Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Stephanie J. Bryant is active.

Publication


Featured researches published by Stephanie J. Bryant.


Tissue Engineering Part B-reviews | 2008

Cell Encapsulation in Biodegradable Hydrogels for Tissue Engineering Applications

Garret D. Nicodemus; Stephanie J. Bryant

Encapsulating cells in biodegradable hydrogels offers numerous attractive features for tissue engineering, including ease of handling, a highly hydrated tissue-like environment for cell and tissue growth, and the ability to form in vivo. Many properties important to the design of a hydrogel scaffold, such as swelling, mechanical properties, degradation, and diffusion, are closely linked to the crosslinked structure of the hydrogel, which is controlled through a variety of different processing conditions. Degradation may be tuned by incorporating hydrolytically or enzymatically labile segments into the hydrogel or by using natural biopolymers that are susceptible to enzymatic degradation. Because cells are present during the gelation process, the number of suitable chemistries and formulations are limited. In this review, we describe important considerations for designing biodegradable hydrogels for cell encapsulation and highlight recent advances in material design and their applications in tissue engineering.


Journal of Biomaterials Science-polymer Edition | 2000

Cytocompatibility of UV and visible light photoinitiating systems on cultured NIH/3T3 fibroblasts in vitro.

Stephanie J. Bryant; Charles R. Nuttelman; Kristi S. Anseth

This work investigates the cytocompatibility of several photoinitiating systems for potential cell encapsulation applications. Both UV and visible light initiating schemes were examined. The UV photoinitiators included 2,2-dimethoxy-2-phenylacetophenone (Irgacure 651), 1-hydroxycyclohexyl phenyl ketone (Irgacure 184), 2-methyl-1-[4-(methylthio) phenyl]-2-(4-morpholinyl)-1-propanone (Irgacure 907), and 2-hydroxy-1-[4-(hydroxyethoxy)phenyl]-2-methyl-1-propanone (Darocur 2959). The visible light initiating systems included camphorquinone (CQ) with ethyl 4-N, N-dimethylaminobenzoate (4EDMAB) and triethanolamine (TEA) and the photosensitizer isopropyl thioxanthone. A cultured fibroblast cell line, NIH/3T3, was exposed to the photoinitiators at varying concentrations from 0.01% (w/w) to 0.1% (w/w) with and without the presence of initiating light. The results demonstrated that at low photoinitiator concentrations (6 0.01% (w/w)), all of the initiator molecules were cytocompatible with the exception of CQ, Irgacure 651, and 4EDMAB which had a relative survival ~ 50% lower than a control. In the presence of low intensity initiating light (~ 6 mW cm-2 of 365 nm UV light and ~ 60 mW cm-2 of 470-490 nm visible light) and initiating radicals, Darocur 2959 at concentrations 6 0.05% (w/w) and CQ at concentrations 6 0.01% (w/w) were the most promising cytocompatible UV and visible light initiating systems, respectively. To demonstrate the potential use of cytocompatible photoinitiating systems in cell encapsulation applications, chondrocytes were encapsulated in a photocrosslinked hydrogel using 0.05% (w/w) Darocur 2959 (cytocompatible) and 0.01% (w/w) Irgacure 651 (cyto-incompatible). After photopolymerizing for 10 minutes with ~ 8 mW cm-2 of 365 nm light, nearly all the chondrocytes survived the process with Darocur 2959 while very few of the chondrocytes survived the process with Irgacure 651.


Journal of Controlled Release | 2002

In situ forming degradable networks and their application in tissue engineering and drug delivery

Kristi S. Anseth; Andrew T. Metters; Stephanie J. Bryant; Penny J. Martens; Jennifer H. Elisseeff; Christopher N. Bowman

Multifunctional macromers based on poly(ethylene glycol) and poly(vinyl alcohol) were photopolymerized to form degradable hydrogel networks. The degradation behavior of the highly swollen gels was characterized by monitoring changes in their mass loss, degree of swelling, and compressive modulus. Experimental results show that the modulus decreases exponentially with time, while the volumetric swelling ratio increases exponentially. A degradation mechanism assuming pseudo first-order hydrolysis kinetics and accounting for the structure of the crosslinked networks successfully predicted the experimentally observed trends in these properties with degradation. Once verified, the proposed degradation mechanism was extended to correlate network degradation kinetics, and subsequent changes in network structure, with release behavior of bioactive molecules from these dynamic systems. A theoretical model utilizing a statistical approach to predict the cleavage of crosslinks within the network was used to predict the complex erosion profiles produced by these hydrogels. Finally, the application of these macromers as in situ forming hydrogel constructs for cartilage tissue engineering is demonstrated.


Biomaterials | 2001

The effects of scaffold thickness on tissue engineered cartilage in photocrosslinked poly(ethylene oxide) hydrogels

Stephanie J. Bryant; Kristi S. Anseth

The thickness of human articular cartilage has been reported to vary from less than 0.5 up to 7 mm. Hence, tissue engineered cartilage scaffolds should be able to span the thickness of native cartilage to fill defects of all shapes and sizes. In this study, we demonstrate the potential for using photopolymerization technology to encapsulate chondrocytes in poly(ethylene oxide) hydrogels, which vary in thickness from 2 to 8 mm. Chondrocytes, encapsulated in an 8 mm thick, photocrosslinked hydrogel and cultured in vitro for 6 weeks, remained viable and produced cartilaginous tissue throughout the construct comparable to a 2 mm thick gel as seen both histologically and biochemically. In addition, the total collagen and glycosaminoglycan contents per wet weight of the 8 mm thick cell-polymer construct were 0.13 +/- 0.01 and 0.25 +/- 0.03%, respectively, and did not vary significantly as a function of spatial position in the construct. The histological evidence and the biochemical content were similar in all constructs of varying thickness. The results suggest that photocrosslinked hydrogels are promising scaffolds for tissue engineering cartilage as cell viability is readily maintained; uniform cell seeding is easy to achieve: and the biochemical content of the extracellular matrix is not compromised as the scaffold thickness is increased from 2 to 8 mm.


Annals of Biomedical Engineering | 2004

Crosslinking density influences chondrocyte metabolism in dynamically loaded photocrosslinked poly(ethylene glycol) hydrogels.

Stephanie J. Bryant; T.T. Chowdhury; David A. Lee; Dan L. Bader; Kristi S. Anseth

In approaches to tissue engineer articular cartilage, an important consideration for in situ forming cell carriers is the impact of mechanical loading on the cell composite structure and function. Photopolymerized hydrogel scaffolds based on poly(ethylene glycol) (PEG) may be synthesized with a range of crosslinking densities and corresponding macroscopic properties. This study tests the hypothesis that changes in the hydrogel crosslinking density influences the metabolic response of encapsulated chondrocytes to an applied load. PEG hydrogels were formulated with two crosslinking densities that resulted in gel compressive moduli ranging from 60 to 670 kPa. When chondrocytes were encapsulated in these PEG gels, an increase in crosslinking density resulted in an inhibition in cell proliferation and proteoglycan synthesis. Moreover, when the gels were dynamically loaded for 48 h in unconfined compression with compressive strains oscillating from 0 to 15% at a frequency of 1 Hz, cell proliferation and proteoglycan synthesis were affected in a crosslinking-density-dependent manner. Cell proliferation was inhibited in both crosslinked gels, but was greater in the highly crosslinked gel. In contrast, dynamic loading did not influence proteoglycan synthesis in the loosely crosslinked gel, but a marked decrease in proteoglycan production was observed in the highly crosslinked gel. In summary, changes in PEG hydrogel properties greatly affect how chondrocytes respond to an applied dynamic load.


Journal of Biomedical Materials Research Part A | 2012

The effects of substrate stiffness on the in vitro activation of macrophages and in vivo host response to poly(ethylene glycol)-based hydrogels.

Anna K. Blakney; Mark D. Swartzlander; Stephanie J. Bryant

Poly(ethylene glycol) (PEG) hydrogels, modified with RGD, are promising platforms for cell encapsulation and tissue engineering. While these hydrogels offer tunable mechanical properties, the extent of the host response may limit their in vivo applicability. The overall objective was to characterize the effects of hydrogel stiffness on the in vitro macrophage response and in vivo host response. We hypothesized that stiffer substrates induce better attachment, adhesion, and increased cell spreading, which elevates the macrophage classically activated phenotype and leads to a more severe foreign body reaction (FBR). PEG-RGD hydrogels were fabricated with compressive moduli of 130, 240, and 840 kPa, and the same RGD concentration. Hydrogel stiffness did not impact macrophage attachment, but elicited differences in cell morphology. Cells retained a round morphology on 130 kPa substrates, with localized and dense F-actin and localized α(V) integrin stainings. Contrarily, cells on stiffer substrates were more spread, with filopodia protruding from the cell, a more defined F-actin, and greater α(V) integrin staining. When stimulated with lipopolysaccharide, macrophages had a classical activation phenotype, with increased expression of TNF-α, IL-1β, and IL-6, however the degree of activation was significantly reduced with the softest hydrogels. A FBR ensued in response to all hydrogels when implanted subcutaneously in mice, but 28 days postimplantation the layer of macrophages at the implant surface was significantly lower in the softest hydrogels. In conclusion, hydrogels with lower stiffness led to reduced macrophage activation and a less severe and more typical FBR, and therefore are more suited for in vivo tissue engineering applications.


Biomacromolecules | 2008

Degradable poly(2-hydroxyethyl methacrylate)-co-polycaprolactone hydrogels for tissue engineering scaffolds.

Sarah Atzet; Scott A. Curtin; Phalen Trinh; Stephanie J. Bryant; Buddy D. Ratner

Biodegradable poly(2-hydroxyethyl methacrylate)(pHEMA) hydrogels for engineered tissue constructs were developed by the use of atom transfer radical polymerization (ATRP), a degradable cross-linker, and a macroinitiator. Hydrogels are appropriate materials for tissue engineering scaffolds because of their tissue-like mechanical compliance and mass transfer properties. However, many hydrogels that have seen wide application in medicine are not biodegradable or cannot be easily cleared from the body. pHEMA was selected for the scaffold material because of its reasonable mechanical strength, elasticity, and long history of successful use in medicine as well as because it can be easily fabricated into numerous configurations. pHEMA was studied at various molecular weights between 2 and 50 kDa. The molecular weight range suitable for renal clearance was an important factor in the experimental design. The fabricated hydrogels contain oligomeric blocks of polycaprolactone (PCL), a hydrolytically and enzymatically degradable polymer, as a cross-linking agent. In addition, a degradable macroinitiator that also contained oligomeric PCL was used to initiate the ATRP. The chain length, cross-link density, and polymerization solvent were found to affect the mechanical properties of the pHEMA hydrogels. Degradation of the pHEMA hydrogels was characterized by the use of 0.007 M NaOH, lipase solutions, and phosphate-buffered saline. The mass loss, swelling ratio, and tensile modulus were evaluated. Degradation products after sodium hydroxide treatment were measured by the use of gel permeation chromatography (GPC) to verify the polymer lengths and polydispersity. Erosion was observed in only the sodium hydroxide and lipase solutions. However, the swelling ratio and tensile modulus indicate bulk degradation in all PCL-containing samples. Degradable hydrogels in enzymatic solutions showed 30% mass loss in 16 weeks. Initial cell toxicity studies indicate no adverse cellular response to the hydrogels or their degradation products. These hydrogels have appropriate mechanical properties and a tunable degradation rate, and they are composed of materials that are currently in FDA-approved devices. Therefore, the degradable pHEMA developed in this study has considerable potential as a scaffold for tissue engineering applications, in cardiac and other applications.


Journal of Biomedical Materials Research Part A | 2012

Student award winner in the undergraduate category for the society of biomaterials 9th World Biomaterials Congress, Chengdu, China, June 1-5, 2012: The effects of substrate stiffness on the in vitro activation of macrophages and in vivo host response to poly(ethylene glycol)-based hydrogels

Anna K. Blakney; Mark D. Swartzlander; Stephanie J. Bryant

Poly(ethylene glycol) (PEG) hydrogels, modified with RGD, are promising platforms for cell encapsulation and tissue engineering. While these hydrogels offer tunable mechanical properties, the extent of the host response may limit their in vivo applicability. The overall objective was to characterize the effects of hydrogel stiffness on the in vitro macrophage response and in vivo host response. We hypothesized that stiffer substrates induce better attachment, adhesion, and increased cell spreading, which elevates the macrophage classically activated phenotype and leads to a more severe foreign body reaction (FBR). PEG-RGD hydrogels were fabricated with compressive moduli of 130, 240, and 840 kPa, and the same RGD concentration. Hydrogel stiffness did not impact macrophage attachment, but elicited differences in cell morphology. Cells retained a round morphology on 130 kPa substrates, with localized and dense F-actin and localized α(V) integrin stainings. Contrarily, cells on stiffer substrates were more spread, with filopodia protruding from the cell, a more defined F-actin, and greater α(V) integrin staining. When stimulated with lipopolysaccharide, macrophages had a classical activation phenotype, with increased expression of TNF-α, IL-1β, and IL-6, however the degree of activation was significantly reduced with the softest hydrogels. A FBR ensued in response to all hydrogels when implanted subcutaneously in mice, but 28 days postimplantation the layer of macrophages at the implant surface was significantly lower in the softest hydrogels. In conclusion, hydrogels with lower stiffness led to reduced macrophage activation and a less severe and more typical FBR, and therefore are more suited for in vivo tissue engineering applications.


Acta Biomaterialia | 2012

Compressive Elasticity of Three-Dimensional Nanofiber Matrix Directs Mesenchymal Stem Cell Differentiation to Vascular Cells with Endothelial or Smooth Muscle Cell Markers

Kathryn Wingate; Walter Bonani; Yan Tan; Stephanie J. Bryant; Wei Tan

The importance of mesenchymal stem cells (MSC) in vascular regeneration is becoming increasingly recognized. However, few in vitro studies have been performed to identify the effects of environmental elasticity on the differentiation of MSC into vascular cell types. Electrospinning and photopolymerization techniques were used to fabricate a three-dimensional (3-D) polyethylene glycol dimethacrylate nanofiber hydrogel matrix with tunable elasticity for use as a cellular substrate. Compression testing demonstrated that the elastic modulus of the hydrated 3-D matrices ranged from 2 to 15 kPa, similar to the in vivo elasticity of the intima basement membrane and media layer. MSC seeded on rigid matrices (8-15 kPa) showed an increase in cell area compared with those seeded on soft matrices (2-5 kPa). Furthermore, the matrix elasticity guided the cells to express different vascular-specific phenotypes with high differentiation efficiency. Around 95% of MSC seeded on the 3-D matrices with an elasticity of 3 kPa showed Flk-1 endothelial markers within 24h, while only 20% of MSC seeded on the matrices with elasticity >8 kPa demonstrated Flk-1 marker. In contrast, ∼80% of MSC seeded on 3-D matrices with elasticity >8 kPa demonstrated smooth muscle α-actin marker within 24h, while fewer than 10% of MSC seeded on 3-D matrices with elasticity <5 kPa showed α-actin markers. The ability to control MSC differentiation into either endothelial or smooth muscle-like cells based purely on the local elasticity of the substrate could be a powerful tool for vascular tissue regeneration.


Journal of Biomechanics | 2008

The role of hydrogel structure and dynamic loading on chondrocyte gene expression and matrix formation

Garret D. Nicodemus; Stephanie J. Bryant

Crosslinked poly(ethylene glycol) (PEG) hydrogels are attractive scaffolds for cartilage tissue engineering because of their ability to mimic the aqueous environment and mechanical properties of native cartilage. In this study, hydrogel crosslinking density was varied to study the influence of gel structure and the application of dynamic loading (continuous, 1 Hz, 15% amplitude strain) on chondrocyte gene expression over approximately 1 week culture. Gene expression was quantified using real-time RT-PCR for collagen II and aggrecan, the major cartilage extracellular matrix (ECM) components, and collagen I, an indicator of chondrocyte de-differentiation. When chondrocytes were encapsulated in PEG gels with low or high crosslinking, a high collagen II expression compared to collagen I expression (1000 or 100,000:1, respectively) indicated the native chondrocyte phenotype was retained. In the absence of loading, relative gene expression for collagen II and aggrecan was significantly higher (e.g., 2-fold and 4-fold, respectively, day 7) in the low crosslinked gels compared to gels with higher crosslinking. Dynamic loading, however, showed little effect on ECM gene expression in both crosslinked systems. To better understand the cellular environment, ECM production was qualitatively assessed using an in situ immunofluorescent technique and standard histology. A pericellular matrix (PCM) was observed as early as day 3 post-encapsulation and the degree of formation was dependent on gel crosslinking. These results suggest the PCM may protect the cells from sensing the applied loads. This study demonstrates that gel structure has a profound effect on chondrocyte gene expression, while dynamic loading has much less of an effect at early culture times.

Collaboration


Dive into the Stephanie J. Bryant's collaboration.

Top Co-Authors

Avatar

Kristi S. Anseth

University of Colorado Boulder

View shared research outputs
Top Co-Authors

Avatar

Justine J. Roberts

University of Colorado Boulder

View shared research outputs
Top Co-Authors

Avatar

Garret D. Nicodemus

University of Colorado Boulder

View shared research outputs
Top Co-Authors

Avatar

Idalis Villanueva

University of Colorado Boulder

View shared research outputs
Top Co-Authors

Avatar

Stacey C. Skaalure

University of Colorado Boulder

View shared research outputs
Top Co-Authors

Avatar

Virginia L. Ferguson

University of Colorado Boulder

View shared research outputs
Top Co-Authors

Avatar

Elizabeth A. Aisenbrey

University of Colorado Boulder

View shared research outputs
Top Co-Authors

Avatar

Franck J. Vernerey

University of Colorado Boulder

View shared research outputs
Top Co-Authors

Avatar

Aaron D. Lynn

University of Colorado Boulder

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge