Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cells Tissues Organs ; 201(3): 159-69, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26989895

RESUMO

The use of valved stents for minimally invasive replacement of semilunar heart valves is expected to change the extracellular matrix and mechanical function of the native artery and may thus impair long-term functionality of the implant. Here we investigate the impact of the stent on matrix remodeling of the pulmonary artery in a sheep model, focusing on matrix composition and collagen (re)orientation of the host tissue. Ovine native pulmonary arteries were harvested 8 (n = 2), 16 (n = 4) and 24 (n = 2) weeks after transapical implantation of self-expandable stented heart valves. Second harmonic generation (SHG) microscopy was used to assess the collagen (re)orientation of fresh tissue samples. The collagen and elastin content was quantified using biochemical assays. SHG microscopy revealed regional differences in collagen organization in all explants. In the adventitial layer of the arterial wall far distal to the stent (considered as the control tissue), we observed wavy collagen fibers oriented in the circumferential direction. These circumferential fibers were more straightened in the adventitial layer located behind the stent. On the luminal side of the wall behind the stent, collagen fibers were aligned along the stent struts and randomly oriented between the struts. Immediately distal to the stent, however, fibers on both the luminal and the adventitial side of the wall were oriented in the axial direction, demonstrating the stent impact on the collagen structure of surrounding arterial tissues. Collagen orientation patterns did not change with implantation time, and biochemical analyses showed no changes in the trend of collagen and elastin content with implantation time or location of the vascular wall. We hypothesize that the collagen fibers on the adventitial side of the arterial wall and behind the stent straighten in response to the arterial stretch caused by oversizing of the stent. However, the collagen organization on the luminal side suggests that stent-induced remodeling is dominated by contact guidance.


Assuntos
Bioprótese , Colágeno/análise , Elastina/análise , Próteses Valvulares Cardíacas , Artéria Pulmonar/ultraestrutura , Stents , Animais , Valvas Cardíacas/cirurgia , Artéria Pulmonar/química , Ovinos , Engenharia Tecidual , Alicerces Teciduais/química
2.
J Cell Mol Med ; 18(11): 2176-88, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25103256

RESUMO

Novel cardiovascular replacements are being developed by using degradable synthetic scaffolds, which function as a temporary guide to induce neotissue formation directly in situ. Priming of such scaffolds with fast-releasing monocyte chemoattractant protein-1 (MCP-1) was shown to improve the formation of functional neoarteries in rats. However, the underlying mechanism has not been clarified. Therefore, the goal of this study was to investigate the effect of a burst-release of MCP-1 from a synthetic scaffold on the local recruitment of circulating leucocytes under haemodynamic conditions. Herein, we hypothesized that MCP-1 initiates a desired healing cascade by recruiting favourable monocyte subpopulations into the implanted scaffold. Electrospun poly(ε-caprolactone) scaffolds were loaded with fibrin gel containing various doses of MCP-1 and exposed to a suspension of human peripheral blood mononuclear cells in static or dynamic conditions. In standard migration assay, a dose-dependent migration of specific CD14(+) monocyte subsets was observed, as measured by flow cytometry. In conditions of pulsatile flow, on the other hand, a marked increase in immediate monocyte recruitment was observed, but without evident selectivity in monocyte subsets. This suggests that the selectivity was dependent on the release kinetics of the MCP-1, as it was overruled by the effect of shear stress after the initial burst-release. Furthermore, these findings demonstrate that local recruitment of specific MCP-1-responsive monocytes is not the fundamental principle behind the improved neotissue formation observed in long-term in vivo studies, and mobilization of MCP-1-responsive cells from the bone marrow into the bloodstream is suggested to play a predominant role in vivo.


Assuntos
Quimiocina CCL2/metabolismo , Vasos Coronários/crescimento & desenvolvimento , Leucócitos Mononucleares/citologia , Alicerces Teciduais , Implantes Absorvíveis , Animais , Contagem de Células , Células Cultivadas , Quimiocina CCL2/química , Vasos Coronários/citologia , Vasos Coronários/metabolismo , Humanos , Leucócitos Mononucleares/metabolismo , Masculino , Poliésteres/química , Ratos
3.
Cell Tissue Res ; 352(3): 727-37, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23430473

RESUMO

Mechanical conditioning is often used to enhance collagen synthesis, remodeling and maturation and, hence, the structural and mechanical properties of engineered cardiovascular tissues. Intermittent straining, i.e., alternating periods of cyclic and static strain, has previously been shown to result in more mature tissue compared with continuous cyclic straining. Nevertheless, the underlying mechanism is unknown. We have determined the short-term effects of continuous cyclic strain and of cyclic strain followed by static strain at the gene expression level to improve insight into the mechano-regulatory mechanism of intermittent conditioning on collagen synthesis, remodeling and maturation. Tissue-engineered constructs, consisting of human vascular-derived cells seeded onto rapidly degrading PGA/P4HB scaffolds, were conditioned with 4% strain at 1 Hz for 3 h in order to study the immediate effects of cyclic strain (n=18). Next, the constructs were either subjected to ongoing cyclic strain (4% at 1 Hz; n=9) or to static strain (n=9). Expression levels of genes involved in collagen synthesis, remodeling and maturation were studied at various time points up to 24 h within each straining protocol. The results indicate that a period of static strain following cyclic strain favors collagen synthesis and remodeling, whereas ongoing cyclic strain shifts this balance toward collagen remodeling and maturation. The data suggest that, with prolonged culture, the conditioning protocol should be changed from intermittent straining to continuous cyclic straining to improve collagen maturation after its synthesis and, hence, the tissue (mechanical) properties.


Assuntos
Sistema Cardiovascular/metabolismo , Colágeno/metabolismo , Matriz Extracelular/metabolismo , Estresse Mecânico , Engenharia Tecidual/métodos , Transcriptoma , Regulação da Expressão Gênica , Humanos , Reprodutibilidade dos Testes , Alicerces Teciduais
4.
J Heart Valve Dis ; 21(5): 670-8, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23167234

RESUMO

BACKGROUND AND AIM OF THE STUDY: Living tissue-engineered heart valves (TEHVs) based on rapidly degrading scaffolds and autologous cells might overcome the limitations of today's valve substitutes. Following minimally invasive trans-apical implantation into an ovine model, TEHVs showed adequate in-vivo functionality, but a thickening of the leaflets was observed. In order to evaluate the impact of the substantial tissue deformations of TEHVs associated with the crimping procedure during minimally invasive delivery, trans-apical and conventional implantation technologies were compared in an ovine model. METHODS: Trileaflet heart valves (n=11) based on PGA/P4HB-scaffolds, integrated into self-expandable stents, were engineered from autologous ovine vascular-derived cells. After in-vitro culture, the TEHVs were either implanted surgically (n=5), replacing the native pulmonary valve, or delivered trans-apically (n=6) into the orthotopic pulmonary valve position. In-vivo functionality was assessed by echocardiography and by angiography for up to eight weeks. The tissue compositions of the explanted TEHVs and corresponding control valves were analyzed. RESULTS: TEHV implantations were successful in all cases. Independent of the implantation method, the explants demonstrated a comparable layered tissue formation with thickening and deposited fibrous layers. Active remodeling of these layers was evident in the explants, as indicated by vascularization of the walls, invasion of the host cells, and the formation of a luminal endothelial layer on the TEHV leaflets. CONCLUSION: This direct comparison of trans-apical and conventional surgical implantation techniques showed that crimping had no adverse effect on the integrity or functional outcome of TEHVs. This suggests that a thickening of TEHVs in vivo is neither caused by nor enhanced by the crimping procedure, but represents a functional tissue remodeling process.


Assuntos
Bioprótese , Implante de Prótese de Valva Cardíaca , Próteses Valvulares Cardíacas , Valva Pulmonar , Animais , Procedimentos Cirúrgicos Minimamente Invasivos , Valva Pulmonar/patologia , Ovinos , Engenharia Tecidual
5.
Circulation ; 119(2): 290-7, 2009 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-19118259

RESUMO

BACKGROUND: Previous attempts in heart valve tissue engineering (TE) failed to produce autologous valve replacements with native-like mechanical behavior to allow for systemic pressure applications. Because hypoxia and insulin are known to promote protein synthesis by adaptive cellular responses, a physiologically relevant oxygen tension and insulin supplements were applied to the growing heart valve tissues to enhance their mechanical properties. METHODS AND RESULTS: Scaffolds of rapid-degrading polyglycolic acid meshes coated with poly-4-hydroxybutyrate were seeded with human saphenous vein myofibroblasts. The tissue-engineered constructs were cultured under normal oxygen tension (normoxia) or hypoxia (7% O(2)) and incubated with or without insulin. Glycosaminoglycan production in the constructs approached that of native values under the influence of hypoxia and under the influence of insulin. Both insulin and hypoxia were associated with enhanced matrix production and improved mechanical properties; however, a synergistic effect was not observed. Although the amount of collagen and cross-links in the engineered tissues was still lower than that in native adult human aortic valves, constructs cultured under hypoxic conditions reached native human aortic valve levels of tissue strength and stiffness after 4 weeks of culturing. CONCLUSIONS: These results indicate that oxygen tension may be a key parameter for the achievement of sufficient tissue quality and mechanical integrity in tissue-engineered heart valves. Engineered tissues of such strength, based on rapid-degrading polymers, have not been achieved to date. These findings bring the potential use of tissue-engineered heart valves for systemic applications a step closer and represent an important improvement in heart valve tissue engineering.


Assuntos
Valva Aórtica/citologia , Valva Aórtica/fisiologia , Técnicas de Cultura de Células/métodos , Próteses Valvulares Cardíacas , Engenharia Tecidual/métodos , Valva Aórtica/efeitos dos fármacos , Fenômenos Biomecânicos/fisiologia , Hipóxia Celular/fisiologia , Células Cultivadas , Próteses Valvulares Cardíacas/normas , Humanos , Insulina/farmacologia , Fatores de Tempo , Engenharia Tecidual/instrumentação
6.
Circulation ; 116(11 Suppl): I64-70, 2007 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-17846327

RESUMO

BACKGROUND: A novel concept providing prenatally tissue engineered human autologous heart valves based on routinely obtained fetal amniotic fluid progenitors as single cell source is introduced. METHODS AND RESULTS: Fetal human amniotic progenitors were isolated from routinely sampled amniotic fluid and sorted using CD133 magnetic beads. After expansion and differentiation, cell phenotypes of CD133- and CD133+ cells were analyzed by immunohistochemistry and flowcytometry. After characterization, CD133- derived cells were seeded onto heart valve leaflet scaffolds (n=18) fabricated from rapidly biodegradable polymers, conditioned in a pulse duplicator system, and subsequently coated with CD133+ derived cells. After in vitro maturation, opening and closing behavior of leaflets was investigated. Neo-tissues were analyzed by histology, immunohistochemistry, and scanning electron microscopy (SEM). Extracellular matrix (ECM) elements and cell numbers were quantified biochemically. Mechanical properties were assessed by tensile testing. CD133- derived cells demonstrated characteristics of mesenchymal progenitors expressing CD44 and CD105. Differentiated CD133+ cells showed features of functional endothelial cells by eNOS and CD141 expression. Engineered heart valve leaflets demonstrated endothelialized tissue formation with production of ECM elements (GAG 80%, HYP 5%, cell number 100% of native values). SEM showed intact endothelial surfaces. Opening and closing behavior was sufficient under half of systemic conditions. CONCLUSIONS: The use of amniotic fluid as single cell source is a promising low-risk approach enabling the prenatal fabrication of heart valves ready to use at birth. These living replacements with the potential of growth, remodeling, and regeneration may realize the early repair of congenital malformations.


Assuntos
Líquido Amniótico/citologia , Bioprótese , Próteses Valvulares Cardíacas , Valvas Cardíacas/citologia , Células-Tronco/citologia , Adulto , Líquido Amniótico/fisiologia , Células Cultivadas , Feminino , Valvas Cardíacas/fisiologia , Humanos , Masculino , Gravidez , Células-Tronco/fisiologia , Engenharia Tecidual/métodos
7.
Thromb Haemost ; 100(2): 271-9, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18690347

RESUMO

Monocytes and platelets are both crucially involved in atherogenesis. Importantly, activated platelets bound to circulating monocytes increase adhesion of the monocytes and thus mediate colocalization of both cell types at the vessel wall. We examined the fate of the platelets upon migration of these potentially pro-atherogenic platelet-monocyte complexes (PMC) across activated endothelium. Platelet-monocyte complex migration was studied both quantitatively by means of Transwell filters coated with endothelial cells, as well as qualitatively with different imaging techniques, and in the absence or presence of flow. Upon PMC transendothelial migration, platelets relocate with monocytic P-selectin glycoprotein ligand-1 (PSGL-1) to the rear of the monocyte, detach, and remain at the endothelial surface. Platelet dissociation appeared not to be due to reduced PSGL-1 expression or reduced platelet-binding capacity of the migrated monocytes. In addition, different endothelial matrix proteins with different platelet-binding capacities coated on the Transwell filter, instead of endothelial cells, did not affect PMC dissociation. In contrast, lowering the mechanical stress that PMC experience during transmigration prevented dissociation of platelets. In conclusion, PMC dissociate during transendothelial migration as a result of monocytic PSGL-1 redistribution and mechanical stress. PMC-mediated deposition of activated platelets at sites of vascular inflammation is likely relevant for cardiovascular disease progression or vascular regeneration.


Assuntos
Aterosclerose/patologia , Plaquetas/citologia , Comunicação Celular/fisiologia , Movimento Celular/fisiologia , Células Endoteliais/citologia , Monócitos/citologia , Aterosclerose/imunologia , Plaquetas/fisiologia , Adesão Celular/fisiologia , Polaridade Celular/fisiologia , Células Endoteliais/imunologia , Humanos , Glicoproteínas de Membrana/metabolismo , Monócitos/imunologia , Monócitos/metabolismo , Estresse Mecânico , Veias Umbilicais/citologia
8.
J Biomech ; 41(12): 2612-7, 2008 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-18701107

RESUMO

Understanding the response of tissue structures to mechanical stress is crucial for optimization of mechanical conditioning protocols in the field of heart valve tissue engineering. In heart valve tissue, it is unclear to what extent mechanical loading affects the collagen fibril morphology. To determine if local stress affects the collagen fibril morphology, in terms of fibril diameter, its distribution, and the fibril density, this was investigated in adult native human aortic valve leaflets. Transmission electron microscopy images of collagen fibrils were analyzed at three locations: the commissures, the belly, and the fixed edge of the leaflets. Subsequently, the mechanical behavior of human aortic valves was used in a computational model to predict the stress distribution in the valve leaflet during the diastolic phase of the cardiac cycle. The local stresses at the three locations were related to the collagen fibril morphology. The fibril diameter and density varied significantly between the measured locations, and appeared inversely related. The average fibril diameter increased from the fixed edge, to the belly, and to the commissures of the leaflets, while fibril density decreased. Interestingly, these differences corresponded well with the level of stress at the locations. The presented data showed that large tissue stress is associated with greater average fibril diameter, lower fibril density, and wider fibril size distribution compared with low stress locations in the leaflets. The findings here provide insight in the effect of mechanical loading on the collagen ultrastructure, and are valuable to improve conditioning protocols for tissue engineering.


Assuntos
Valva Aórtica/fisiologia , Valva Aórtica/ultraestrutura , Colágenos Fibrilares/fisiologia , Colágenos Fibrilares/ultraestrutura , Próteses Valvulares Cardíacas , Mecanotransdução Celular/fisiologia , Modelos Cardiovasculares , Bioprótese , Simulação por Computador , Módulo de Elasticidade , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Conformação Proteica , Estresse Mecânico , Engenharia Tecidual
9.
Front Cardiovasc Med ; 5: 105, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30159315

RESUMO

The creation of living heart valve replacements via tissue engineering is actively being pursued by many research groups. Numerous strategies have been described, aimed either at culturing autologous living valves in a bioreactor (in vitro) or inducing endogenous regeneration by the host via resorbable scaffolds (in situ). Whereas a lot of effort is being invested in the optimization of heart valve scaffold parameters and culturing conditions, the pathophysiological in vivo remodeling processes to which tissue-engineered heart valves are subjected upon implantation have been largely under-investigated. This is partly due to the unavailability of suitable immunohistochemical tools specific to sheep, which serves as the gold standard animal model in translational research on heart valve replacements. Therefore, the goal of this study was to comprise and validate a comprehensive sheep-specific panel of antibodies for the immunohistochemical analysis of tissue-engineered heart valve explants. For the selection of our panel we took inspiration from previous histopathological studies describing the morphology, extracellular matrix composition and cellular composition of native human heart valves throughout development and adult stages. Moreover, we included a range of immunological markers, which are particularly relevant to assess the host inflammatory response evoked by the implanted heart valve. The markers specifically identifying extracellular matrix components and cell phenotypes were tested on formalin-fixed paraffin-embedded sections of native sheep aortic valves. Markers for inflammation and apoptosis were tested on ovine spleen and kidney tissues. Taken together, this panel of antibodies could serve as a tool to study the spatiotemporal expression of proteins in remodeling tissue-engineered heart valves after implantation in a sheep model, thereby contributing to our understanding of the in vivo processes which ultimately determine long-term success or failure of tissue-engineered heart valves.

10.
Circulation ; 114(1 Suppl): I125-31, 2006 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-16820561

RESUMO

BACKGROUND: Heart valve tissue engineering is a promising strategy to overcome the lack of autologous growing replacements, particularly for the repair of congenital malformations. Here, we present a novel concept using human prenatal progenitor cells as new and exclusive cell source to generate autologous implants ready for use at birth. METHODS AND RESULTS: Human fetal mesenchymal progenitors were isolated from routinely sampled prenatal chorionic villus specimens and expanded in vitro. A portion was cryopreserved. After phenotyping and genotyping, cells were seeded onto synthetic biodegradable leaflet scaffolds (n=12) and conditioned in a bioreactor. After 21 days, leaflets were endothelialized with umbilical cord blood-derived endothelial progenitor cells and conditioned for additional 7 days. Resulting tissues were analyzed by histology, immunohistochemistry, biochemistry (amounts of extracellular matrix, DNA), mechanical testing, and scanning electron microscopy (SEM) and were compared with native neonatal heart valve leaflets. Fresh and cryopreserved cells showed comparable myofibroblast-like phenotypes. Genotyping confirmed their fetal origin. Neo-tissues exhibited organization, cell phenotypes, extracellular matrix production, and DNA content comparable to their native counterparts. Leaflet surfaces were covered with functional endothelia. SEM showed cellular distribution throughout the polymer and smooth surfaces. Mechanical profiles approximated those of native heart valves. CONCLUSIONS: Prenatal fetal progenitors obtained from routine chorionic villus sampling were successfully used as an exclusive, new cell source for the engineering of living heart valve leaflets. This concept may enable autologous replacements with growth potential ready for use at birth. Combined with the use of cell banking technology, this approach may be applied also for postnatal applications.


Assuntos
Bioprótese , Amostra da Vilosidade Coriônica , Vilosidades Coriônicas , Próteses Valvulares Cardíacas , Células-Tronco Mesenquimais/citologia , Engenharia Tecidual/métodos , Coleta de Tecidos e Órgãos , Implantes Absorvíveis , Biodegradação Ambiental , Reatores Biológicos , Separação Celular , Técnicas de Cocultura , Criopreservação , DNA/metabolismo , Células Endoteliais/citologia , Matriz Extracelular/metabolismo , Sangue Fetal/citologia , Citometria de Fluxo , Genótipo , Idade Gestacional , Humanos , Doadores Vivos , Masculino , Teste de Materiais , Células-Tronco Mesenquimais/metabolismo , Microscopia Eletrônica de Varredura , Técnicas de Cultura de Órgãos/métodos , Fenótipo , Resistência à Tração , Engenharia Tecidual/instrumentação , Preservação de Tecido , Transplante Autólogo
11.
Circulation ; 114(1 Suppl): I159-66, 2006 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-16820566

RESUMO

BACKGROUND: Living autologous vascular grafts with the capacity for regeneration and growth may overcome the limitations of contemporary artificial prostheses. Particularly in congenital cardiovascular surgery, there is an unmet medical need for growing replacement materials. Here we investigate growth capacity of tissue-engineered living pulmonary arteries in a growing lamb model. METHODS AND RESULTS: Vascular grafts fabricated from biodegradable scaffolds (ID 18+/-l mm) were sequentially seeded with vascular cells. The seeded constructs were grown in vitro for 21 days using biomimetic conditions. Thereafter, these tissue-engineered vascular grafts (TEVGs) were surgically implanted as main pulmonary artery replacements in 14 lambs using cardiopulmonary bypass and followed up for < or = 100 weeks. The animals more than doubled their body weight during the 2-year period. The TEVG showed good functional performance demonstrated by regular echocardiography at 20, 50, 80, and 100 weeks and computed tomography-angiography. In particular, there was no evidence of thrombus, calcification, stenosis, suture dehiscence, or aneurysm. There was a significant increase in diameter by 30% and length by 45%. Histology showed tissue formation reminiscent of native artery. Biochemical analysis revealed cellularity and proteoglycans and increased collagen contents in all of the groups, analogous to those of native vessels. The mechanical profiles of the TEVG showed stronger but less elastic tissue properties than native pulmonary arteries. CONCLUSIONS: This study provides evidence of growth in living, functional pulmonary arteries engineered from vascular cells in a full growth animal model.


Assuntos
Prótese Vascular , Implantes Experimentais , Artéria Pulmonar/cirurgia , Engenharia Tecidual , Implantes Absorvíveis , Animais , Biodegradação Ambiental , Biomarcadores , Fenômenos Biomecânicos , Implante de Prótese Vascular , Colágeno/biossíntese , Fibroblastos/citologia , Mioblastos/citologia , Complicações Pós-Operatórias , Proteoglicanas/biossíntese , Artéria Pulmonar/diagnóstico por imagem , Ovinos , Resistência à Tração , Engenharia Tecidual/instrumentação , Engenharia Tecidual/métodos , Tomografia Computadorizada por Raios X , Transplante Autólogo , Ultrassonografia , Aumento de Peso
12.
Circulation ; 114(1 Suppl): I152-8, 2006 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-16820565

RESUMO

BACKGROUND: Tissue engineering represents a promising approach for the development of living heart valve replacements. In vivo animal studies of tissue-engineered autologous heart valves have focused on pulmonary valve replacements, leaving the challenge to tissue engineer heart valves suitable for systemic application using human cells. METHODS AND RESULTS: Tissue-engineered human heart valves were analyzed up to 4 weeks and conditioning using bioreactors was compared with static culturing. Tissue formation and mechanical properties increased with time and when using conditioning. Organization of the tissue, in terms of anisotropic properties, increased when conditioning was dynamic in nature. Exposure of the valves to physiological aortic valve flow demonstrated proper opening motion. Closure dynamics were suboptimal, most likely caused by the lower degree of anisotropy when compared with native aortic valve leaflets. CONCLUSIONS: This study presents autologous tissue-engineered heart valves based on human saphenous vein cells and a rapid degrading synthetic scaffold. Tissue properties and mechanical behavior might allow for use as living aortic valve replacements.


Assuntos
Valva Aórtica , Bioprótese , Próteses Valvulares Cardíacas , Engenharia Tecidual , Implantes Absorvíveis , Idoso , Anisotropia , Biodegradação Ambiental , Reatores Biológicos , Células Cultivadas/citologia , Células Cultivadas/metabolismo , Colágeno/biossíntese , Fibroblastos/citologia , Fibroblastos/metabolismo , Glicosaminoglicanos/biossíntese , Humanos , Masculino , Teste de Materiais , Mioblastos/citologia , Mioblastos/metabolismo , Reologia , Veia Safena/citologia , Stents , Resistência à Tração , Engenharia Tecidual/instrumentação , Engenharia Tecidual/métodos
13.
Tissue Eng ; 13(7): 1501-11, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17518750

RESUMO

A major challenge in tissue engineering of functional heart valves is to determine and mimic the dominant tissue structures that regulate heart valve function and in vivo survival. In native heart valves, the anisotropic matrix architecture assures sustained and adequate functioning under high-pressure conditions. Collagen, being the main load-bearing matrix component, contributes significantly to the biomechanical strength of the tissue. This study investigates the relationship between collagen content, collagen cross-links, and biomechanical behavior in human aortic heart valve leaflets and in tissue-engineered constructs. In the main loading direction (circumferential) of native valve leaflets, a significant positive linear correlation between modulus of elasticity and collagen cross-link concentration was found, whereas no correlation between modulus of elasticity and collagen content was found. Similar findings were observed in tissue-engineered constructs, where cross-link concentration was higher for dynamically strained constructs then for statically cultured controls. These findings suggest a dominant role for collagen cross-links over collagen content with respect to biomechanical tissue behavior in human heart valve leaflets. They further suggest that dynamic tissue straining in tissue engineering protocols can enhance cross-link concentration and biomechanical function.


Assuntos
Valva Aórtica/anatomia & histologia , Valva Aórtica/fisiologia , Fenômenos Biomecânicos , Colágeno/fisiologia , Engenharia Tecidual , Feminino , Humanos , Masculino , Pessoa de Meia-Idade
14.
J Biomech ; 40(2): 325-34, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-16529755

RESUMO

Mathematical models can provide valuable information to assess and evaluate the mechanical behavior of tissue-engineered constructs. In this study, a structurally based model is applied to describe and analyze the mechanics of tissue-engineered human heart valve leaflets. The results from two orthogonal uniaxial tensile tests are used to determine the model parameters of the constructs after two, three and four weeks of culturing. Subsequently, finite element analyses are performed to simulate the mechanical response of the engineered leaflets to a pressure load. The stresses in the leaflets induced by the pressure load increase monotonically with culture time due to a decrease in the construct's thickness. The strains, on the other hand, eventually decrease as a result of an increase in the elastic modulus. Compared to native porcine leaflets, the mechanical response of the engineered tissues after four weeks of culturing is more linear, stiffer and less anisotropic.


Assuntos
Fenômenos Biomecânicos , Próteses Valvulares Cardíacas , Modelos Biológicos , Engenharia Tecidual , Análise de Elementos Finitos , Humanos
15.
J Leukoc Biol ; 79(3): 499-507, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16415171

RESUMO

Human monocytes adhere to activated platelets, resulting in the formation of platelet-monocyte complexes (PMC). Complex formation depends on the interaction between platelet-displayed P-selectin and the specific ligand for P-selectin on leukocytes, P-selectin glycoprotein ligand-1 (PSGL-1). We have recently shown that monocytes within PMC have increased adhesive capacity to the activated endothelium. To better understand the effect of platelet binding on the capacity of monocytes to adhere to activated endothelium, the P-selectin-PSGL-1 interaction-induced changes in integrin functionality were studied. The binding of platelets to monocytes via P-selectin-PSGL-1 interactions was shown to increase expression and activity of alpha4beta1 and alphaMbeta2integrin, with a concomitant decrease in L-selectin expression. Furthermore, the binding of platelets to monocytes resulted in increased monocyte adhesion to intercellular adhesion molecule-1, vascular cell adhesion molecule-1, and fibronectin. Platelet binding was also responsible for an increase in monocyte transendothelial migration. Similar effects were observed after engagement of PSGL-1 with specific antibodies or with P-selectin immunoglobulin protein. Our data suggest that platelets, by binding via P-selectin to PSGL-1 on monocytes, induce up-regulation and activation of beta1 and beta2integrins and increased adhesion of monocytes to activated endothelium. Hence, monocytes within PMC are in a higher state of activation and may have, therefore, an increased atherogenic capacity.


Assuntos
Plaquetas/metabolismo , Antígenos CD18/metabolismo , Membrana Celular/metabolismo , Integrina beta1/metabolismo , Monócitos/metabolismo , Regulação para Cima/imunologia , Anticorpos/farmacologia , Plaquetas/imunologia , Antígenos CD18/imunologia , Adesão Celular/imunologia , Membrana Celular/imunologia , Movimento Celular/imunologia , Células Cultivadas , Regulação para Baixo/imunologia , Células Endoteliais/imunologia , Fibronectinas/imunologia , Fibronectinas/metabolismo , Humanos , Integrina beta1/imunologia , Molécula 1 de Adesão Intercelular/imunologia , Molécula 1 de Adesão Intercelular/metabolismo , Selectina L/imunologia , Selectina L/metabolismo , Glicoproteínas de Membrana/antagonistas & inibidores , Glicoproteínas de Membrana/imunologia , Glicoproteínas de Membrana/metabolismo , Monócitos/imunologia , Selectina-P/imunologia , Selectina-P/metabolismo , Molécula 1 de Adesão de Célula Vascular/imunologia , Molécula 1 de Adesão de Célula Vascular/metabolismo
16.
Methods Mol Med ; 140: 319-30, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-18085217

RESUMO

Heart valve replacement represents the most common surgical therapy for end-stage valvular heart diseases. A major drawback all contemporary heart valve replacements have in common is the lack of growth, repair, and remodeling capabilities. To overcome these limitations, the emerging field of tissue engineering is focusing on the in vitro generation of functional, living heart valve replacements. The basic approach uses starter matrices of either decellularized xenogeneic or biopolymeric materials configured in the shape of the heart valve and subsequent cell seeding. Moreover, in vitro strategies using mechanical loading in bioreactor systems have been developed to improve tissue maturation. This chapter gives a short overview of the current concepts and provides detailed methods for in vitro heart valve tissue engineering.


Assuntos
Bioprótese , Próteses Valvulares Cardíacas , Valvas Cardíacas/anatomia & histologia , Engenharia Tecidual/métodos , Animais , Reatores Biológicos , Técnicas de Cultura de Células , Células Cultivadas , Humanos
17.
Tissue Eng Part A ; 23(19-20): 1142-1151, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28314377

RESUMO

Decellularized tissue-engineered heart valves (DTEHVs) showed remarkable results in translational animal models, leading to recellularization within hours after implantation. This is crucial to enable tissue remodeling. To investigate if the presence of scaffold remnants before implantation is responsible for the fast recellularization of DTEHVs, an in vitro mesofluidic system was used. Human granulocyte and agranulocyte fractions were isolated, stained, brought back in suspension, and implemented in the system. Three different types of biomaterials were exposed to the circulating blood cells, consisting of decellularized tissue-engineered constructs (DTECs) with or without scaffold remnants or only bare scaffold. After 5 h of testing, the granulocyte fraction depleted faster from the circulation than the agranulocyte fraction. However, only granulocytes infiltrated into the DTEC with scaffold, migrating toward the scaffold remnants. The agranulocyte population, on the other hand, was only observed on the outer surface. Active cell infiltration was associated with increased levels of matrix metalloproteinase-1 secretion in the DTEC, including scaffold remnants. Proinflammatory cytokines such as interleukin (IL)-1α, IL-6, and tumor necrosis factor alpha (TNFα) were significantly upregulated in the DTEC without scaffold remnants. These results indicate that scaffold remnants can influence the immune response in DTEC, being responsible for rapid cell infiltration.


Assuntos
Células Sanguíneas/metabolismo , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Adulto , Citocinas/metabolismo , Citometria de Fluxo , Coração/fisiologia , Humanos , Masculino
18.
3D Print Addit Manuf ; 4(1): 19-29, 2017 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-32953940

RESUMO

The evolution of minimally invasive implantation procedures and the in vivo remodeling potential of decellularized tissue-engineered heart valves require stents with growth capacity to make these techniques available for pediatric patients. By means of computational tools and 3D printing technology, this proof-of-concept study demonstrates the design and manufacture of a polymer stent with a mechanical performance comparable to that of conventional nitinol stents used for heart valve implantation in animal trials. A commercially available 3D printing polymer was selected, and crush and crimping tests were conducted to validate the results predicted by the computational model. Finally, the degradability of the polymer was assessed via accelerated hydrolysis.

19.
Tissue Eng ; 12(11): 3223-32, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17518636

RESUMO

This study demonstrates the engineering of biologically active heart valve leaflets using prenatally available human umbilical cord-derived progenitor cells as the only cell source. Wharton's Jelly-derived cells and umbilical cord blood-derived endothelial progenitor cells were subsequently seeded on biodegradable scaffolds and cultured in a biomimetic system under biochemical or mechanical stimulation or both. Depending on the stimulation, leaflets showed mature layered tissue formation with functional endothelia and extracellular matrix production comparable with that of native tissues. This demonstrates the feasibility of heart valve leaflet fabrication from prenatal umbilical cord-derived progenitor cells as a further step in overcoming the lack of living autologous replacements with growth and regeneration potential for the repair of congenital malformation.


Assuntos
Bioprótese , Sangue Fetal/citologia , Próteses Valvulares Cardíacas , Células-Tronco/citologia , Engenharia Tecidual/métodos , Cordão Umbilical/citologia , Materiais Biocompatíveis , Materiais Biomiméticos , Células Cultivadas , Matriz Extracelular/química , Estudos de Viabilidade , Humanos , Imuno-Histoquímica , Microscopia Eletrônica de Varredura , Técnicas de Cultura de Órgãos , Poliésteres/química , Ácido Poliglicólico/química , Estresse Mecânico , Resistência à Tração , Engenharia Tecidual/instrumentação , Cordão Umbilical/irrigação sanguínea , Veias Umbilicais/citologia
20.
Tissue Eng Part A ; 22(1-2): 123-32, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26466917

RESUMO

Synthetic polymers are widely used to fabricate porous scaffolds for the regeneration of cardiovascular tissues. To ensure mechanical integrity, a balance between the rate of scaffold absorption and tissue formation is of high importance. A higher rate of tissue formation is expected in fast-degrading materials than in slow-degrading materials. This could be a result of synthetic cells, which aim to compensate for the fast loss of mechanical integrity of the scaffold by deposition of collagen fibers. Here, we studied the effect of fast-degrading polyglycolic acid scaffolds coated with poly-4-hydroxybutyrate (PGA-P4HB) and slow-degrading poly-ɛ-caprolactone (PCL) scaffolds on amount of tissue, composition, and mechanical characteristics in time, and compared these engineered values with values for native human heart valves. Electrospun PGA-P4HB and PCL scaffolds were either kept unseeded in culture or were seeded with human vascular-derived cells. Tissue formation, extracellular matrix (ECM) composition, remaining scaffold weight, tissue-to-scaffold weight ratio, and mechanical properties were analyzed every week up to 6 weeks. Mass of unseeded PCL scaffolds remained stable during culture, whereas PGA-P4HB scaffolds degraded rapidly. When seeded with cells, both scaffold types demonstrated increasing amounts of tissue with time, which was more pronounced for PGA-P4HB-based tissues during the first 2 weeks; however, PCL-based tissues resulted in the highest amount of tissue after 6 weeks. This study is the first to provide insight into the tissue-to-scaffold weight ratio, therewith allowing for a fair comparison between engineered tissues cultured on scaffolds as well as between native heart valve tissues. Although the absolute amount of ECM components differed between the engineered tissues, the ratio between ECM components was similar after 6 weeks. PCL-based tissues maintained their shape, whereas the PGA-P4HB-based tissues deformed during culture. After 6 weeks, PCL-based engineered tissues showed amounts of cells and ECM that were comparable to the number of human native heart valve leaflets, whereas values were lower in the PGA-P4HB-based tissues. Although increasing in time, the number of collagen crosslinks were below native values in all engineered tissues. In conclusion, this study indicates that slow-degrading scaffold materials are favored over fast-degrading materials to create organized ECM-rich tissues in vitro, which keep their three-dimensional structure before implantation.


Assuntos
Valvas Cardíacas , Poliésteres/química , Ácido Poliglicólico/química , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Células Cultivadas , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA