Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
ACS Appl Mater Interfaces ; 11(16): 14548-14559, 2019 Apr 24.
Article in English | MEDLINE | ID: mdl-30943004

ABSTRACT

Osteosarcoma is one of the most common metastatic bone cancers, which results in significant morbidity and mortality. Unfolding of effectual therapeutic strategies against osteosarcoma is impeded because of the absence of adequate animal models, which can truly recapitulate disease biology of humans. Tissue engineering provides an opportunity to develop physiologically relevant, reproducible, and tunable in vitro platforms to investigate the interactions of osteosarcoma cells with its microenvironment. Adipose-derived stem cells (ASCs) are detected adjacent to osteosarcoma masses and are considered to have protumor effects. Hence, the present study focuses on investigating the role of reactive ASCs in formation of spheroids of osteosarcoma cells (Saos 2) within a three-dimensional (3D) niche, which is created using gellan gum (GG)-silk fibroin. By modifying the blending ratio of GG-silk, the optimum stiffness of the resultant hydrogels such as GG and GG75: S25 is obtained for cancer spheroid formation. This work indicates that the co-existence of cancer and stem cells can form a spheroid, the hallmark of cancer, only in particular microenvironment stiffness. The incorporation of fibrillar silk fibroin within the hydrophilic network of GG in GG75: S25 spongy-like hydrogels closely mimics the stiffness of commercially established cancer biomaterials (e.g., Matrigel, HyStem). The GG75: S25 hydrogel maintains the metabolically active construct for a longer time with elevated expression of osteopontin, osteocalcin, RUNX 2, and bone sialoprotein genes, the biomarkers of osteosarcoma, compared to GG. The GG75: S25 construct also exhibits intense alkaline phosphatase expression in immunohistochemistry compared to GG, indicating itspotentiality to serve as biomimetic niche to model osteosarcoma. Taken together, the GG-silk fibroin-blended spongy-like hydrogel is envisioned as an alternative low-cost platform for 3D cancer modeling.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/metabolism , Bone Neoplasms/metabolism , Fibroins/chemistry , Hydrogels/chemistry , Models, Biological , Osteosarcoma/metabolism , Spheroids, Cellular/metabolism , Stem Cells/metabolism , Tissue Scaffolds/chemistry , Adipocytes/pathology , Adipose Tissue/pathology , Bone Neoplasms/pathology , Cell Line, Tumor , Humans , Osteosarcoma/pathology , Spheroids, Cellular/pathology , Stem Cells/pathology , Tissue Engineering
2.
Biomaterials ; 149: 98-115, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29024838

ABSTRACT

Most cancer patients do not die from the primary tumor but from its metastasis. Current in vitro and in vivo cancer models are incapable of satisfactorily predicting the outcome of various clinical treatments on patients. This is seen as a serious limitation and efforts are underway to develop a new generation of highly predictive cancer models with advanced capabilities. In this regard, organ-on-chip models of cancer metastasis emerge as powerful predictors of disease progression. They offer physiological-like conditions where the (hypothesized) mechanistic determinants of the disease can be assessed with ease. Combined with high-throughput characteristics, the employment of organ-on-chip technology would allow pharmaceutical companies and clinicians to test new therapeutic compounds and therapies. This will permit the screening of a large battery of new drugs in a fast and economic manner, to accelerate the diagnosis of the disease in the near future, and to test personalized treatments using cells from patients. In this review, we describe the latest advances in the field of organ-on-chip models of cancer metastasis and their integration with advanced imaging, screening and biosensing technologies for future precision medicine applications. We focus on their clinical applicability and market opportunities to drive us forward to the next generation of tumor models for improved cancer patient theranostics.


Subject(s)
Lab-On-A-Chip Devices , Neoplasm Metastasis/diagnosis , Neoplasm Metastasis/therapy , Precision Medicine/methods , Theranostic Nanomedicine/methods , Tissue Array Analysis , High-Throughput Screening Assays , Humans , Microfluidics , Precision Medicine/instrumentation
3.
Biofabrication ; 7(1): 015008, 2015 Jan 21.
Article in English | MEDLINE | ID: mdl-25607240

ABSTRACT

Degeneration of the intervertebral disc (IVD) represents a significant musculoskeletal disease burden. Tissue engineering has proposed several strategies comprising the use of biodegradable materials to prepare scaffolds that can present mechanical properties similar to those of native IVD tissues. However, this might be insufficient, since the patient's intervertebral space geometry must be replicated to allow for appropriate implant fixation and integration. Herein, we propose the use of reverse engineering and rapid prototyping techniques with the goal of preparing custom-tailored annulus fibrosus scaffolds; these techniques have previously been applied to rabbit models. The IVD reverse-engineered architecture was obtained by means of microcomputed tomography acquisition and three-dimensional modelling, resulting in a computer-aided design (CAD) that replicates the original rabbit IVD. Later, a fused deposition-modelling three-dimensional printer was used to produce the scaffolds with different geometries provided by the CAD, using polycaprolactone (PCL) with 100% infill density. The microstructure of the PCL scaffolds was investigated by scanning electron microscopy (SEM), which allowed us to observe an adequate fusion adhesion between the layers. The SEM images revealed that, up to the point of moderate resolution, the porosities manually designed in the CAD model were successfully replicated. The PCL scaffolds' three-dimensional architecture was also assessed by means of microcomputed tomography analysis. Compressive stiffness was determined using a mechanical testing system. Results showed higher values than those of human IVDs (5.9-6.7 kN mm(-1) versus 1.2 kN mm(-1), respectively). In vitro studies were performed to investigate the possible cytotoxicity of the polycaprolactone scaffolds' leachables. The results showed that the custom-tailored PCL scaffolds do not have any deleterious cytotoxic effect over annulus fibrosus cells or the mouse lung fibroblast's cell line. This study proposed a simple, rapid, and low-cost strategy to fabricate custom-tailored annulus fibrosus scaffolds. In the future, this strategy might be used in association with nucleus pulposus regeneration strategies to facilitate the development of tissue-engineered total disc replacement implants specific to each patient, with a goal of full IVD regeneration.


Subject(s)
Polyesters/pharmacology , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Total Disc Replacement , Animals , Cell Death/drug effects , Cell Line , Computer Simulation , Imaging, Three-Dimensional , Intervertebral Disc/diagnostic imaging , Intervertebral Disc/drug effects , Intervertebral Disc/ultrastructure , Lumbar Vertebrae/diagnostic imaging , Lumbar Vertebrae/drug effects , Materials Testing , Mice , Porosity , Rabbits , Radiography
4.
J Tissue Eng Regen Med ; 9(6): 714-23, 2015 Jun.
Article in English | MEDLINE | ID: mdl-24155167

ABSTRACT

Paracrine signalling from chondrocytes has been reported to increase the synthesis and expression of cartilage extracellular matrix (ECM) by stem cells. The use of conditioned medium obtained from chondrocytes for stimulating stem cells chondrogenic differentiation may be a very interesting alternative for moving into the clinical application of these cells, as chondrocytes could be partially replaced by stem cells for this type of application. In the present study we aimed to achieve chondrogenic differentiation of two different sources of stem cells using conditioned medium, without adding growth factors. We tested both human bone marrow-derived mesenchymal stem cells (hBSMCs) and human Wharton's jelly-derived stem cells (hWJSCs). Conditioned medium obtained from a culture of human articular chondrocytes was used to feed the cells during the experiment. Cultures were performed in previously produced three-dimensional (3D) scaffolds, composed of a blend of 50:50 chitosan:poly(butylene succinate). Both types of stem cells were able to undergo chondrogenic differentiation without the addition of growth factors. Cultures using hWJSCs showed significantly higher GAGs accumulation and expression of cartilage-related genes (aggrecan, Sox9 and collagen type II) when compared to hBMSCs cultures. Conditioned medium obtained from articular chondrocytes induced the chondrogenic differentiation of MSCs and ECM formation. Obtained results showed that this new strategy is very interesting and should be further explored for clinical applications.


Subject(s)
Cell Differentiation/drug effects , Chondrocytes/cytology , Chondrogenesis/drug effects , Culture Media, Conditioned/pharmacology , Mesenchymal Stem Cells/cytology , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cartilage, Articular/cytology , Cell Differentiation/genetics , Cells, Cultured , Chondrocytes/drug effects , Chondrocytes/metabolism , Chondrogenesis/genetics , DNA/metabolism , Extracellular Matrix/metabolism , Glycosaminoglycans/metabolism , Humans , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Real-Time Polymerase Chain Reaction , Staining and Labeling
5.
Mater Sci Eng C Mater Biol Appl ; 42: 615-21, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25063161

ABSTRACT

The development of tissue engineering field entails the creation of micro/nanoscale features for cellular alignment and biocompatibility improvement. As replication techniques, hot embossing and soft lithography can be used to produce micro/nanoscale features on biodegradable membranes. Subsequently the generation of 3D scaffolds can be done by means of assembling techniques. Using the described techniques, high resolution of features, as small as 5 nm, can be achieved. Nevertheless membrane assembling must be fully studied to avoid feature fluctuations and even collapse of the scaffold. The present review focuses on the state-of-the-art in the replication techniques used to create micro/nanoscale features on biodegradable polymers and assembling approaches to construct scaffolds with the aim of exploring existing advances and limitations of the reported methods.


Subject(s)
Cell Culture Techniques/instrumentation , Nanotechnology/methods , Tissue Engineering/instrumentation , Tissue Scaffolds , Printing, Three-Dimensional
6.
J Biomater Appl ; 28(9): 1430-42, 2014 May.
Article in English | MEDLINE | ID: mdl-24255003

ABSTRACT

To understand the role of chitosan in chitosan-poly(butylene succinate) scaffolds (50% wt), 50%, 25%, and 0% of chitosan were used to produce different scaffolds. These scaffolds were in vitro seeded and cultured with human bone marrow stromal cells in osteogenic conditions, revealing that higher percentage of chitosan showed enhanced cell viability over time, adhesion, proliferation, and osteogenic differentiation. Scaffolds were also implanted in cranial defects and iliac submuscular region in Wistar rats, and the results evidenced that chitosan-containing scaffolds displayed mild inflammatory response and good integration with surrounding tissues, showed by connective tissue colonization and the presence of new blood vessels. Scaffolds without chitosan-evidenced necrotic tissue in scaffolds' interior, proving that chitosan exerts a positive effect over cell behavior and displays a milder host inflammatory response in vivo.


Subject(s)
Cell Differentiation , Inflammation/pathology , Mesenchymal Stem Cells/cytology , Osteogenesis , Tissue Scaffolds , Animals , Base Sequence , DNA Primers , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction , X-Ray Microtomography
7.
Biomed Microdevices ; 16(1): 69-78, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24122322

ABSTRACT

The use of bottom-up approaches in tissue engineering applications is advantageous since they enable the combination of various layers that could be made from different materials and/or incorporate different biochemical cues. Regarding the complex structure and the vascular system of the bone tissue, the aim of this study was to develop an innovative bottom-up approach that allows the construction of 3D biodegradable scaffolds from 2D microfabricated membranes with precise shape, pore size and porosity. For that purpose, poly (caprolactone) (PCL) and starch ­ poly (caprolactone) (SPCL (30 % starch)) blended sheets were used as substrates to produce the microfabricated membranes using micro hotembossing. The use of this micro fabrication process allowed accurately imprinting micropillars and microholes in reproducible way. The assembling of the microfabricated membranes was performed using an easy, highly reproducible and inexpensive approach based on its successive stacking. Additionaly, the suitability of the microfabricated membranes to support the attachment and the cytoskeletal organization of human bone marrow stem cells (hBMSCs), macrovascular endothelial cells and osteoblasts derived from hBMSCs was demonstrated. Furthermore, hBMSCs proliferated and maintained the expression of the stromal progenitor marker STRO-1 when cultured on both PCL and SPCL microfabricated membranes. The proposed methodology constitutes a promising alternative to the traditional processing methods used to prepare tissue engineering scaffolds.


Subject(s)
Bone and Bones/chemistry , Microtechnology/methods , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Cell Adhesion , Cell Differentiation , Cells, Cultured , Human Umbilical Vein Endothelial Cells , Humans , Immunohistochemistry , Mesenchymal Stem Cells/chemistry , Microscopy, Electron, Scanning , Osteoblasts/chemistry , Polymers/chemistry , Porosity
8.
J Tissue Eng Regen Med ; 6(1): 1-11, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21294275

ABSTRACT

The first stem cells considered for the reconstruction of bone were bone marrow mesenchymal stem cells (BMSCs). Subsequently, cells with similar marker expression panel and differentiation potential were found in new sources of cells, such as adipose tissue. This source of stem cells has a promising future in tissue-engineering applications, considering the abundance of this tissue in the human body, the easy harvesting and the high number of stem cells that are available from such a small amount of tissue. The isolation of the adipose stem cells is generally performed by means of enzymatic digestion of the tissues, followed by a natural selection of the stem cells based on their capacity to adhere to the culture flasks, leading to a quite heterogeneous population. This constitutes a major drawback for the use of these cells, since the heterogeneity of the cell culture obtained can compromise their proliferation and differentiation potential. In the present study we have analysed the in vitro and in vivo behaviour of two selected subpopulations with high osteogenic potential. For this purpose, ASCs(CD29+) and ASCs (STRO-1+)subpopulations were isolated and in vitro cultured onto a biodegradable polymeric scaffold, using osteogenic medium, before implantation in a nude mice model. The biodegradable polymeric scaffold used is a fibre-mesh structure based on a blend of starch and polycaprolatone (SPCL) that has been successfully used in several bone tissue-engineering studies. The implanted ASCs-scaffold constructs promoted the formation of new bone tissue in nude mice. However, the results obtained show differences in the behaviour of the two ASCs subpopulations under study, particularly regarding their potential to differentiate into the osteogenic lineage, and allowed the indentification of ASCs (STRO-1+) as the best subpopulation for bone tissue-engineering applications.


Subject(s)
Adipose Tissue/cytology , Osteogenesis/physiology , Stem Cells/cytology , Tissue Engineering/methods , Adipocytes/cytology , Animals , Female , Green Fluorescent Proteins/chemistry , Humans , Immunohistochemistry/methods , Mice , Mice, Nude , Regenerative Medicine/methods , Tissue Scaffolds , X-Ray Microtomography/methods
9.
J Tissue Eng Regen Med ; 6(1): 21-8, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21312336

ABSTRACT

Tissue engineering sustains the need of a three-dimensional (3D) scaffold to promote the regeneration of tissues in volume. Usually, scaffolds are seeded with an adequate cell population, allowing their growth and maturation upon implantation in vivo. Previous studies obtained by our group evidenced significant growth patterns and osteogenic differentiation of human bone marrow mesenchymal stem cells (hBMSCs) when seeded and cultured on melt-based porous chitosan fibre mesh scaffolds (cell constructs). Therefore, it is crucial to test the in vivo performance of these in vitro 3D cell constructs. In this study, chitosan-based scaffolds were seeded and cultured in vitro with hBMSCs for 3 weeks under osteogenic stimulation conditions and analysed for cell adhesion, proliferation and differentiation. Implantation of 2 weeks precultured cell constructs in osteogenic culture conditions was performed into critical cranial size defects in nude mice. The objective of this study was to verify the scaffold integration and new bone formation. At 8 weeks of implantation, scaffolds were harvested and prepared for micro-computed tomography (µCT) analysis. Retrieved implants showed good integration with the surrounding tissue and significant bone formation, more evident for the scaffolds cultured and implanted with human cells. The results of this work demonstrated that chitosan-based scaffolds, besides supporting in vitro proliferation and osteogenic differentiation of hBMSCs, induced bone formation in vivo. Thus, their osteogenic potential in orthotopic location in immunodeficient mice was validated, evidencing good prospects for their use in bone tissue-engineering therapies.


Subject(s)
Bone Marrow Cells/cytology , Butylene Glycols/chemistry , Chitosan/chemistry , Polymers/chemistry , Stromal Cells/cytology , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Alkaline Phosphatase/metabolism , Animals , Bone and Bones/cytology , Bone and Bones/metabolism , Cell Survival , Cells, Cultured , Humans , Mice , Mice, Nude , Phenotype , Porosity , X-Ray Microtomography/methods
10.
J Tissue Eng Regen Med ; 5(9): 722-32, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21953870

ABSTRACT

Native articular cartilage is subjected to synovial fluid flow during normal joint function. Thus, it is believed that the morphogenesis of articular cartilage may be positively regulated by the application of similar stimulation in vitro. In the present study, the effect of fluid flow over the chondrogenic differentiation of human bone marrow-derived mesenchymal stem cells (hBM-MSCs) was investigated. We intended to find out whether the shear stress caused by perfusion of the medium through the constructs was capable of augmenting the differentiation process. Human BMSCs were isolated from bone marrow aspirates and were characterized by flow cytometry. After expansion, hBM-MSCs were seeded statically onto fibre mesh scaffolds, consisting of a blend of 50:50 chitosan:poly(butylene terephthalate adipate) (CPBTA). Constructs were cultured in a flow-perfusion bioreactor for 28 days, using complete medium for chondrogenesis supplemented by TGFß3. An enhanced ECM deposition and collagen type II production was observed in the bioreactor samples when compared to the static controls. Moreover, it was observed that hBM-MSCs, in static cultures, take longer to differentiate. ECM accumulation in these samples is lower than in the bioreactor sections, and there is a significant difference in the expression of collagen type I. We found that the flow-induced shear stress has a beneficial effect on the chondrogenic differentiation of hMSCs.


Subject(s)
Bioreactors , Bone Marrow Cells/cytology , Cell Differentiation/drug effects , Chitosan/pharmacology , Chondrogenesis/drug effects , Mesenchymal Stem Cells/cytology , Tissue Engineering/instrumentation , Biomarkers/metabolism , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Bone Marrow Cells/ultrastructure , Cell Separation , Cell Shape/drug effects , Cells, Cultured , Chondrogenesis/genetics , Collagen Type I/metabolism , Collagen Type II/metabolism , DNA/metabolism , Flow Cytometry , Humans , Immunohistochemistry , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/ultrastructure , Perfusion , Polyesters/pharmacology , Real-Time Polymerase Chain Reaction , Rheology/drug effects , Staining and Labeling , Tissue Scaffolds/chemistry
11.
Acta Biomater ; 6(3): 1149-57, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19788942

ABSTRACT

Naturally derived polymers have been extensively used in scaffold production for cartilage tissue engineering. The present work aims to evaluate and characterize extracellular matrix (ECM) formation in two types of chitosan-based scaffolds, using bovine articular chondrocytes (BACs). The influence of these scaffolds' porosity, as well as pore size and geometry, on the formation of cartilagineous tissue was studied. The effect of stirred conditions on ECM formation was also assessed. Chitosan-poly(butylene succinate) (CPBS) scaffolds were produced by compression moulding and salt leaching, using a blend of 50% of each material. Different porosities and pore size structures were obtained. BACs were seeded onto CPBS scaffolds using spinner flasks. Constructs were then transferred to the incubator, where half were cultured under stirred conditions, and the other half under static conditions for 4 weeks. Constructs were characterized by scanning electron microscopy, histology procedures, immunolocalization of collagen type I and collagen type II, and dimethylmethylene blue assay for glycosaminoglycan (GAG) quantification. Both materials showed good affinity for cell attachment. Cells colonized the entire scaffolds and were able to produce ECM. Large pores with random geometry improved proteoglycans and collagen type II production. However, that structure has the opposite effect on GAG production. Stirred culture conditions indicate enhancement of GAG production in both types of scaffold.


Subject(s)
Biocompatible Materials/chemistry , Butylene Glycols/chemistry , Cartilage/growth & development , Chitosan/chemistry , Chondrocytes/physiology , Extracellular Matrix/physiology , Polymers/chemistry , Tissue Scaffolds/chemistry , Absorption , Animals , Biomimetic Materials/chemistry , Cartilage/cytology , Cattle , Cell Culture Techniques/methods , Cells, Cultured , Chondrocytes/cytology , Crystallization/methods , Extracellular Matrix Proteins/metabolism , Materials Testing , Particle Size , Porosity , Surface Properties , Tissue Engineering/methods
12.
J Biomed Mater Res A ; 91(2): 489-504, 2009 Nov.
Article in English | MEDLINE | ID: mdl-18985771

ABSTRACT

Blends of chitosan and synthetic aliphatic polyesters (polybutylene succinate, polybutylene succinate adipate, polycaprolactone, and polybutylene terepthalate adipate) were compounded with and without hydroxyapatite, a bioactive mineral filler known to enhance osteoconduction. The blends and composites were compression molded with two different granulometric salt sizes (63-125 microm and 250-500 microm) having different levels of salt content (60, 70, and 80%) by weight. By leaching the salt particles, it was possible to produce porous scaffolds with distinct morphologies. The relationship between scaffold morphology and mechanical properties was evaluated using scanning electron microscopy, microcomputed tomography, compression testing, differential scanning calorimetry, small-angle X-ray scattering (SAXS), and wide-angle X-ray scattering. The produced scaffolds are characterized by having different morphologies depending on the average particle size and the amount of NaCl used. Specimens with higher porosity level have a less organized pore structure but increased interconnectivity of the pores. The stress-strain curve under compression displayed a linear elasticity followed by a plateau whose characteristics depend on the scaffold polymer composition. A decrease in the salt particle size used to create the porosity caused in general a decrease in the mechanical properties of the foams. Composites with hydroxyapatite had a sharp reduction in yield stress, modulus, and strain at break. The melting temperature decreased with increased chitosan content. SAXS results indicate no preferential crystalline orientation in the scaffolds. Cytotoxicity evaluation were carried out using standard tests (accordingly to ISO/EN 10993 part 5 guidelines), namely MTS test with a 24-h extraction period, revealing that L929 cells had comparable metabolic activities to that obtained for the negative control.


Subject(s)
Chitosan/chemistry , Polyesters/chemistry , Tissue Scaffolds/chemistry , Animals , Calorimetry, Differential Scanning , Cell Line , Cell Survival , Crystallization , Fibroblasts/cytology , Materials Testing , Porosity , Rats , Stress, Mechanical , Tensile Strength
SELECTION OF CITATIONS
SEARCH DETAIL
...