Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Cell Mol Life Sci ; 71(11): 2103-18, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24399290

RESUMEN

This review will focus on two elements that are essential for functional arterial regeneration in vitro: the mechanical environment and the bioreactors used for tissue growth. The importance of the mechanical environment to embryological development, vascular functionality, and vascular graft regeneration will be discussed. Bioreactors generate mechanical stimuli to simulate biomechanical environment of arterial system. This system has been used to reconstruct arterial grafts with appropriate mechanical strength for implantation by controlling the chemical and mechanical environments in which the grafts are grown. Bioreactors are powerful tools to study the effect of mechanical stimuli on extracellular matrix architecture and mechanical properties of engineered vessels. Hence, biomimetic systems enable us to optimize chemo-biomechanical culture conditions to regenerate engineered vessels with physiological properties similar to those of native arteries. In addition, this article reviews various bioreactors designed especially to apply axial loading to engineered arteries. This review will also introduce and examine different approaches and techniques that have been used to engineer biologically based vascular grafts, including collagen-based grafts, fibrin-gel grafts, cell sheet engineering, biodegradable polymers, and decellularization of native vessels.


Asunto(s)
Arterias/patología , Prótesis Vascular , Células Endoteliales/citología , Ingeniería de Tejidos , Arterias/inmunología , Arterias/cirugía , Materiales Biocompatibles/metabolismo , Fenómenos Biomecánicos , Reactores Biológicos , Colágeno/metabolismo , Células Endoteliales/fisiología , Endotelio Vascular/citología , Endotelio Vascular/fisiología , Matriz Extracelular/metabolismo , Matriz Extracelular/fisiología , Fibrina/metabolismo , Supervivencia de Injerto/inmunología , Humanos , Mecanotransducción Celular , Técnicas de Cultivo de Tejidos , Andamios del Tejido
2.
Bone ; 43(2): 292-301, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18550463

RESUMEN

A three dimensional (3D) computational simulation of dynamic process of trabecular bone remodeling was developed with all the parameters derived from physiological and clinical data. Contributions of the microstructural bone formation deficits: trabecular plate perforations, trabecular rod breakages, and isolated bone fragments, to the rapid bone loss and disruption of trabecular microarchitecture during menopause were studied. Eighteen human trabecular bone samples from femoral neck (FN) and spine were scanned using a micro computed tomography (microCT) system. Bone resorption and formation were simulated as a computational cycle corresponding to 40-day resorption/160-day formation. Resorption cavities were randomly created over the bone surface according to the activation frequency, which was strictly based on clinical data. Every resorption cavity was refilled during formation unless it caused trabecular plate perforation, trabecular rod breakage or isolated fragments. A 20-year-period starting 5 years before and ending 15 years after menopause was simulated for each specimen. Elastic moduli, standard and individual trabeculae segmentation (ITS)-based morphological parameters were evaluated for each simulated 3D image. For both spine and FN groups, the time courses of predicted bone loss pattern by microstructural bone formation deficits were fairly consistent with the clinical measurements. The percentage of bone loss due to trabecular plate perforation, trabecular rod breakage, and isolated bone fragments were 73.2%, 18.9% and 7.9% at the simulated 15 years after menopause. The ITS-based plate fraction (pBV/BV), mean plate surface area (pTb.S), plate number density (pTb.N), and mean rod thickness (rTb.Th) decreased while rod fraction (rBV/BV) and rod number density (rTb.N) increased after the simulated menopause. The dynamic bone remodeling simulation based on microstructural bone formation deficits predicted the time course of menopausal bone loss pattern of spine and FN. Microstructural plate perforation could be the primary cause of menopausal trabecular bone loss. The combined effect of trabeculae perforation, breakage, and isolated fragments resulted in fewer and smaller trabecular plates and more but thinner trabecular rods.


Asunto(s)
Remodelación Ósea , Huesos/fisiología , Simulación por Computador , Menopausia/fisiología , Anciano , Fenómenos Biomecánicos , Femenino , Fémur/fisiología , Humanos , Masculino , Persona de Mediana Edad , Tamaño de los Órganos , Columna Vertebral/fisiología
3.
Tissue Eng Part C Methods ; 22(6): 524-33, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27108525

RESUMEN

Tissue-engineered blood vessels (TEVs) are typically produced using the pulsatile, uniaxial circumferential stretch to mechanically condition and strengthen the arterial grafts. Despite improvements in the mechanical integrity of TEVs after uniaxial conditioning, these tissues fail to achieve critical properties of native arteries such as matrix content, collagen fiber orientation, and mechanical strength. As a result, uniaxially loaded TEVs can result in mechanical failure, thrombus, or stenosis on implantation. In planar tissue equivalents such as artificial skin, biaxial loading has been shown to improve matrix production and mechanical properties. To date however, multiaxial loading has not been examined as a means to improve mechanical and biochemical properties of TEVs during culture. Therefore, we developed a novel bioreactor that utilizes both circumferential and axial stretch that more closely simulates loading conditions in native arteries, and we examined the suture strength, matrix production, fiber orientation, and cell proliferation. After 3 months of biaxial loading, TEVs developed a formation of mature elastic fibers that consisted of elastin cores and microfibril sheaths. Furthermore, the distinctive features of collagen undulation and crimp in the biaxial TEVs were absent in both uniaxial and static TEVs. Relative to the uniaxially loaded TEVs, tissues that underwent biaxial loading remodeled and realigned collagen fibers toward a more physiologic, native-like organization. The biaxial TEVs also showed increased mechanical strength (suture retention load of 303 ± 14.53 g, with a wall thickness of 0.76 ± 0.028 mm) and increased compliance. The increase in compliance was due to combinatorial effects of mature elastic fibers, undulated collagen fibers, and collagen matrix orientation. In conclusion, biaxial stretching is a potential means to regenerate TEVs with improved matrix production, collagen organization, and mechanical properties.


Asunto(s)
Arterias/citología , Colágeno/química , Tejido Elástico/citología , Regeneración/fisiología , Estrés Mecánico , Ingeniería de Tejidos/métodos , Animales , Arterias/química , Reactores Biológicos , Tejido Elástico/química , Matriz Extracelular/metabolismo , Humanos
4.
Tissue Eng Part A ; 22(17-18): 1086-97, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27520928

RESUMEN

Here we report the creation of a novel tracheal construct in the form of an engineered, acellular tissue-stent biocomposite trachea (TSBT). Allogeneic or xenogeneic smooth muscle cells are cultured on polyglycolic acid polymer-metal stent scaffold leading to the formation of a tissue comprising cells, their deposited collagenous matrix, and the stent material. Thorough decellularization then produces a final acellular tubular construct. Engineered TSBTs were tested as end-to-end tracheal replacements in 11 rats and 3 nonhuman primates. Over a period of 8 weeks, no instances of airway perforation, infection, stent migration, or erosion were observed. Histological analyses reveal that the patent implants remodel adaptively with native host cells, including formation of connective tissue in the tracheal wall and formation of a confluent, columnar epithelium in the graft lumen, although some instances of airway stenosis were observed. Overall, TSBTs resisted collapse and compression that often limit the function of other decellularized tracheal replacements, and additionally do not require any cells from the intended recipient. Such engineered TSBTs represent a model for future efforts in tracheal regeneration.


Asunto(s)
Bioprótesis , Ensayo de Materiales , Stents , Ingeniería de Tejidos , Andamios del Tejido/química , Tráquea , Animales , Bovinos , Chlorocebus aethiops , Humanos , Ratas
5.
Stem Cells Transl Med ; 3(12): 1535-43, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25378654

RESUMEN

The utility of human induced pluripotent stem cells (hiPSCs) to create tissue-engineered vascular grafts was evaluated in this study. hiPSC lines were first induced into a mesenchymal lineage via a neural crest intermediate using a serum-free, chemically defined differentiation scheme. Derived cells exhibited commonly known mesenchymal markers (CD90, CD105, and CD73 and negative marker CD45) and were shown to differentiate into several mesenchymal lineages (osteogenic, chondrogenic, and adipogenic). Functional vascular grafts were then engineered by culturing hiPSC-derived mesenchymal progenitor cells in a pulsatile bioreactor system over 8 weeks to induce smooth muscle cell differentiation and collagenous matrix generation. Histological analyses confirmed layers of calponin-positive smooth muscle cells in a collagen-rich matrix. Mechanical tests revealed that grafts had an average burst pressure of 700 mmHg, which is approximately half that of native veins. Additionally, studies revealed that karyotypically normal mesenchymal stem cell clones led to generation of grafts with predicted features of engineered vascular grafts, whereas derived clones having chromosomal abnormalities generated calcified vessel constructs, possibly because of cell apoptosis during culture. Overall, these results provide significant insight into the utility of hiPS cells for vascular graft generation. They pave the way for creating personalized, patient-specific vascular grafts for surgical applications, as well as for creating experimental models of vascular development and disease.


Asunto(s)
Prótesis Vascular , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Ingeniería de Tejidos/métodos , Antígenos de Diferenciación/biosíntesis , Línea Celular , Matriz Extracelular/metabolismo , Humanos
6.
Tissue Eng Part A ; 20(9-10): 1499-507, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24320793

RESUMEN

Tissue-engineered small-diameter vascular grafts have been developed as a promising alternative to native veins or arteries for replacement therapy. However, there is still a crucial need to improve the current approaches to render the tissue-engineered blood vessels more favorable for clinical applications. A completely biological blood vessel (3-mm inner diameter) was constructed by culturing a 50:50 mixture of bovine smooth muscle cells (SMCs) with neonatal human dermal fibroblasts in fibrin gels. After 30 days of culture under pulsatile stretching, the engineered blood vessels demonstrated an average burst pressure of 913.3±150.1 mmHg (n=6), a suture retention (53.3±15.4 g) that is suitable for implantation, and a compliance (3.1%±2.5% per 100 mmHg) that is comparable to native vessels. These engineered grafts contained circumferentially aligned collagen fibers, microfibrils and elastic fibers, and differentiated SMCs, mimicking a native artery. These promising mechanical and biochemical properties were achieved in a very short culture time of 30 days, suggesting the potential of co-culturing SMCs with fibroblasts in fibrin gels to generate functional small-diameter vascular grafts for vascular reconstruction surgery.


Asunto(s)
Prótesis Vascular , Vasos Sanguíneos/crecimiento & desarrollo , Fibrina/química , Fibroblastos/fisiología , Miocitos del Músculo Liso/fisiología , Ingeniería de Tejidos/instrumentación , Andamios del Tejido , Animales , Vasos Sanguíneos/citología , Bovinos , Células Cultivadas , Técnicas de Cocultivo , Fibroblastos/citología , Humanos , Miocitos del Músculo Liso/citología , Diseño de Prótesis , Ingeniería de Tejidos/métodos
7.
J Vis Exp ; (52)2011 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-21694696

RESUMEN

Much effort has been devoted to develop and advance the methodology to regenerate functional small-diameter arterial bypasses. In the physiological environment, both mechanical and chemical stimulation are required to maintain the proper development and functionality of arterial vessels. Bioreactor culture systems developed by our group are designed to support vessel regeneration within a precisely controlled chemo-mechanical environment mimicking that of native vessels. Our bioreactor assembly and maintenance procedures are fairly simple and highly repeatable. Smooth muscle cells (SMCs) are seeded onto a tubular polyglycolic acid (PGA) mesh that is threaded over compliant silicone tubing and cultured in the bioreactor with or without pulsatile stimulation for up to 12 weeks. There are four main attributes that distinguish our bioreactor from some predecessors. 1) Unlike other culture systems that simulate only the biochemical surrounding of native blood vessels, our bioreactor also creates a physiological pulsatile environment by applying cyclic radial strain to the vessels in culture. 2) Multiple engineered vessels can be cultured simultaneously under different mechanical conditions within a controlled chemical environment. 3) The bioreactor allows a mono layer of endothelial cells (EC) to be easily coated onto the luminal side of engineered vessels for animal implantation models. 4) Our bioreactor can also culture engineered vessels with different diameter size ranged from 1 mm to 3 mm, saving the effort to tailor each individual bioreactor to fit a specific diameter size. The engineered vessels cultured in our bioreactor resemble native blood vessels histologically to some degree. Cells in the vessel walls express mature SMC contractile markers such as smooth muscle myosin heavy chain (SMMHC). A substantial amount of collagen is deposited within the extracellular matrix, which is responsible for ultimate mechanical strength of the engineered vessels. Biochemical analysis also indicates that collagen content of engineered vessels is comparable to that of native arteries. Importantly, the pulsatile bioreactor has consistently regenerated vessels that exhibit mechanical properties that permit successful implantation experiments in animal models. Additionally, this bioreactor can be further modified to allow real-time assessment and tracking of collagen remodeling over time, non-invasively, using a non-linear optical microscopy (NLOM). To conclude, this bioreactor should serve as an excellent platform to study the fundamental mechanisms that regulate the regeneration of functional small-diameter vascular grafts.


Asunto(s)
Reactores Biológicos , Prótesis Vascular , Ingeniería de Tejidos/métodos , Animales , Colágeno/metabolismo , Células Endoteliales/citología , Células Endoteliales/metabolismo , Humanos , Músculo Liso/citología , Músculo Liso/metabolismo , Cadenas Pesadas de Miosina/metabolismo , Ácido Poliglicólico
8.
Biophys J ; 90(5): 1804-9, 2006 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-16339875

RESUMEN

Cells require optimal substrate stiffness for normal function and differentiation. The mechanisms for sensing matrix rigidity and durotaxis, however, are not clear. Here we showed that control, Shp2-/-, integrin beta1-/-, and talin1-/- cell lines all spread to a threefold greater area on fibronectin (FN)-coated rigid polyacrylamide surfaces than soft. In contrast, RPTPalpha-/- cells spread to the same area irrespective of rigidity on FN surfaces but spread 3x greater on rigid collagen IV-coated surfaces than soft. RPTPalpha and alphavbeta3 integrins were shown previously to be colocalized at leading edges and antibodies to alphavbeta3 blocked FN rigidity sensing. When FN beads were held with a rigid laser trap at the leading edge, stronger bonds to the cytoskeleton formed than when held with a soft trap; whereas back from the leading edge and in RPTPalpha-/- cells, weaker bonds were formed with both rigid and soft laser traps. From the rigidity of the trap, we calculate that a force of 10 pN generated in 1 s is sufficient to activate the rigidity response. We suggest that RPTPalpha and alphavbeta3 at the leading edge are critical elements for sensing FN matrix rigidity possibly through SFK activation at the edge and downstream signaling.


Asunto(s)
Matriz Extracelular/fisiología , Fibroblastos/citología , Fibroblastos/metabolismo , Integrina alfaVbeta3/metabolismo , Mecanotransducción Celular/fisiología , Proteínas Tirosina Fosfatasas/metabolismo , Receptores de Superficie Celular/metabolismo , Animales , Adhesión Celular/fisiología , Movimiento Celular/fisiología , Células Cultivadas , Elasticidad , Dureza , Ratones , Proteínas Tirosina Fosfatasas Clase 4 Similares a Receptores , Estrés Mecánico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA