros/farmacologia , Alicerces Teciduais/química , Actinas/metabolismo , Forma Celular/efeitos dos fármacos , Células Cultivadas , Quitosana/química , Citocinas/metabolismo , Humanos , Ácido Láctico/química , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Microscopia Eletrônica de Varredura , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Poliésteres , Polímeros/química , Coloração e Rotulagem
13.
Organogenesis ; 9(4): 239-44, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-23959206

RESUMO

Rapid prototyping (RP), also known as additive manufacturing (AM), has been well received and adopted in the biomedical field. The capacity of this family of techniques to fabricate customized 3D structures with complex geometries and excellent reproducibility has revolutionized implantology and regenerative medicine. In particular, nozzle-based systems allow the fabrication of high-resolution polylactic acid (PLA) structures that are of interest in regenerative medicine. These 3D structures find interesting applications in the regenerative medicine field where promising applications including biodegradable templates for tissue regeneration purposes, 3D in vitro platforms for studying cell response to different scaffolds conditions and for drug screening are considered among others. Scaffolds functionality depends not only on the fabrication technique, but also on the material used to build the 3D structure, the geometry and inner architecture of the structure, and the final surface properties. All being crucial parameters affecting scaffolds success. This Commentary emphasizes the importance of these parameters in scaffolds' fabrication and also draws the attention toward the versatility of these PLA scaffolds as a potential tool in regenerative medicine and other medical fields.


Assuntos
Ácido Láctico/química , Polímeros/química , Medicina Regenerativa/instrumentação , Medicina Regenerativa/métodos , Alicerces Teciduais/química , Animais , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/ultraestrutura , Poliésteres , Impressão , Ratos , Propriedades de Superfície
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...