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










Base de dados
Intervalo de ano de publicação
1.
Bioengineering (Basel) ; 10(4)2023 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-37106654

RESUMO

Articular cartilage (AC) tissue repair and regeneration remains an ongoing challenge. One component of the challenge is the limited ability to scale an engineered cartilage graft to clinically relevant sizes while maintaining uniform properties. In this paper, we report on the evaluation of our polyelectrolyte complex microcapsule (PECM) platform technology as a technique for generating cartilage-like spherical modules. Bone marrow-derived mesenchymal stem cells (bMSCs) or primary articular chondrocytes were encapsulated within PECMs composed of methacrylated hyaluronan, collagen I, and chitosan. The formation of cartilage-like tissue in the PECMs over a 90-day culture was characterized. The results showed that chondrocytes exhibited superior growth and matrix deposition compared to either chondrogenically-induced bMSCs or a mixed PECM culture containing both chondrocytes and bMSCs. The chondrocyte-generated matrix filled the PECM and produced substantial increases in capsule compressive strength. The PECM system thus appears to support intracapsular cartilage tissue formation and the capsule approach promotes efficient culture and handling of these micro tissues. Since previous studies have proven the feasibility of fusing such capsules into large tissue constructs, the results suggest that encapsulating primary chondrocytes in PECM modules may be a viable route toward achieving a functional articular cartilage graft.

2.
Ann Biomed Eng ; 47(5): 1223-1236, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30796550

RESUMO

The bioinspired, microscale tissue engineering approach has emerged in recent years as a promising alternative to preformed scaffolds. Using this approach, complex tissues and organs can be efficiently engineered from microscale modules to replicate the intricate architecture and physiology of vascularized organs and tissues. Previously, we demonstrated assembly of a prototype, engineered liver tissue, formed by the fusion of hepatocyte-containing capsules. Here, we analyzed the effects of various controllable system parameters with the aim of predicting the operating limits of our modular tissue in high cell density, perfused cultures. Both the capsule diameter and construct height were limited by mass transfer requirements, while the shear stress on the capsule wall and the pressure drop across the packed capsule bed were dictated by the capsule diameter and permissible flow rates of the system. Our analysis predicts that capsules with a 200 µm radius can efficiently maintain hepatocytes at cell densities comparable to liver tissue. Some model predictions were validated by packed bed perfusion cultures. Flow-induced bed compaction hysteresis was tested experimentally and found to have minimal effect on flow characteristics. The effectiveness factor (η) for the overall oxygen transfer within packed beds of capsule modules was estimated to be 0.72 for all conditions. Primary hepatocytes encapsulated in the capsules exhibited normal metabolism and formed spheroids during a 7-day culture. The model predictions can be useful to study mass transfer and shear stress in high-density perfusion cultures. Overall, analysis of a perfused, capsule-based, modular tissue demonstrated the feasibility of the technology as a platform for fabrication of highly metabolic solid organs.


Assuntos
Órgãos Bioartificiais , Hepatócitos/metabolismo , Fígado Artificial , Fígado/metabolismo , Modelos Biológicos , Engenharia Tecidual , Animais , Transporte Biológico Ativo , Reatores Biológicos , Técnicas de Cultura de Células , Ratos
3.
Discoveries (Craiova) ; 7(1): e90, 2019 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-32309608

RESUMO

Skeletal muscle tissue has inherent capacity for regeneration in response to minor injuries. However, in the case of severe trauma, tumor ablations, or in congenital muscle defects, these myopathies can cause irreversible loss of muscle mass and function, a condition referred to as volumetric muscle loss (VML). The natural muscle repair mechanisms are overwhelmed, prompting the search for new muscle regenerative strategies, such as using biomaterials that can provide regenerative signals to either transplanted or host muscle cells. Recent studies involve the use of suitable biomaterials which may be utilized as a template to guide tissue reorganization and ultimately provide optimum micro-environmental conditions to cells. These strategies range from approaches that utilize biomaterials alone to those that combine materials with exogenous growth factors, and ex vivo cultured cells. A number of scaffold materials have been used in the development of grafts to treat VML. In this brief review, we outline the natural skeletal regeneration process, available treatments used in the clinic for muscle injury and promising tissue bioengineering and regenerative approaches for muscle loss treatment.

4.
Bioeng Transl Med ; 3(3): 265-274, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30377665

RESUMO

Polyelectrolyte microcapsules are modular constructs which facilitate cell handling and assembly of cell-based tissue constructs. In this study, an electrospray (ES) encapsulation apparatus was developed for the encapsulation of mesenchymal stem cells (MSCs). Ionic complexation between glycosaminoglycans (GAGs) and chitosan formed a polyelectrolyte complex membrane at the interface. To optimize the capsules, the effect of voltage, needle size and GAG formulation on capsule size were investigated. It was observed that by increasing the voltage and decreasing the needle size, the capsule size would decrease but at voltages above 12 kV, capsule size distribution broadened significantly which yields lower circularity. Increase in GAG viscosity resulted in larger microcapsules and cell viability exhibited no significant changes during the encapsulation procedure. These results suggest that ES is a highly efficient, and scalable approach to the encapsulation of MSCs for subsequent use in bioprinting and other modular tissue engineering or regenerative medicine applications.

5.
J Biomed Mater Res A ; 105(6): 1725-1735, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28198136

RESUMO

Heparin has shown promise as a component of various biomaterial formulations, but its variable properties and inhibitory effects on some cell types have raised interest in use of dextran sulfate as an alternative. In this study, we characterized the interactions of vascular smooth muscle (SMC) and endothelial cells (EC) with heparin and dextran sulfate immobilized onto chitosan-based films. Films were modified by blending chitosan with type I collagen and covalently attaching heparin or dextran sulfate at various levels. Cell-material interactions were evaluated by quantifying cell spreading, shape and proliferation rate. ECs proliferated well on chitosan, but the polymer was a mediocre substrate for SMC growth. Immobilizing heparin on chitosan further inhibited SMC proliferation. However, blending collagen reversed the heparin inhibition of SMC growth, resulting in a pro-proliferative effect of heparin immobilized on chitosan-collagen films. Dextran sulfate surfaces supported both SMC and EC proliferation with or without the presence of collagen. The results indicate that inhibitory effects of heparin on SMC are reversed by immobilization in the presence of collagen, and that dextran sulfate may be superior to heparin as a biomaterial additive for promoting vascular cell growth in chitosan-based scaffolds. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 1725-1735, 2017.


Assuntos
Anticoagulantes/farmacologia , Materiais Biocompatíveis/química , Proliferação de Células/efeitos dos fármacos , Sulfato de Dextrana/farmacologia , Células Endoteliais/efeitos dos fármacos , Heparina/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Animais , Anticoagulantes/administração & dosagem , Separação Celular , Células Cultivadas , Quitosana/química , Colágeno Tipo I/química , Sulfato de Dextrana/administração & dosagem , Sistemas de Liberação de Medicamentos/métodos , Liberação Controlada de Fármacos , Células Endoteliais/citologia , Heparina/administração & dosagem , Músculo Liso Vascular/citologia , Ratos , Suínos
6.
Ann Biomed Eng ; 43(10): 2467-76, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25777293

RESUMO

The onset of post-traumatic osteoarthritis (PTOA) remains prevalent following traumatic joint injury such as anterior cruciate ligament (ACL) rupture, and animal models are important for studying the pathomechanisms of PTOA. Noninvasive ACL injury using the tibial compression model in the rat has not been characterized, and it may represent a more clinically relevant model than the common surgical ACL transection model. This study employed four loading profiles to induce ACL injury, in which motion capture analysis was performed, followed by quantitative joint laxity testing. High-speed, high-displacement loading repeatedly induces complete ACL injury, which causes significant increases in anterior-posterior and varus laxity. No loading protocol induced valgus laxity. Tibial internal rotation and anterior subluxation occurs up to the point of ACL failure, after which the tibia rotates externally as it subluxes over the femoral condyles. High displacement was more determinative of ACL injury compared to high speed. Low-speed protocols induced ACL avulsion from the femoral footprint whereas high-speed protocols caused either midsubstance rupture, avulsion, or a combination injury of avulsion and midsubstance rupture. This repeatable, noninvasive ACL injury protocol can be utilized in studies assessing PTOA or ACL reconstruction in the rat.


Assuntos
Lesões do Ligamento Cruzado Anterior , Ligamento Cruzado Anterior/patologia , Ligamento Cruzado Anterior/fisiopatologia , Tíbia/fisiopatologia , Animais , Modelos Animais de Doenças , Feminino , Osteoartrite/patologia , Osteoartrite/fisiopatologia , Ratos , Ratos Endogâmicos Lew , Tíbia/patologia
7.
PLoS One ; 9(1): e84287, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24465401

RESUMO

The limited ability to vascularize and perfuse thick, cell-laden tissue constructs has hindered efforts to engineer complex tissues and organs, including liver, heart and kidney. The emerging field of modular tissue engineering aims to address this limitation by fabricating constructs from the bottom up, with the objective of recreating native tissue architecture and promoting extensive vascularization. In this paper, we report the elements of a simple yet efficient method for fabricating vascularized tissue constructs by fusing biodegradable microcapsules with tunable interior environments. Parenchymal cells of various types, (i.e. trophoblasts, vascular smooth muscle cells, hepatocytes) were suspended in glycosaminoglycan (GAG) solutions (4%/1.5% chondroitin sulfate/carboxymethyl cellulose, or 1.5 wt% hyaluronan) and encapsulated by forming chitosan-GAG polyelectrolyte complex membranes around droplets of the cell suspension. The interior capsule environment could be further tuned by blending collagen with or suspending microcarriers in the GAG solution These capsule modules were seeded externally with vascular endothelial cells (VEC), and subsequently fused into tissue constructs possessing VEC-lined, inter-capsule channels. The microcapsules supported high density growth achieving clinically significant cell densities. Fusion of the endothelialized, capsules generated three dimensional constructs with an embedded network of interconnected channels that enabled long-term perfusion culture of the construct. A prototype, engineered liver tissue, formed by fusion of hepatocyte-containing capsules exhibited urea synthesis rates and albumin synthesis rates comparable to standard collagen sandwich hepatocyte cultures. The capsule based, modular approach described here has the potential to allow rapid assembly of tissue constructs with clinically significant cell densities, uniform cell distribution, and endothelialized, perfusable channels.


Assuntos
Glicosaminoglicanos/química , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Animais , Linhagem Celular , Células Cultivadas , Células Endoteliais/citologia , Humanos , Ratos
8.
J Mech Behav Biomed Mater ; 20: 217-26, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23465267

RESUMO

The low mechanical properties of hydrogel materials such as chitosan hinder their broad utility for tissue engineering applications. Previous research efforts improved the mechanical properties of chitosan fiber through chemical and physical modifications; however, unfavorable toxicity effects on cells were reported. In this paper, we report the preparation of chitosan fibers with improved mechanical and biocompatibility properties. The structure-property relationships of extruded chitosan fibers were explored by varying acetic acid (AA) concentration, ammonia concentration, annealing temperature and degree of heparin crosslinking. Results showed that optimizing AA concentration to 2vol% improved fiber strength and stiffness by 2-fold. Extruding chitosan solution into 25wt% of ammonia solution reduced fiber diameters and improved fiber strength by 2-fold and stiffness by 3-fold, due to an increase in crystallinity as confirmed by XRD. Fiber annealing further reduced fiber diameter and improved fiber strength and stiffness as temperature increased. Chitosan fibers crosslinked with heparin had increased diameter but lower strength and stiffness properties and higher breaking strain values. When individual parameters were combined, further improvement in fiber mechanical properties was achieved. All mechanically improved fibers and heparin crosslinked fibers promoted valvular interstitial cells (VIC) attachment and growth over 10 day cultures. Our results demonstrate the ability to substantially improve the mechanical properties of chitosan fibers without adversely affecting their biological properties. The investigated treatments offer numerous advantages over previous physical/chemical modifications and thus are expected to expand the utility of chitosan fibers with tunable mechanical properties in various tissue engineering applications.


Assuntos
Materiais Biocompatíveis/química , Células Endoteliais/citologia , Células Endoteliais/fisiologia , Hidrogéis/química , Engenharia Tecidual/métodos , Animais , Proliferação de Células , Sobrevivência Celular/fisiologia , Células Cultivadas , Módulo de Elasticidade , Teste de Materiais , Suínos , Resistência à Tração
9.
J Mech Behav Biomed Mater ; 5(1): 171-80, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22100092

RESUMO

Chitosan is being widely studied for tissue engineering applications due to its biocompatibility and biodegradability. However, its use in load-bearing applications is limited due to low mechanical properties. In this study, we investigated the effectiveness of a chitosan fiber reinforcement approach to enhancing the mechanical properties of chitosan scaffolds. Chitosan fibers were fabricated using a solution extrusion and neutralization method and incorporated into porous chitosan scaffolds. The effects of fiber/scaffold mass ratio, fiber mechanical properties and fiber length on scaffold mechanical properties were studied. The results showed that incorporating fibers improved scaffold strength and stiffness in proportion to the fiber/scaffold mass ratio. A fiber-reinforced, heart valve scaffold achieved leaflet tensile strength values of 220±17 kPa, comparable to the radial values of human pulmonary valve leaflets. Additionally, the effects of 2 mm fibers were found to be up to threefold greater than 10 mm fibers at identical mass ratios. Heparin crosslinking of fibers produced a reduction in fiber strength, and thus failed to produce additional improvements to fiber-reinforced scaffold properties. Despite this reduction in fiber strength, heparin-modified fibers still improved the mechanical properties of reinforced scaffolds, but to a lesser extent than unmodified fibers. The results demonstrate that chitosan fiber reinforcement can be used to achieve porous chitosan scaffold strength approaching that of tissue, and that fiber length and mechanical properties are important parameters in defining the degree of mechanical improvement.


Assuntos
Quitosana/química , Valvas Cardíacas/citologia , Fenômenos Mecânicos , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Animais , Quitosana/metabolismo , Liofilização , Heparina/metabolismo , Humanos , Porosidade
10.
J Biomed Mater Res A ; 96(4): 682-92, 2011 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-21268241

RESUMO

Application of umbilical cord blood (UCB) transplantation in adults as a treatment post-chemotherapy is hampered due to delayed platelet recovery. A potential solution suggested is the transfusion of ex vivo expanded megakaryocytes (Mks) from hematopoietic stem cells (HSCs). Alternatively, large-scale production of platelets in vitro has also been attempted with the goal of transfusing them into patients with thrombocytopenia. Glycosaminoglycans (GAGs) have been shown to influence the proliferation and differentiation of HSCs. This study sought to examine the effects of immobilized GAGs on the expansion, apoptosis, and platelet release activity of CD41a+ Mk progenitors in vitro. Freshly isolated HSCs from UCB were cultured in serum-free media supplemented with thrombopoietin on GAG-derivatized chitosan membranes for 17 days. Controls consisted of uncoated and chitosan-coated wells. Wells were demidepopulated at periodic intervals and analyzed by flow cytometry. Heparin and dermatan sulfate surfaces significantly enhanced total cell and Mk cell expansion (p < 0.05) compared to both the controls. The apoptotic Mk fraction was significantly lower on GAG surfaces (p < 0.05) compared to the polystyrene control during the early stages of the culture (days 7 and 11). However, by day 17, the apoptotic Mk fraction was comparable on all surfaces. The cumulative number of platelets generated on dermatan sulfate and heparan sulfate surfaces was significantly higher (p < 0.05) than on both the controls. These results suggest that immobilized GAGs delay Mk apoptosis and thereby enhance Mk expansion and platelet production.


Assuntos
Plaquetas/metabolismo , Glicosaminoglicanos/metabolismo , Células Progenitoras de Megacariócitos/metabolismo , Adulto , Animais , Antígenos CD34/metabolismo , Plaquetas/citologia , Proliferação de Células , Células Cultivadas , Sangue Fetal/citologia , Glicosaminoglicanos/química , Transplante de Células-Tronco Hematopoéticas , Humanos , Células Progenitoras de Megacariócitos/citologia , Células Progenitoras de Megacariócitos/transplante , Ratos , Ratos Sprague-Dawley
11.
Biotechnol Prog ; 26(6): 1567-79, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20967921

RESUMO

In this work, a novel optimization-based metabolic control analysis (OMCA) method is introduced for reducing data requirement for metabolic control analysis (MCA). It is postulated that using the optimal control approach, the fluxes in a metabolic network are correlated to metabolite concentrations and enzyme activities as a state-feedback control system that is optimal with respect to a homeostasis objective. It is then shown that the optimal feedback gains are directly related to the elasticity coefficients (ECs) of MCA. This approach requires determination of the relative "importance" of metabolites and fluxes for the system, which is possible with significantly reduced experimental data, as compared with typical MCA requirements. The OMCA approach is applied to a top-down control model of glycolysis in hepatocytes. It is statistically demonstrated that the OMCA model is capable of predicting the ECs observed experimentally with few exceptions. Further, an OMCA-based model reconciliation study shows that the modification of four assumed stoichiometric coefficients in the model can explain most of the discrepancies, with the exception of elasticities with respect to the NADH/NAD ratio.


Assuntos
Algoritmos , Simulação por Computador , Enzimas/metabolismo , Modelos Biológicos , Cinética
12.
Acta Biomater ; 6(6): 2126-31, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19925888

RESUMO

Cell and tissue responses to polymeric materials are orchestrated in part by the conformations of adsorbed plasma proteins. Thus, the chemical properties of a polymer membrane that govern protein adsorption behavior can play an important role in determining the biological properties of tissue engineered scaffolds derived from that polymer. In this study, we explored the role of membrane thickness as a factor influencing cell adhesion and proliferation on chitosan membranes with and without covalently attached glycosaminoglycans. Rat mesenchymal stem cells (MSCs) cultured on chitosan membranes of various thicknesses demonstrated significantly improved cell adhesion, spreading and proliferation as membrane thickness was increased. Rat hepatocytes displayed increased spreading on the substrate with increasing membrane thickness, similar to MSCs. Increased thickness reduced the overall crystallinity of the membrane, and the data indicate that the improved cellular responses were likely due to enhanced adsorption of serum vitronectin, presumably due to reduced membrane crystallinity. These results demonstrate that membrane thickness is an important design variable that can be manipulated in chitosan-based scaffolds to achieve enhanced cell spreading, proliferation and function.


Assuntos
Materiais Biocompatíveis/química , Quitosana/química , Membranas Artificiais , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , Alicerces Teciduais , Animais , Adesão Celular , Movimento Celular , Proliferação de Células , Células Cultivadas , Masculino , Teste de Materiais , Ratos , Ratos Sprague-Dawley , Propriedades de Superfície
13.
Tissue Eng Part A ; 15(11): 3499-512, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19456238

RESUMO

Mesenchymal stem cells (MSCs) are adult stem cells with potential for multilineage differentiation. They represent an attractive cell source alternative to embryonic stem cells for therapeutic applications. Optimal utilization of MSCs for tissue engineering requires improved biomaterials that can enhance their growth and direct differentiation. The biological activity of glycosaminoglycans (GAGs) has been previously exploited for use in tissue engineering applications. In this study, MSC proliferation and differentiation was studied on GAG-derivatized chitosan membranes. The GAGs included heparin, heparan sulfate, dermatan sulfate, chondroitin 4-sulfate, chondroitin 6-sulfate, and hyaluronic acid. The covalent GAG immobilization method and amount of immobilized GAG were varied. It was found that MSC growth increased as much as fivefold on GAG-immobilized surfaces compared to tissue culture plastic and chitosan-only controls. The MSC growth rates increased significantly with increasing GAG density on the culture surfaces. The MSC proliferation rates on heparin, heparan sulfate, dermatan sulfate, and chondroitin 6-sulfate exhibited nonlinear increases with the level of fibronectin binding on these surfaces. In contrast, MSC proliferation on hyaluronic acid and chondroitin 4-sulfate was found to be independent of fibronectin or vitronectin binding on the surfaces, suggesting that these GAGs influenced MSC proliferation through different mechanisms. In conclusion, the results indicate that GAG immobilization on chitosan scaffolds provides an effective means of manipulating MSC proliferation and has promising potential for directing MSC differentiation in tissue engineering applications employing chitosan.


Assuntos
Materiais Biocompatíveis/química , Quitosana/química , Glicosaminoglicanos/química , Glicosaminoglicanos/farmacologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , Engenharia Tecidual/métodos , Absorção , Adsorção , Animais , Técnicas de Cultura de Células/métodos , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Cristalização/métodos , Matriz Extracelular/química , Masculino , Teste de Materiais , Células-Tronco Mesenquimais/efeitos dos fármacos , Porosidade , Ratos , Ratos Sprague-Dawley , Propriedades de Superfície
14.
Acta Biomater ; 5(5): 1575-81, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19231304

RESUMO

The demand for biodegradable implant materials has fueled interest in chitosan as a biomaterial. In previous work, branched chitosans were synthesized and structurally characterized. In this study the biological properties of branched chitosans were explored. Branched chitosans were synthesized by grafting low molecular weight chitosan chains (1.6, 16 and 80 kDa) to high molecular weight (600 kDa) linear chitosans via reductive amination. Films of the branched materials were evaluated with regard to: lysozyme-mediated degradation; protein adsorption; cell adhesion and proliferation. Branched chitosan with a 1.6 kDa branch length exhibited higher degradation rates than either linear or higher branch length materials. Branched chitosans also exhibited reduced adsorption of bovine serum albumin that was more pronounced with thicker films. Branched chitosans supported proliferation of rat endothelial cells, but growth rates were significantly lower than on linear chitosan. The results of this study demonstrate that control of many aspects of chitosan's physical and biological properties can be achieved by changes in molecular architecture.


Assuntos
Proteínas Sanguíneas/metabolismo , Quitosana/química , Quitosana/farmacologia , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Adsorção/efeitos dos fármacos , Animais , Bovinos , Proliferação de Células/efeitos dos fármacos , Fibronectinas/metabolismo , Muramidase , Ratos , Soroalbumina Bovina/metabolismo , Soluções , Vitronectina/metabolismo , Água
15.
J Biomed Mater Res A ; 86(1): 98-107, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17941019

RESUMO

In vitro expansion of hematopoietic stem cells (HSCs) has been employed to obtain sufficient numbers of stem cells for successful engraftment after HSC transplantation. A three-dimensional perfusion bioreactor system with a heparin-chitosan scaffold was designed and evaluated for its capability to support maintenance and expansion of HSCs. Porous chitosan scaffolds were fabricated by a freeze-drying technique and N-desulfated heparin was covalently immobilized within the scaffolds using carbodiimide chemistry. CD34+ HSCs isolated from umbilical cord blood by immunomagnetic separation were cultured within the porous scaffold in a perfusion bioreactor system. Control cultures were maintained on dishes coated with similar heparin-chitosan films. Oxygen uptake was measured during the culture period. After 7 days of culture, scaffolds were harvested for analysis. Cellular phenotype and HSC characteristics were evaluated via flow cytometry and colony forming unit assays. The results indicate good cell retention and proliferation within the perfused scaffolds. Oxygen consumption in the perfusion bioreactor system increased continuously during the culture, indicating steady cell growth. Cells from the perfused scaffold cultures showed higher percentages of primitive progenitors and exhibited superior colony forming unit performance as compared to cells from static cultures. In addition, perfusion culture at low oxygen (5%) enhanced the expansion of CD34+ cells and colony-forming activity compared to high oxygen (19%) cultures. The results suggest that perfusion culture of cord blood CD34+ cells under bone marrow-like conditions enhances HSC expansion compared to static cultures.


Assuntos
Materiais Biocompatíveis/química , Técnicas de Cultura de Células/instrumentação , Técnicas de Cultura de Células/métodos , Sangue Fetal/citologia , Glicosaminoglicanos/química , Células-Tronco/citologia , Antígenos CD34/biossíntese , Reatores Biológicos , Quitosana/química , Desenho de Equipamento , Heparina/química , Humanos , Oxigênio/metabolismo , Consumo de Oxigênio , Cordão Umbilical/citologia
16.
Artigo em Inglês | MEDLINE | ID: mdl-18097786

RESUMO

The production of a fully functional bioartificial liver assist device (BLAD) would greatly enhance available treatment options for patients suffering from acute liver failure. Currently, inadequate oxygen provision to hepatocytes seeded within hollow fiber bioreactors hampers development of a viable hollow fiber-based BLAD. Experimentally, oxygen provision to primary rat hepatocytes cultured within hollow fiber bioreactors was measured, it was observed that supplementation with an oxygen carrier (bovine red blood cells at approximately 2% human hematocrit) did not significantly improve oxygenation compared to the absence of an oxygen carrier. Therefore, an oxygen transport model of an individual hollow fiber within the bioreactor was developed and simulated (up to approximately 10% human hematocrit) to more fully examine the effect of oxygen carrier supplementation on oxygenation within the bioreactor. The modeling analysis, supported via the experimental results, was utilized to predict optimal bioreactor operating conditions for the delivery of in vivo-like oxygen gradients to cultured hepatocytes in clinically relevant settings.


Assuntos
Reatores Biológicos , Sistemas de Liberação de Medicamentos/métodos , Hemoglobinas/farmacologia , Hepatócitos/metabolismo , Oxigênio/administração & dosagem , Animais , Técnicas de Cultura de Células/instrumentação , Respiração Celular , Células Cultivadas , Simulação por Computador , Desenho de Equipamento , Hepatócitos/efeitos dos fármacos , Fígado Artificial , Modelos Biológicos , Oxigênio/metabolismo , Ratos
17.
J Biomed Mater Res A ; 82(1): 201-12, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17266018

RESUMO

Chitosan continues to be studied as a promising biomaterial for tissue repair and regeneration applications. However, chitosan structures show a large reduction in tensile strength in the wet state. Methods for improving the wet strength of chitosan materials may broaden its applicability as a tissue scaffold for applications requiring significant load bearing capacity. In this study, the role of molecular architecture in defining the mechanical properties of hydrated chitosan membranes was examined. Specifically, branched chitosan molecules were synthesized with a range of branch lengths and branch densities. Physical and mechanical properties were characterized using viscometry, FTIR spectroscopy, and tensile testing measurements, and the results were correlated with the postulated architecture of the linear and branched chitosan materials. Both branch density and branch length were found to influence the mechanical properties of chitosan membranes. For example, high-molecular-weight (600 kDa) chitosans grafted with 80 kDa branches exhibited up to twofold increases in both tensile strength and extensibility. FTIR results indicated that these increases correlated with enhanced levels of hydrogen bonding in the branched materials. Vascular smooth muscle cells cultured on cast membranes of the branched chitosans exhibited no differences in adhesion or spreading as compared to the linear polymer. The results indicate that the mechanical properties of chitosan materials can be improved by the induction of a branched molecular architecture.


Assuntos
Materiais Biocompatíveis/química , Quitosana/química , Animais , Fenômenos Biomecânicos , Adesão Celular , Proliferação de Células , Ligação de Hidrogênio , Técnicas In Vitro , Teste de Materiais , Estrutura Molecular , Peso Molecular , Miócitos de Músculo Liso/citologia , Ratos , Reologia , Espectroscopia de Infravermelho com Transformada de Fourier , Resistência à Tração , Água/química
18.
Biotechnol Bioeng ; 97(3): 622-37, 2007 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-17058287

RESUMO

Using optimization based methods to predict fluxes in metabolic flux balance models has been a successful approach for some microorganisms, enabling construction of in silico models and even inference of some regulatory motifs. However, this success has not been translated to mammalian cells. The lack of knowledge about metabolic objectives in mammalian cells is a major obstacle that prevents utilization of various metabolic engineering tools and methods for tissue engineering and biomedical purposes. In this work, we investigate and identify possible metabolic objectives for hepatocytes cultured in vitro. To achieve this goal, we present a special data-mining procedure for identifying metabolic objective functions in mammalian cells. This multi-level optimization based algorithm enables identifying the major fluxes in the metabolic objective from MFA data in the absence of information about critical active constraints of the system. Further, once the objective is determined, active flux constraints can also be identified and analyzed. This information can be potentially used in a predictive manner to improve cell culture results or clinical metabolic outcomes. As a result of the application of this method, it was found that in vitro cultured hepatocytes maximize oxygen uptake, coupling of urea and TCA cycles, and synthesis of serine and urea. Selection of these fluxes as the metabolic objective enables accurate prediction of the flux distribution in the system given a limited amount of flux data; thus presenting a workable in silico model for cultured hepatocytes. It is observed that an overall homeostasis picture is also emergent in the findings.


Assuntos
Técnicas de Cultura de Células/métodos , Fenômenos Fisiológicos Celulares , Simulação por Computador , Hepatócitos/metabolismo , Células Cultivadas , Biologia Computacional/métodos , Metabolismo Energético/fisiologia , Hepatócitos/citologia
19.
J Biomed Mater Res A ; 75(2): 316-23, 2005 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-16059890

RESUMO

Fibronectin (Fn) is a matrix protein known to induce cell attachment and spreading through its cell binding site and related synergy sites. Fn-coated surfaces are therefore useful in tissue engineering and other cell contacting applications, but a problem with many immobilization strategies is a random distribution of molecular orientations. We sought to control Fn orientation, and thus enhance the availability of its cell binding site, by immobilizing Fn via a carboxymethyl dextran layer onto which are chemically attached monoclonal antibodies specific to a region near to Fn's C terminus (and thus away from the cell binding site). Using optical waveguide lightmode spectroscopy, we show the presence of chemically coupled antibodies to yield a considerably denser and thicker Fn layer, consistent with a more vertically aligned protein. Human umbilical vein endothelial cells spread significantly faster, and in a more spherically symmetric way, on an oriented Fn layer (i.e., in the presence of immobilized monoclonal antibodies) as compared with a control Fn layer (i.e., in the absence of bound antibodies). However, we observe human umbilical vein endothelial cell spreading on the oriented Fn layer to be similar to that on a Fn layer in the absence of a carboxymethyl dextran layer, suggesting that although orienting Fn is a promising strategy, coupling strategies using linkers other than dextran may be needed.


Assuntos
Anticorpos Monoclonais/metabolismo , Materiais Revestidos Biocompatíveis/química , Células Endoteliais/metabolismo , Fibronectinas/química , Adsorção , Sítios de Ligação , Movimento Celular , Células Cultivadas , Materiais Revestidos Biocompatíveis/metabolismo , Dextranos/química , Dextranos/metabolismo , Células Endoteliais/citologia , Endotélio Vascular/citologia , Fibronectinas/metabolismo , Humanos , Teste de Materiais , Ligação Proteica , Fatores de Tempo , Veias Umbilicais/anatomia & histologia
20.
J Biomed Mater Res ; 59(3): 585-90, 2002 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-11774317

RESUMO

Chitosan scaffolds appear to be suitable for a variety of tissue engineering applications. This study addressed the biocompatibility of chitosan in a mouse implantation model. Porous chitosan scaffolds were implanted in mice, and animals were sacrificed after 1, 2, 4, 8, or 12 weeks. Macroscopic inspection of the implantation site revealed no pathological inflammatory responses. Histological assessment indicated marked neutrophil accumulation within the implant, which resolved with increasing implantation time. Gram staining and limulus assays revealed no evidence of infection or endotoxin. Collagen was observed within the chitosan pore spaces, indicating that connective tissue matrix was deposited within the implant. Angiogenic activity associated with the external implant surface was also observed. Cellular immune responses were determined by lymphocyte proliferation assays, and antibody responses were measured using ELISA techniques. These assays indicated a very low incidence of chitosan-specific reactions. Although there was a large migration of neutrophils into the implantation area, there were minimal signs of any inflammatory reaction to the material itself. This preliminary study demonstrates that chitosan has a high degree of biocompatibility in this animal model. Overall, the findings suggest that chitosan may be suitable for the development of implantable materials.


Assuntos
Materiais Biocompatíveis/farmacologia , Substitutos Ósseos/química , Quitina/análogos & derivados , Quitina/farmacologia , Animais , Materiais Biocompatíveis/normas , Substitutos Ósseos/farmacologia , Divisão Celular , Quitina/uso terapêutico , Quitosana , Colágeno/metabolismo , Feminino , Imunidade Celular , Camundongos , Camundongos Endogâmicos BALB C , Modelos Animais , Neovascularização Fisiológica , Neutrófilos , Implantação de Prótese , Engenharia Tecidual/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...