Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Biotechnol Bioeng ; 117(10): 3108-3123, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32557561

RESUMEN

The development of a minimally invasive, robust, and inexpensive technique that permits real-time monitoring of cell responses on biomaterial scaffolds can improve the eventual outcomes of scaffold-based tissue engineering strategies. Towards establishing correlations between in situ biological activity and cell fate, we have developed a comprehensive workflow for real-time volumetric imaging of spatiotemporally varying cytosolic calcium oscillations in pure microglial cells cultured on electrospun meshes. Live HMC3 cells on randomly oriented electrospun fibers were stained with a fluorescent dye and imaged using a laser scanning confocal microscope. Resonance scanning provided high-resolution in obtaining the time-course of intracellular calcium levels without compromising spatial and temporal resolution. Three-dimensional reconstruction and depth-coding enabled the visualization of cell location and intracellular calcium levels as a function of sample thickness. Importantly, changes in cell morphology and in situ calcium spiking were quantified in response to a soluble biochemical cue and varying matrix architectures (i.e., randomly oriented and aligned fibers). Importantly, raster plots generated from spiking data revealed calcium signatures specific to culture conditions. In the future, our approach can be used to elucidate correlations between calcium signatures and cell phenotype/activation, and facilitate the rational design of scaffolds for biomedical applications.


Asunto(s)
Señalización del Calcio , Calcio/metabolismo , Citosol/metabolismo , Microglía/metabolismo , Microscopía Confocal/métodos , Ingeniería de Tejidos/métodos , Andamios del Tejido/química , Materiales Biocompatibles/química , Células Cultivadas , Humanos , Imagenología Tridimensional/métodos , Microglía/citología , Microscopía Electrónica de Rastreo , Nanofibras/química
2.
Polymer (Guildf) ; 77: 227-238, 2015 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-29332957

RESUMEN

Poly(ethylene glycol) diacrylate (PEGDA) hydrogels are widely used in biotechnology due to their in situ crosslinking capacity and tunable physical properties. However, as with all single component hydrogels, the modulus of PEGDA networks cannot be tailored independently of mesh size. This interdependence places significant limitations on their use for defined, 3D cell-microenvironment studies and for certain controlled release applications. The incorporation of secondary reactive species (SRS) into PEGDA hydrogels has previously been shown to allow the identification of up to 6 PEGDA hydrogel formulations for which distinct moduli can be obtained at consistent average mesh size (or vice versa). However, the modulus and mesh size ranges which can be probed by these formulations are quite restricted. This work presents an in-depth study of SRS incorporation into PEGDA hydrogels, with the goal of expanding the space for which "decoupled" examination of modulus and mesh size effects is achievable. Towards this end, over 100 PEGDA hydrogels containing either N-vinyl pyrrolidone or star PEG-tetraacrylate as SRS were characterized. To our knowledge, this is the first study to demonstrate that SRS incorporation allows for the identification of a number of modulus ranges that can be probed at consistent average mesh size (or vice versa).

3.
Biotechnol Bioeng ; 111(12): 2549-59, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24898875

RESUMEN

Although bone-patellar tendon-bone (B-PT-B) autografts are the gold standard for repair of anterior cruciate ligament ruptures, they suffer from drawbacks such as donor site morbidity and limited supply. Engineered tissues modeled after B-PT-B autografts are promising alternatives because they have the potential to regenerate connective tissue and facilitate osseointegration. Towards the long-term goal of regenerating ligaments and their bony insertions, the objective of this study was to construct 2D meshes and 3D cylindrical composite scaffolds - possessing simultaneous region-wise differences in fiber orientation, diameter, chemistry and mechanical properties - by electrospinning two different polymers from off-set spinnerets. Using a dual drum collector, 2D meshes consisting of an aligned polycaprolactone (PCL) fiber region, randomly oriented poly(lactide-co-glycolide) (PLGA) fiber region and a transition region (comprised of both PCL and PLGA fibers) were prepared, and region-wise differences were confirmed by microscopy and tensile testing. Bone marrow stromal cells (BMSCs) cultured on these meshes exhibited random orientations and low aspect ratios on the random PLGA regions, and high aspect ratios and alignment on the aligned PCL regions. Next, meshes containing an aligned PCL region flanked by two transition regions and two randomly oriented PLGA regions were prepared and processed into 3D cylindrical composite scaffolds using an interpenetrating photo-crosslinkable polyethylene glycol diacrylate hydrogel to recapitulate the shape of B-PT-B autografts. Tensile testing indicated that cylindrical composites were mechanically robust, and eventually failed due to stress concentration in the aligned PCL region. In summary, this study demonstrates a process to fabricate electrospun meshes possessing region-wise differences in properties that can elicit region-dependent cell responses, and be readily processed into scaffolds with the shape of B-PT-B autografts.


Asunto(s)
Huesos/citología , Técnicas Electroquímicas/métodos , Ligamentos/citología , Ingeniería de Tejidos/métodos , Andamios del Tejido , Animales , Péptidos Catiónicos Antimicrobianos/química , Células de la Médula Ósea , Células Cultivadas , Diseño de Equipo , Femenino , Poliésteres/química , Ratas , Resistencia a la Tracción
4.
Int J Pharm ; 651: 123768, 2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38176477

RESUMEN

Chronic inflammation is a leading cause of neurodegeneration and vision loss in hyperglycemia-associated conditions such as diabetic retinopathy. Corticosteroid injections are widely used for treatment but suffer from limitations such as rapid drug clearance, short drug half-lives and frequent administration. While drug release from biomaterial carriers can overcome these shortcomings, evaluating the combined effects of corticosteroids and polymeric matrices under hyperglycemic stress is an important step towards aiding translation. In this study, we investigated the effects of dexamethasone (DEX) and electrospun mesh combination on primary human mixed retinal cells under normal and hyperglycemic culture conditions. DEX-incorporated poly(lactide-co-glycolide) (PLGA) meshes were prepared and characterized for architecture, chemistry, drug distribution and in vitro release. The meshes exhibited cumulative in vitro drug release of 39.5 % over 2 months at a near constant rate. Under normal culture conditions, DEX-PLGA meshes promoted significantly higher viability of mixed retinal cells than the control groups but without adverse phenotypic activation. Under hyperglycemic conditions, DEX supplementation resulted in higher viability than the control, although the highest viability was achieved only when DEX was added to cells cultured on PLGA fibers. The combination of DEX and PLGA fibers also promoted higher mRNA expression of the antioxidant GSH under hyperglycemia. Importantly, the largest reduction in the production of pro-inflammatory cytokines viz., MMP-9, IL-6, IL-8 and VEGF-R1 was observed for the DEX and PLGA combination. Our study reveals a combined effect of DEX and electrospun fibers in combating hyperglycemia-driven pro-inflammatory responses, which can aid the development of DEX-loaded electrospun implants for diabetes-driven retinal conditions.


Asunto(s)
Hiperglucemia , Mallas Quirúrgicas , Humanos , Materiales Biocompatibles , Polímeros , Dexametasona , Hiperglucemia/tratamiento farmacológico
5.
Acta Biomater ; 163: 228-247, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-35675893

RESUMEN

Electrospun fibrous meshes are widely used for tissue repair due to their ability to guide a host of cell responses including phenotypic differentiation and tissue maturation. A critical factor determining the eventual biological outcomes of mesh-based regeneration strategies is the early innate immune response following implantation. The natural healing process involves a sequence of tightly regulated, temporally varying and delicately balanced pro-/anti-inflammatory events which together promote mesh integration with host tissue. Matrix designs that do not account for the immune milieu can result in dysregulation, chronic inflammation and fibrous capsule formation, thus obliterating potential therapeutic outcomes. In this review, we provide systematic insights into the effects of specific fiber/mesh properties and mechanical stimulation on the responses of early innate immune modulators viz., neutrophils, monocytes and macrophages. We identify matrix characteristics that promote anti-inflammatory immune phenotypes, and we correlate such responses with pro-regenerative in vivo outcomes. We also discuss recent advances in 3D fabrication technologies, bioactive functionalization approaches and biomimetic/bioinspired immunomodulatory mesh design strategies for tissue repair and wound healing. The mechanobiological insights and immunoregulatory strategies discussed herein can help improve the translational outcomes of fiber-based regeneration. STATEMENT OF SIGNIFICANCE: The crucial role played by immune cells in promoting biomaterial-based tissue regeneration is being increasingly recognized. In this review focusing on the interactions of innate immune cells with electrospun fibrous meshes, we systematically elucidate the effects of the fiber microenvironment and mechanical stimulation on biological responses, and build upon these insights to inform the rational design of immunomodulatory meshes for effective tissue repair. We discuss state-of-the-art fabrication methods and mechanobiological advances that permit the orchestration of temporally controlled phenotypic switches in immune cells during different phases of healing. The design strategies discussed herein can also be leveraged to target several complex autoimmune and inflammatory diseases.


Asunto(s)
Inmunidad Innata , Mallas Quirúrgicas , Macrófagos , Cicatrización de Heridas , Antiinflamatorios
6.
ACS Biomater Sci Eng ; 8(1): 1-15, 2022 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-34958569

RESUMEN

Functionalization of electrospun meshes with growth factors (GFs) is a common strategy for guiding specific cell responses in tissue engineering. GFs can exert their intended biological effects only when they retain their bioactivity and can be subsequently delivered in a temporally controlled manner. However, adverse processing conditions encountered in electrospinning can potentially disrupt GFs and diminish their biological efficacy. Further, meshes prepared using conventional approaches often promote an initial burst and rely solely on intrinsic fiber properties to provide extended release. Sequential delivery of multiple GFs─a strategy that mimics the natural tissue repair cascade─is also not easily achievable with traditional fabrication techniques. These limitations have hindered the effective use and translation of mesh-based strategies for tissue repair. An attractive alternative is the use of carrier vehicles (e.g., nanoparticles, microspheres) for GF incorporation into meshes. This review presents advances in the development of particle-integrated electrospun composites for safe and effective delivery of GFs. Compared to traditional approaches, we reveal how particles can protect GF activity, permit the incorporation of multiple GFs, decouple release from fiber properties, help achieve spatiotemporal control over delivery, enhance surface bioactivity, exert independent biological effects, and augment matrix mechanics. In presenting innovations in GF functionalization and composite engineering strategies, we also discuss specific in vitro and in vivo biological effects and their implications for diverse tissue engineering applications.


Asunto(s)
Sistemas de Liberación de Medicamentos , Nanopartículas , Péptidos y Proteínas de Señalización Intercelular , Ingeniería de Tejidos , Cicatrización de Heridas
7.
Biomed Mater ; 16(4)2021 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-34116516

RESUMEN

In this study, we offer new insights into the contrasting effects of electrospun fiber orientation on microglial polarization under normoxia and hypoxia, and establish for the first time, the intrinsically protective roles of electrospun meshes against hypoxia-induced microglial responses. First, resting microglia were cultured under normoxia on poly(caprolactone) fibers possessing two distinctly different fiber orientations. Matrix-guided differences in cell shape/orientation and differentially expressed Rho GTPases (RhoA, Rac1, Cdc42) were well-correlated with the randomly oriented fibers inducing a pro-inflammatory phenotype and the aligned fibers sustaining a resting phenotype. Upon subsequent hypoxia induction, both sets of meshes offered protection from hypoxia-induced damage by promoting a radical phenotypic switch and beneficially altering the M2/M1 ratio to different extents. Compared to 2D hypoxic controls, meshes significantly suppressed the expression of pro-inflammatory markers (IL-6, TNF-α) and induced drastically higher expression of anti-inflammatory (IL-4, IL-10, VEGF-189) and neuroprotective (Nrf-2) markers. Consistent with this M2 polarization, the expression of Rho GTPases was significantly lower in the mesh groups under hypoxia compared to normoxic culture. Moreover, meshes-particularly with aligned fibers-promoted higher cell viability, suppressed caspase 3/8 and LC-3 expression and promoted LAMP-1 and LAMP-2 expression, which suggested the mitigation of apoptotic/autophagic cell death via a lysosomal membrane-stabilization mechanism. Notably, all protective effects under hypoxia were observed in the absence of additional soluble cues. Our results offer promise for leveraging the intrinsic therapeutic potential of electrospun meshes in degenerative diseases where microglial dysfunction, hypoxia and inflammation are implicated.


Asunto(s)
Materiales Biocompatibles , Hipoxia de la Célula/fisiología , Polaridad Celular , Microglía/citología , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Línea Celular , Polaridad Celular/efectos de los fármacos , Polaridad Celular/fisiología , Citocinas/metabolismo , Técnicas Electroquímicas , Humanos , Inflamación/metabolismo , Microglía/metabolismo , Oxígeno/metabolismo , Proteínas de Unión al GTP rho/metabolismo
8.
ACS Omega ; 5(26): 15865-15876, 2020 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-32656407

RESUMEN

Despite advances in the development of degradable polymers for drug delivery, effective translation of drug-loaded materials is often hindered due to a poor understanding of matrix property combinations that promote controlled and sustained release. In this study, we investigated the influence of dominant factors on the release of a hydrophobic glucocorticoid dexamethasone (DEX) from electrospun meshes. Polycaprolactone meshes released 98% of the drug within 24 h, while poly(l-lactide) meshes exhibited negligible release even after 28 days despite both polymers being slow-degrading. Differences in drug-polymer interactions and drug-polymer miscibility-but neither matrix degradation nor differences in bulk hydrophobicity-influenced DEX release from these semi-crystalline matrices. Poly(d,l-lactide-co-glycolide) 50:50 meshes possessing two different fiber diameters exhibited a sequential burst and sustained release, while poly(d,l-lactide-co-glycolide) 85:15 meshes cumulatively released 26% drug in a controlled manner. Although initial drug release from these matrices was driven by differences in matrix architecture and solid-state drug solubility, release toward the later stages was influenced by a combination of fiber swelling and matrix degradation as evidenced by gross and microstructural changes to the mesh network. We suggest that drug release from polymeric matrices can be better understood via investigation of critical matrix characteristics influencing release, as well as concomitant examination of drug-polymer interactions and miscibility. Our findings offer rational matrix design criteria to achieve controlled/extended drug release for promoting sustained biological responses.

9.
J Biomed Mater Res B Appl Biomater ; 107(6): 2019-2029, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30549205

RESUMEN

Synovium-derived mesenchymal stem cells (SMSCs) are an emerging cell source for regenerative medicine applications, including osteochondral defect (OCD) repair. However, in contrast to bone marrow MSCs, scaffold compositions which promote SMSC chondrogenesis/osteogenesis are still being identified. In the present manuscript, we examine poly(ethylene) glycol (PEG)-based scaffolds containing zonally-specific biochemical cues to guide SMSC osteochondral differentiation. Specifically, SMSCs were encapsulated in PEG-based scaffolds incorporating glycosaminoglycans (hyaluronan or chondroitin-6-sulfate [CSC]), low-dose of chondrogenic and osteogenic growth factors (TGFß1 and BMP2, respectively), or osteoinductive poly(dimethylsiloxane) (PDMS). Initial studies suggested that PEG-CSC-TGFß1 scaffolds promoted enhanced SMSC chondrogenic differentiation, as assessed by significant increases in Sox9 and aggrecan. Conversely, PEG-PDMS-BMP2 scaffolds stimulated increased levels of osteoblastic markers with significant mineral deposition. A "Transition" zone formulation was then developed containing a graded mixture of the chondrogenic and osteogenic signals present in the PEG-CSC-TGFß1 and PEG-PDMS-BMP2 constructs. SMSCs within the "Transition" formulation displayed a phenotypic profile similar to hypertrophic chondrocytes, with the highest expression of collagen X, intermediate levels of osteopontin, and mineralization levels equivalent to "bone" formulations. Overall, these results suggest that a graded transition from PEG-CSC-TGFß1 to PEG-PDMS-BMP2 scaffolds elicits a gradual SMSC phenotypic shift from chondrocyte to hypertrophic chondrocyte to osteoblast-like. As such, further development of these scaffold formulations for use in SMSC-based OCD repair is warranted. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater 107B: 2019-2029, 2019.


Asunto(s)
Condrogénesis , Células Madre Mesenquimatosas/metabolismo , Osteogénesis , Membrana Sinovial/metabolismo , Ingeniería de Tejidos , Andamios del Tejido/química , Animales , Antígenos de Diferenciación/biosíntesis , Dimetilpolisiloxanos/química , Perros , Humanos , Masculino
10.
Tissue Eng Part A ; 23(3-4): 101-114, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27736317

RESUMEN

The goal of the present study was to develop a fully three-dimensional (3D) coculture system that would allow for systematic evaluation of the interplay between activated macrophages (AMs) and chondrocytes in osteoarthritic disease progression and treatment. Toward this end, our coculture system was first validated against existing in vitro osteoarthritis models, which have generally cultured healthy normal chondrocytes (NCs)-in two-dimensional (2D) or 3D-with proinflammatory AMs in 2D. In this work, NCs and AMs were both encapsulated within poly(ethylene glycol) diacrylate hydrogels to mimic the native 3D environments of both cell types within the osteoarthritic joint. As with previous studies, increases in matrix metalloproteinases (MMPs) and proinflammatory cytokines associated with the early stages of osteoarthritis were observed during NC-AM coculture, as were decreases in protein-level Aggrecan and collagen II. Thereafter, the coculture system was extended to osteoarthritic chondrocytes (OACs) and AMs to evaluate the potential effects of AMs on pre-existing osteoarthritic phenotypes. OACs in coculture with AMs expressed significantly higher levels of MMP-1, MMP-3, MMP-9, MMP-13, IL-1ß, TNF-α, IL-6, IL-8, and IFN-γ compared to OACs in mono-culture, indicating that proinflammatory macrophages may intensify the abnormal matrix degradation and cytokine secretion already associated with OACs. Likewise, AMs cocultured with OACs expressed significantly more IL-1ß and VEGF-A compared to AM mono-culture controls, suggesting that OACs may intensify abnormal macrophage activation. Finally, OACs cultured in the presence of nonactivated macrophages produced lower levels of MMP-9 and proinflammatory cytokines IL-1ß, TNF-α, and IFN-γ compared to OACs in the OAC-AM system, results that are consistent with anti-inflammatory agents temporarily reducing certain OA symptoms. In summary, the 3D coculture system developed herein captures several key features of inflammatory OA and may prove useful in future screening of therapeutic agents and/or assessment of disease progression mechanisms.


Asunto(s)
Condrocitos/metabolismo , Macrófagos/metabolismo , Modelos Biológicos , Osteoartritis/metabolismo , Células Cultivadas , Condrocitos/patología , Técnicas de Cocultivo , Colagenasas/metabolismo , Citocinas/metabolismo , Humanos , Inflamación/metabolismo , Inflamación/patología , Macrófagos/patología , Osteoartritis/patología
11.
J Biomed Mater Res A ; 105(6): 1712-1724, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28218444

RESUMEN

Bioactive coatings which support the adhesion of late-outgrowth peripheral blood endothelial progenitor cells (EOCs) are actively being investigated as a means to promote rapid endothelialization of "off-the-shelf," small-caliber arterial graft prostheses following implantation. In the present work, we evaluated the behavior of EOCs on thromboresistant graft coatings based on the collagen-mimetic protein Scl2-2 and poly(ethylene glycol) (PEG) diacrylate. Specifically, the attachment, proliferation, migration, and phenotype of EOCs on PEG-Scl2-2 hydrogels were evaluated as a function of Scl2-2 concentration (4, 8, and 12 mg/mL) relative to human umbilical vein endothelial cells (HUVECs). Results demonstrate the ability of each PEG-Scl2-2 hydrogel formulation to support EOC and HUVEC adhesion, proliferation, and spreading. However, only the 8 and 12 mg/mL PEG-Scl2-2 hydrogels were able to support stable EOC and HUVEC confluence. These PEG-Scl2-2 formulations were, therefore, selected for evaluation of their impact on EOC and HUVEC phenotype relative to PEG-collagen hydrogels. Cumulatively, both gene and protein level data indicated that 8 mg/mL PEG-Scl2-2 hydrogels supported similar or improved levels of EOC maturation relative to PEG-collagen controls based on evaluation of CD34, VEGFR2, PECAM-1, and VE-Cadherin. The 8 mg/mL PEG-Scl2-2 hydrogels also appeared to support similar or improved levels of EOC homeostatic marker expression relative to PEG-collagen hydrogels based on von Willebrand factor, collagen IV, NOS3, thrombomodulin, and E-selectin assessment. Combined, the present results indicate that PEG-Scl2-2 hydrogels warrant further investigation as "off-the-shelf" graft coatings. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 1712-1724, 2017.


Asunto(s)
Materiales Biocompatibles/química , Colágeno/química , Células Endoteliales/citología , Células Progenitoras Endoteliales/citología , Hidrogeles/química , Polietilenglicoles/química , Venas Umbilicales/citología , Adhesión Celular , Movimiento Celular , Proliferación Celular , Células Endoteliales de la Vena Umbilical Humana , Humanos , Integrinas/análisis , Ensayo de Materiales
12.
J Mater Chem B ; 3(40): 7912-7919, 2015 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-28989705

RESUMEN

This work evaluates the response of human aortic endothelial cells (HAECs) to thromboresistant collagen-mimetic hydrogel coatings toward improving the biocompatibility of existing "off-the-shelf" small-caliber vascular grafts. Specifically, bioactive hydrogels - previously shown to support α1/α2 integrin-mediated cell adhesion but to resist platelet activation - were fabricated by combining poly(ethylene glycol) (PEG) with a 120 kDa, triple-helical collagen-mimetic protein(Scl2-2) containing the GFPGER adhesion sequence. Analysis of HAECs seeded onto the resulting PEG-Scl2-2 hydrogels demonstrated that HAEC adhesion increased with increasing Scl2-2 concentration, while HAEC migration rate decreased over this same concentration range. In addition, evaluation of HAEC phenotype at confluence indicated significant differences in the gene expression of NOS3, thrombomodulin, and E-selectin on the PEG-Scl2-2 hydrogels relative to PEG-collagen controls. At the protein level, however, only NOS3 was significantly different between the PEG-Scl2-2 and PEG-collagen surfaces. Furthermore, PECAM-1 and VE-cadherin expression on PEG-Scl2-2 hydrogels versus PEG-collagen controls could not be distinguished at either the gene or protein level. Cumulatively, these data indicate the PEG-Scl2-2 hydrogels warrant further investigation as "off-the-shelf" graft coatings. In future studies, the Scl2-2 protein can potentially be modified to include additional extracellular matrix or cytokine binding sites to further improve endothelial cell responses.

13.
ACS Biomater Sci Eng ; 1(12): 1220-1230, 2015 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-33304994

RESUMEN

Recently, a novel shape memory polymer foam based on the photopolymerization of poly(ε-caprolactone) diacrylate (PCLDA) has been developed. These PCLDA foams enter a temporary softened state when briefly treated with warm saline (T saline > T m of PCLDA), allowing them to conform to irregular bone defect "boundaries" prior to shape setting. When coated with a mechanically stable polydopamine (PD) layer, these PCLDA foams have previously been demonstrated to induce hydroxyapatite deposition. In the present study, the osteoinductivity of these "self-fitting" PD-coated PCLDA (PD-PCLDA) materials was evaluated relative to uncoated PCLDA (U-PCLDA) controls using bone marrow-derived human mesenchymal stem cells (h-MSCs). When cultured in the absence of osteogenic media supplements, PD-PCLDA scaffolds expressed similar levels of Runx2, alkaline phosphatase, and osteopontin protein as U-PCLDA scaffolds cultured in the presence of osteogenic media supplements. In addition, PD-PCLDA scaffolds cultured without osteogenic supplements did not significantly promote undesired lineage progression (e.g., adipogenesis or chondrogenesis) of h-MSCs. Cumulatively, these data indicate that PD-PCLDA materials display increased osteoinductivity relative to U-PCLDA substrates. Future studies will examine tethered osteogenic factors or peptides toward augmenting the osteoinductive properties of the PD-PCLDA foams.

14.
Acta Biomater ; 9(9): 8037-45, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23791671

RESUMEN

Calcium phosphate ceramics (CPCs) have been widely used as biomaterials for the regeneration of bone tissue because of their ability to induce osteoblastic differentiation in progenitor cells. Despite the progress made towards fabricating CPCs possessing a range of surface features and chemistries, the influence of material properties in orchestrating cellular events such as adhesion and differentiation is still poorly understood. Specifically, questions such as why certain CPCs may be more osteoinductive than others, and how material properties contribute to osteoinductivity/osteoconductivity remain unanswered. Therefore, this review article systematically discusses the effects of the physical (e.g. surface roughness) and chemical properties (e.g. solubility) of CPCs on protein adsorption, cell adhesion and osteoblastic differentiation in vitro. The review also provides a summary of possible signaling pathways involved in osteoblastic differentiation in the presence of CPCs. In summary, these insights on the contribution of material properties towards osteoinductivity and the role of signaling molecules involved in osteoblastic differentiation can potentially aid the design of CPC-based biomaterials that support bone regeneration without the need for additional biochemical supplements.


Asunto(s)
Sustitutos de Huesos/química , Fosfatos de Calcio/química , Cerámica/química , Osteoblastos/citología , Osteoblastos/fisiología , Osteogénesis/fisiología , Ingeniería de Tejidos/instrumentación , Animales , Adhesión Celular/fisiología , Diferenciación Celular/fisiología , Proliferación Celular , Supervivencia Celular/fisiología , Diseño de Equipo , Humanos , Propiedades de Superficie
15.
Biomaterials ; 33(31): 7727-35, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22835644

RESUMEN

Biomaterial scaffolds with gradients in architecture, mechanical and chemical properties have the potential to improve the osseointegration of ligament grafts by recapitulating phenotypic gradients that exist at the natural ligament-bone (L-B) interface. Towards the larger goal of regenerating the L-B interface, this in vitro study was performed to investigate the potential of two scaffolds with mineral gradients in promoting a spatial gradient of osteoblastic differentiation. Specifically, the first graded scaffold was fabricated by co-electrospinning two polymer solutions (one doped with nano-hydroxyapatite particles) from offset spinnerets, while the second was created by immersing the first scaffold in a 5 × simulated body fluid. Rat bone marrow stromal cells, cultured in the presence of osteogenic supplements, were found to be metabolically active on all regions of both scaffolds after 1 and 7 days of culture. Gene expression of bone morphogenic protein-2 and osteopontin was elevated on mineral-containing regions as compared to regions without mineral, while the expression of alkaline phosphatase mRNA revealed the opposite trend. Finally, the presence of osteopontin and bone sialoprotein confirmed osteoblastic phenotypic maturation by day 28. This study indicates that co-electrospun scaffolds with gradients in mineral content can guide the formation of phenotypic gradients and may thus promote the regeneration of the L-B interface.


Asunto(s)
Huesos/fisiología , Ligamentos/fisiología , Células Madre Mesenquimatosas/citología , Ingeniería de Tejidos/métodos , Andamios del Tejido/química , Animales , Biomarcadores/metabolismo , Huesos/efectos de los fármacos , Recuento de Células , Diferenciación Celular/efectos de los fármacos , Citoesqueleto/efectos de los fármacos , Citoesqueleto/metabolismo , Durapatita/farmacología , Proteínas de la Matriz Extracelular/metabolismo , Técnica del Anticuerpo Fluorescente , Regulación de la Expresión Génica/efectos de los fármacos , Ligamentos/efectos de los fármacos , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Nanopartículas/ultraestructura , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley
16.
Acta Biomater ; 7(12): 4131-8, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21791254

RESUMEN

Current scaffolds for the regeneration of anterior cruciate ligament injuries are unable to capture intricate mechanical and chemical gradients present in the natural ligament-bone interface. As a result, stress concentrations can develop at the scaffold-bone interface, leading to poor osseointegration. Hence, scaffolds that possess appropriate mechano-chemical gradients would help establish normal loading properties at the interface, while promoting scaffold integration with bone. With the long-term goal of investigating regeneration of the ligament-bone interface, this feasibility study aimed to fabricate a continuously graded mesh. Specifically, graded meshes were fabricated by co-electrospinning nanohydroxyapatite/polycaprolactone (nHAP-PCL) and poly(ester urethane) urea elastomer solutions from offset spinnerets. Next, mineral crystallites were selectively deposited on the nHAP-PCL fibers by treatment with a 5× simulated body fluid (5× SBF). X-ray diffraction and energy-dispersive spectroscopy indicated calcium-deficient hydroxyapatite-like mineral crystallites with an average Ca/P ratio of 1.48. Tensile testing demonstrated the presence of a mechanical gradient, which became more pronounced upon treatment with 5× SBF. Finally, biocompatibility of the graded meshes was verified using an MC3T3-E1 osteoprogenitor cell line. The study demonstrates that graded meshes, for potential application in interfacial tissue engineering, can be fabricated by co-electrospinning.


Asunto(s)
Huesos , Ligamentos , Modelos Teóricos , Regeneración , Microscopía Electrónica de Rastreo , Difracción de Rayos X
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA