Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Am J Nephrol ; 49(2): 156-164, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30677763

RESUMEN

BACKGROUND: Chronic hemodialysis requires a mode of vascular access through an arteriovenous fistula (AVF), a prosthetic arteriovenous graft (AVG), or a central venous catheter (CVC). AVF is recommended over AVG or CVC due to increased patency and decreased intervention rates for those that mature. AVG are preferred over CVC due to decreased infection and mortality risk. The aims of this study were to evaluate the lifespan of AVF and AVG in maturation, sustained access use, and abandonment. METHODS: The United States Renal Data System (USRDS), Medicare claims, and CROWNWeb were used to identify access placements. Patients with a first end-stage renal disease (ESRD) service from January 1, 2012 to June 30, 2014 with continuous coverage with Medicare as primary payer and ≥1 AVF or AVG placed after ESRD onset were included. Maturation was defined as the first use of the access for hemodialysis recorded in CROWNWeb. Sustained access use was defined as 3 consecutive months of use without catheter placement or replacement. Accesses that were never used at any time post-placement were considered abandoned. RESULTS: The cohort included 38,035 AVF placements and 12,789 AVG placements. Sixty-nine percent of AVF and 72% of AVG matured. Fifty-two percent of AVF and 51% of AVG achieved sustained access use. One quarter of AVF and 14% of AVG were abandoned without use as recorded in CROWNWeb. CONCLUSION: Although considered the gold standard for vascular access, only half of AVF and AVG placements achieved sustained access use. The USRDS database has inherent limitations but provides useful clinical insight into maturation, sustained use, and abandonment.


Asunto(s)
Derivación Arteriovenosa Quirúrgica/efectos adversos , Implantación de Prótesis Vascular/efectos adversos , Oclusión de Injerto Vascular/epidemiología , Fallo Renal Crónico/terapia , Diálisis Renal/efectos adversos , Reclamos Administrativos en el Cuidado de la Salud/estadística & datos numéricos , Adolescente , Adulto , Anciano , Bases de Datos Factuales/estadística & datos numéricos , Femenino , Estudios de Seguimiento , Oclusión de Injerto Vascular/etiología , Humanos , Masculino , Medicare/estadística & datos numéricos , Persona de Mediana Edad , Diálisis Renal/métodos , Resultado del Tratamiento , Estados Unidos/epidemiología , Grado de Desobstrucción Vascular , Adulto Joven
2.
J Surg Res ; 221: 143-151, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29229120

RESUMEN

BACKGROUND: Synthetic expanded polytetrafluorethylene (ePTFE) grafts are routinely used for vascular repair and reconstruction but prone to sustained bacterial infections. Investigational bioengineered human acellular vessels (HAVs) have shown clinical success and may confer lower susceptibility to infection. Here we directly compared the susceptibility of ePTFE grafts and HAV to bacterial contamination in a preclinical model of infection. MATERIALS AND METHODS: Sections (1 cm2) of ePTFE (n = 42) or HAV (n = 42) were inserted within bilateral subcutaneous pockets on the dorsum of rats and inoculated with Staphylococcus aureus (107 CFU/0.25 mL) or Escherichia coli (108 CFU/0.25 mL) before wound closure. Two weeks later, the implant sites were scored for abscess formation and explanted materials were halved for quantification of microbial recovery and histological analyses. RESULTS: The ePTFE implants had significantly higher abscess formation scores for both S. aureus and E. coli inoculations compared to that of HAV. In addition, significantly more bacteria were recovered from explanted ePTFE compared to HAV. Gram staining of explanted tissue sections revealed interstitial bacterial contamination within ePTFE, whereas no bacteria were identified in HAV tissue sections. Numerous CD45+ leukocytes, predominantly neutrophils, were found surrounding the ePTFE implants but minimal intact neutrophils were observed within the ePTFE matrix. The host cells surrounding and infiltrating the HAV explants were primarily nonleukocytes (CD45-). CONCLUSIONS: In an established animal model of infection, HAV was significantly less susceptible to bacterial colonization and abscess formation than ePTFE. The preclinical findings presented in this manuscript, combined with previously published clinical observations, suggest that bioengineered HAV may exhibit low rates of infection.


Asunto(s)
Prótesis Vascular , Infecciones/etiología , Politetrafluoroetileno , Infecciones Relacionadas con Prótesis/etiología , Injerto Vascular/efectos adversos , Animales , Escherichia coli , Masculino , Ratas Sprague-Dawley , Staphylococcus aureus
3.
Sci Transl Med ; 11(485)2019 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-30918113

RESUMEN

Traditional vascular grafts constructed from synthetic polymers or cadaveric human or animal tissues support the clinical need for readily available blood vessels, but often come with associated risks. Histopathological evaluation of these materials has shown adverse host cellular reactions and/or mechanical degradation due to insufficient or inappropriate matrix remodeling. We developed an investigational bioengineered human acellular vessel (HAV), which is currently being studied as a hemodialysis conduit in patients with end-stage renal disease. In rare cases, small samples of HAV were recovered during routine surgical interventions and used to examine the temporal and spatial pattern of the host cell response to the HAV after implantation, from 16 to 200 weeks. We observed a substantial influx of alpha smooth muscle actin (αSMA)-expressing cells into the HAV that progressively matured and circumferentially aligned in the HAV wall. These cells were supported by microvasculature initially formed by CD34+/CD31+ cells in the neoadventitia and later maintained by CD34-/CD31+ endothelial cells in the media and lumen of the HAV. Nestin+ progenitor cells differentiated into either αSMA+ or CD31+ cells and may contribute to early recellularization and self-repair of the HAV. A mesenchymal stem cell-like CD90+ progenitor cell population increased in number with duration of implantation. Our results suggest that host myogenic, endothelial, and progenitor cell repopulation of HAVs transforms these previously acellular vessels into functional multilayered living tissues that maintain blood transport and exhibit self-healing after cannulation injury, effectively rendering these vessels like the patient's own blood vessel.


Asunto(s)
Prótesis Vascular , Vasos Sanguíneos/citología , Vasos Sanguíneos/trasplante , Ingeniería de Tejidos/métodos , Injerto Vascular/métodos , Adulto , Anciano , Vasos Sanguíneos/crecimiento & desarrollo , Diferenciación Celular , Movimiento Celular , Células Cultivadas , Células Endoteliales/citología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Miocitos del Músculo Liso/citología , Neovascularización Fisiológica , Diálisis Renal , Análisis Espacio-Temporal , Células Madre/citología , Andamios del Tejido , Investigación Biomédica Traslacional , Dispositivos de Acceso Vascular
4.
Lancet ; 365(9477): 2122-4, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15964449

RESUMEN

Tissue engineering has made considerable progress in the past decade, but advances have stopped short of clinical application for most tissues. We postulated that an obstacle in engineering human tissues is the limited replicative capacity of adult somatic cells. To test this hypothesis, the effectiveness of telomerase expression to extend cellular lifespan was assessed in a model of human vascular tissue engineering. Telomerase expression in vascular cells isolated from elderly patients enabled the successful culture of engineered autologous blood vessels. Engineered vessels may one day provide a source of bypass conduit for patients with atherosclerotic disease.


Asunto(s)
Vasos Sanguíneos , Ingeniería de Tejidos/métodos , Anciano , Vasos Sanguíneos/enzimología , Técnicas de Cultivo de Célula , Proteínas de Unión al ADN/biosíntesis , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Vena Safena/citología , Telomerasa/biosíntesis , Transfección
5.
Tissue Eng ; 12(2): 291-300, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16548687

RESUMEN

It is well established that, in multicellular systems, conventional cryopreservation results in damaging ice formation, both in the cells and in the surrounding extracellular matrix. As an alternative to conventional cryopreservation, we performed a feasibility study using vitrification (ice-free cryopreservation) to cryopreserve tissue-engineered blood vessels. Fresh, frozen, and vitrified tissue-engineered blood vessels were compared using histological methods, cellular viability, and mechanical properties. Cryosubstitution methods were used to determine the location of ice in conventionally cryopreserved engineered vessels. Ice formation was negligible (0.0 +/- 0.0% of vessel area) in the vitrified specimens, and extensive (68.3 +/- 4.5% of vessel area) in the extracellular matrix of frozen specimens. The metabolic assay and TUNEL staining results indicated that vitrified tissue had similar viability to fresh controls. The contractility results for vitrified samples were >82.7% of fresh controls and, in marked contrast, the results for frozen samples were only 10.7% of fresh controls (p < 0.001). Passive mechanical testing revealed enhanced tissue strength after both freezing and vitrification. Vitrification is a feasible storage method for tissue-engineered blood vessel constructs, and their successful storage brings these constructs one step closer to clinical utility.


Asunto(s)
Materiales Biocompatibles/química , Criopreservación/métodos , Músculo Liso Vascular/citología , Animales , Apoptosis , Fenómenos Biomecánicos , Arterias Carótidas/citología , Arterias Carótidas/fisiología , Arterias Carótidas/ultraestructura , Adhesión Celular , Técnicas de Cultivo de Célula , Supervivencia Celular , Células Cultivadas , Medios de Cultivo/química , Endotelina-1/farmacología , Endotelio Vascular/citología , Endotelio Vascular/fisiología , Endotelio Vascular/ultraestructura , Estudios de Factibilidad , Congelación , Glucosa/metabolismo , Contracción Muscular/efectos de los fármacos , Músculo Liso Vascular/fisiología , Músculo Liso Vascular/ultraestructura , Soluciones Preservantes de Órganos , Papaverina/farmacología , Permeabilidad , Ácido Poliglicólico/química , Porcinos , Factores de Tiempo , Ingeniería de Tejidos/métodos
6.
Cell Transplant ; 14(6): 367-74, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16180655

RESUMEN

In many cases, the mechanical strengths of tissue-engineered arteries do not match the mechanical strengths of native arteries. Ultimate arterial strength is primarily dictated by collagen in the extracellular matrix, but collagen in engineered arteries is not as dense, as organized, or as mature as collagen in native arteries. One step in the maturation process of collagen is the formation of hydroxylysyl pyridinoline (HP) cross-links between and within collagen molecules. HP cross-link formation, which is triggered by the copper-activated enzyme lysyl oxidase, greatly increases collagen fibril stability and enhances tissue strength. Increased cross-link formation, in addition to increased collagen production, may yield a stronger engineered tissue. In this article, the effect of increasing culture medium copper ion concentration on engineered arterial tissue composition and mechanics was investigated. Engineered vessels grown in low copper ion concentrations for the first 4 weeks of culture, followed by higher copper ion concentrations for the last 3 weeks of culture, had significantly elevated levels of cross-link formation compared to those grown in low copper ion concentrations. In contrast, vessels grown in high copper ion concentrations throughout culture failed to develop higher collagen cross-link densities than those grown in low copper ion concentrations. Although the additional cross-linking of collagen in engineered vessels may provide collagen fibril stability and resistance to proteolysis, it failed to enhance global tissue strength.


Asunto(s)
Arterias Carótidas/fisiología , Cobre/farmacología , Matriz Extracelular , Ingeniería de Tejidos , Aminoácidos/metabolismo , Animales , Arterias Carótidas/citología , Arterias Carótidas/crecimiento & desarrollo , Colágeno/metabolismo , Matriz Extracelular/efectos de los fármacos , Técnicas de Cultivo de Órganos , Porcinos , Porcinos Enanos , Ingeniería de Tejidos/métodos
7.
Cell Transplant ; 14(10): 861-8, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16454361

RESUMEN

In many cases, the mechanical strengths of tissue-engineered arteries do not match the mechanical strengths of native arteries. Ultimate arterial strength is primarily dictated by collagen in the extracellular matrix. but collagen in engineered arteries is not as dense, as organized, or as mature as collagen in native arteries. One step in the maturation process of collagen is the formation of hydroxylysyl pyridinoline (HP) cross-links between and within collagen molecules. HP cross-link formation, which is triggered by the copper-activated enzyme lysyl oxidase, greatly increases collagen fibril stability and enhances tissue strength. Increased cross-link formation, in addition to increased collagen production, may yield a stronger engineered tissue. In this article, the effect of increasing culture medium copper ion concentration on engineered arterial tissue composition and mechanics was investigated. Engineered vessels grown in low copper ion concentrations for the first 4 weeks of culture, followed by higher copper ion concentrations for the last 3 weeks of culture, had significantly elevated levels of cross-link formation compared to those grown in low copper ion concentrations. In contrast, vessels grown in high copper ion concentrations throughout culture failed to develop higher collagen cross-link densities than those grown in low copper ion concentrations. Although the additional cross-linking of collagen in engineered vessels may provide collagen fibril stability and resistance to proteolysis, it failed to enhance global tissue strength.


Asunto(s)
Arterias Carótidas/fisiología , Cobre/farmacología , Matriz Extracelular , Ingeniería de Tejidos , Aminoácidos/metabolismo , Animales , Arterias Carótidas/citología , Arterias Carótidas/crecimiento & desarrollo , Colágeno/metabolismo , Matriz Extracelular/química , Matriz Extracelular/efectos de los fármacos , Técnicas de Cultivo de Órganos , Porcinos , Porcinos Enanos , Ingeniería de Tejidos/métodos
8.
Cell Transplant ; 12(6): 659-66, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-14579934

RESUMEN

More than 570,000 coronary artery bypass grafts are implanted each year, creating an important demand for small-diameter vascular grafts. For patients who lack adequate internal mammary artery or saphenous vein, tissue-engineered arteries may prove useful. However, the time needed to tissue engineer arteries (7 weeks or more) is too long for many patients. Decellularized cadaveric human arteries are another possible source of vascular conduit, but limited availability and the potential for disease transmission limit their widespread use. In contrast, decellularized tissue-engineered arteries could serve as grafts for immediate implantation, as scaffolds onto which patients' cells could be seeded, or as carriers for genetically engineered cells to aid cell transplantation. The goal of this study was to quantify the effects of decellularization on vascular matrix and mechanical properties. Specifically, we compared cellular elimination, extracellular matrix retention, and mechanical characteristics of porcine carotid arteries before and after treatment with three decellularization methods. In addition, for the first time, tissue-engineered arteries were decellularized. Decellularized native arteries were also used as a scaffold onto which vascular cells were seeded. These studies identified a decellularization method for native and engineered arteries that maximized cellular elimination, without greatly compromising mechanical integrity. We showed that engineered tissues could be decellularized, and demonstrated the feasibility of reseeding decellularized vessels with vascular cells.


Asunto(s)
Materiales Biocompatibles/farmacología , Arterias Carótidas/trasplante , Separación Celular/métodos , Ingeniería de Tejidos/métodos , Trasplante de Tejidos/métodos , Trasplante Heterólogo/métodos , Animales , Órganos Artificiales/tendencias , Materiales Biocompatibles/uso terapéutico , Arterias Carótidas/citología , Arterias Carótidas/inmunología , Bovinos , Colágeno/metabolismo , Puente de Arteria Coronaria/métodos , ADN/metabolismo , Matriz Extracelular/inmunología , Matriz Extracelular/metabolismo , Humanos , Músculo Liso Vascular/citología , Músculo Liso Vascular/fisiología , Músculo Liso Vascular/trasplante , Estrés Mecánico , Sus scrofa , Soporte de Peso/fisiología
10.
Trends Cardiovasc Med ; 21(3): 83-9, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22626247

RESUMEN

Surgical treatments for vascular disease have progressed during the past century from autologous bypass conduits to synthetic materials, animal-derived tissues, cryopreserved grafts, and, finally, bioengineered conduits. In all cases, alternative vascular grafting materials have been developed with the goal of treating patients who have severe vascular disease requiring bypass but who have no suitable autologous conduit. Synthetic vascular grafts, animal-derived tissues, and cryopreserved grafts all have drawbacks in terms of availability and functionality that have limited their routine clinical adoption. Although bioengineered vascular graft technologies remain early and highly investigational, they have the potential to revolutionize the way in which severe vascular disease is treated. However, before they can have a clinical impact, bioengineered grafts must be available immediately and "off-the-shelf."


Asunto(s)
Bioingeniería , Bioprótesis , Implantación de Prótesis Vascular/instrumentación , Prótesis Vascular , Diseño de Prótesis , Enfermedades Vasculares/cirugía , Animales , Implantación de Prótesis Vascular/efectos adversos , Humanos , Factores de Tiempo , Listas de Espera
11.
J Cardiovasc Transl Res ; 4(5): 674-82, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21748530

RESUMEN

Intimal hyperplasia is one of the prominent failure mechanisms for arteriovenous fistulas and arteriovenous access grafts. Human tissue-engineered vascular grafts (TEVGs) were implanted as arteriovenous grafts in a novel baboon model. Ultrasound was used to monitor flow rates and vascular diameters throughout the study. Intimal hyperplasia in the outflow vein of TEVGs was assessed at the anastomosis and at juxta-anastomotic regions via histological analysis, and was compared to intimal hyperplasia with polytetrafluoroethylene (PTFE) grafts in the baboon model and in literature reports from other animal models. Less venous intimal hyperplasia was observed in histological sections with arteriovenous TEVGs than with arteriovenous PTFE grafts. TEVGs were associated with a mild, noninflammatory intimal hyperplasia. The extent of intimal tissue that formed with TEVG placement correlated with the rate of blood flow through tissue engineered vascular grafts at 2 weeks postimplant. Outflow vein dilatation was observed with increased flow rate. Both mid-graft flow rates and outflow vein diameters reached a plateau by week 4, which suggested that venous remodeling and intimal hyperplasia largely occurred within the first 4 weeks of implant in the baboon model. Given their compliant and noninflammatory nature, TEVGs appear resistant to triggers for venous intimal hyperplasia that are common for PTFE arteriovenous grafts, including (1) abundant proinflammatory macrophage populations that are associated with PTFE grafts and (2) compliance mismatch between PTFE grafts and the outflow vein. Our findings suggest that arteriovenous TEVGs develop only a mild form of venous intimal hyperplasia, which results from the typical hemodynamic changes that are associated with arteriovenous settings.


Asunto(s)
Derivación Arteriovenosa Quirúrgica/instrumentación , Implantación de Prótesis Vascular/instrumentación , Prótesis Vascular , Proliferación Celular , Oclusión de Injerto Vascular/prevención & control , Ingeniería de Tejidos/métodos , Túnica Íntima/patología , Animales , Humanos
12.
Sci Transl Med ; 3(68): 68ra9, 2011 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-21289273

RESUMEN

Autologous or synthetic vascular grafts are used routinely for providing access in hemodialysis or for arterial bypass in patients with cardiovascular disease. However, some patients either lack suitable autologous tissue or cannot receive synthetic grafts. Such patients could benefit from a vascular graft produced by tissue engineering. Here, we engineer vascular grafts using human allogeneic or canine smooth muscle cells grown on a tubular polyglycolic acid scaffold. Cellular material was removed with detergents to render the grafts nonimmunogenic. Mechanical properties of the human vascular grafts were similar to native human blood vessels, and the grafts could withstand long-term storage at 4 °C. Human engineered grafts were tested in a baboon model of arteriovenous access for hemodialysis. Canine grafts were tested in a dog model of peripheral and coronary artery bypass. Grafts demonstrated excellent patency and resisted dilatation, calcification, and intimal hyperplasia. Such tissue-engineered vascular grafts may provide a readily available option for patients without suitable autologous tissue or for those who are not candidates for synthetic grafts.


Asunto(s)
Prótesis Vascular , Ingeniería de Tejidos/métodos , Injerto Vascular/métodos , Adolescente , Adulto , Animales , Materiales Biocompatibles , Cadáver , Células Cultivadas , Perros , Humanos , Masculino , Ensayo de Materiales , Persona de Mediana Edad , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Papio anubis , Estrés Mecánico , Andamios del Tejido , Adulto Joven
13.
Cell Transplant ; 18(8): 915-21, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19500474

RESUMEN

It has been shown that mechanical stimulation affects the physical properties of multiple types of engineered tissues. However, the optimum regimen for applying cyclic radial stretch to engineered arteries is not well understood. To this end, the effect of mechanical stretch on the development of engineered blood vessels was analyzed in constructs grown from porcine vascular smooth muscle cells. Cyclic radial distension was applied during vessel culture at three rates: 0 beats per minute (bpm), 90 bpm, and 165 bpm. At the end of the 7-week culture period, harvested vessels were analyzed with respect to physical characteristics. Importantly, mechanical stretch at 165 bpm resulted in a significant increase in rupture strength in engineered constructs over nonstretched controls. Stress-strain data and maximal elastic moduli from vessels grown at the three stretch rates indicate enhanced physical properties with increasing pulse rate. In order to investigate the role of collagen cross-linking in the improved mechanical characteristics, collagen cross-link density was quantified by HPLC. Vessels grown with mechanical stretch had somewhat more collagen and higher burst pressures than nonpulsed control vessels. Pulsation did not increase collagen cross-link density. Thus, increased wall thickness and somewhat elevated collagen concentrations, but not collagen cross-link density, appeared to be responsible for increased burst strength.


Asunto(s)
Vasos Sanguíneos/fisiología , Colágeno/metabolismo , Estrés Mecánico , Implantes Absorbibles , Animales , Fenómenos Biomecánicos , Vasos Sanguíneos/metabolismo , Células Cultivadas , Colágeno/efectos de los fármacos , Reactivos de Enlaces Cruzados/farmacología , Ensayo de Materiales , Músculo Liso Vascular/citología , Músculo Liso Vascular/fisiología , Técnicas de Cultivo de Órganos , Resistencia al Corte/fisiología , Porcinos , Porcinos Enanos , Resistencia a la Tracción/fisiología , Ingeniería de Tejidos
14.
Ann Biomed Eng ; 36(11): 1782-92, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18720007

RESUMEN

Mechanical models have potential to guide the development and use of engineered blood vessels as well as other engineered tissues. This paper presents a microstructurally motivated, pseudoelastic, mechanical model of the biaxial mechanics of engineered vessels in the physiologic pressure range. The model incorporates experimentally measured densities and alignments of engineered collagen. Specifically, these microstructural and associated mechanical inputs were measured directly from engineered blood vessels that were cultured over periods of 5-7.5 weeks. To the best of our knowledge, this is the first successful application of either a phenomenological or a microstructurally motivated mechanical model to engineered vascular tissues. Model development revealed the need to use novel theoretical configurations to describe the strain history of engineered vessels. The constitutive equations developed herein suggested that collagen remodeled between 5 and 7.5 weeks during a 7.5-week culture period. This remodeling led to strain energies for collagen that differed with alignment, which likely resulted from undulations that varied with alignment. Finally, biaxial data emphasized that axial extensions increase stresses in engineered vessels in the physiologic pressure range, thereby providing a guideline for surgical use: engineered vessels should be implanted at appropriate axial extension to minimize adverse stress responses.


Asunto(s)
Vasos Sanguíneos , Modelos Cardiovasculares , Ingeniería de Tejidos/métodos , Animales , Anisotropía , Fenómenos Biomecánicos , Células Cultivadas , Humanos , Estrés Mecánico , Porcinos
15.
Ann Biomed Eng ; 35(10): 1749-55, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17566861

RESUMEN

Collagen is the structural molecule that is most correlated with strength in blood vessels. In this study, we compared the properties of collagen in engineered and native blood vessels. Transmission electron microscopy (TEM) was used to image sections of engineered and native arteries. Band periodicities of engineered and native collagen fibrils indicated that spacing between collagen molecules was similar in engineered and native tissues. Engineered arteries, however, had thinner collagen fibrils and fibers than native arteries. Further, collagen fibrils were more loosely packed within collagen fibers in engineered arteries than in native arteries. The sensitivity of TEM analysis allowed measurement of the relative frequency of observation for alignment of collagen. These observations showed that collagen in both engineered and native arteries was aligned circumferentially, helically, and axially, but that engineered arteries had less circumferential collagen and more axial collagen than native arteries. Given that collagen is primarily responsible for dictating the ultimate mechanical properties of arterial tissue, future efforts should focus on using relative frequency of observation for alignment of collagen as a descriptive input for models of the mechanical properties of engineered or native tissues.


Asunto(s)
Arterias/ultraestructura , Bioprótesis , Colágeno/ultraestructura , Ingeniería de Tejidos/métodos , Animales , Anisotropía , Bovinos , Análisis de Falla de Equipo , Conformación Molecular
16.
Ann Biomed Eng ; 35(3): 348-55, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17206488

RESUMEN

With the goal of mimicking the mechanical properties of a given native tissue, tissue engineers seek to culture replacement tissues with compositions similar to those of native tissues. In this report, differences between the mechanical properties of engineered arteries and native arteries were correlated with differences in tissue composition. Engineered arteries failed to match the strengths or compliances of native tissues. Lower strengths of engineered arteries resulted partially from inferior organization of collagen, but not from differences in collagen density. Furthermore, ultimate strengths of engineered vessels were significantly reduced by the presence of residual polyglycolic acid polymer fragments, which caused stress concentrations in the vessel wall. Lower compliances of engineered vessels resulted from minimal smooth muscle cell contractility and a lack of organized extracellular elastin. Organization of elastin and collagen in engineered arteries may have been partially hindered by high concentrations of sulfated glycosaminoglycans. Tissue engineers should continue to regulate cell phenotype and promote synthesis of proteins that are known to dominate the mechanical properties of the associated native tissue. However, we should also be aware of the potential negative impacts of polymer fragments and glycosaminoglycans on the mechanical properties of engineered tissues.


Asunto(s)
Arterias/química , Arterias/fisiología , Ingeniería de Tejidos , Animales , Arterias/anatomía & histología , Ácido Poliglicólico , Porcinos
17.
Cell Transplant ; 12(6): 659-666, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28866947

RESUMEN

More than 570,000 coronary artery bypass grafts are implanted each year, creating an important demand for small-diameter vascular grafts. For patients who lack adequate internal mammary artery or saphenous vein, tissue-engineered arteries may prove useful. However, the time needed to tissue engineer arteries (7 weeks or more) is too long for many patients. Decellularized cadaveric human arteries are another possible source of vascular conduit, but limited availability and the potential for disease transmission limit their widespread use. In contrast, decellularized tissue-engineered arteries could serve as grafts for immediate implantation, as scaffolds onto which patients' cells could be seeded, or as carriers for genetically engineered cells to aid cell transplantation. The goal of this study was to quantify the effects of decellularization on vascular matrix and mechanical properties. Specifically, we compared cellular elimination, extracellular matrix retention, and mechanical characteristics of porcine carotid arteries before and after treatment with three decellularization methods. In addition, for the first time, tissue-engineered arteries were decellularized. Decellularized native arteries were also used as a scaffold onto which vascular cells were seeded. These studies identified a decellularization method for native and engineered arteries that maximized cellular elimination, without greatly compromising mechanical integrity. We showed that engineered tissues could be decellularized, and demonstrated the feasibility of reseeding decellularized vessels with vascular cells.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA