Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros










Base de dados
Tipo de estudo
Intervalo de ano de publicação
1.
J Tissue Eng Regen Med ; 10(10): E443-E453, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-24130082

RESUMO

Valvular interstitial cells (VICs) respond to 3D matrix interactions in a complex manner, but understanding these effects on VIC function better is important for applications ranging from valve tissue engineering to studying valve disease. Here, we encapsulated VICs in poly(ethylene glycol) (PEG) hydrogels modified with three different adhesive ligands, derived from fibronectin (RGDS), elastin (VGVAPG) and collagen-1 (P15). By day 14, VICs became significantly more elongated in RGDS-containing gels compared to VGVAPG or P15. This difference in cell morphology appeared to correlate with global matrix metalloproteinase (MMP) activity, as VICs encapsulated in RGDS-functionalized hydrogels secreted higher levels of active MMP at day 2. VIC activation to a myofibroblast phenotype was also characterized by staining for α-smooth muscle actin (αSMA) at day 14. The percentage of αSMA+ VICs in the VGVAPG gels was the highest (56%) compared to RGDS (33%) or P15 (38%) gels. Matrix deposition and composition were also characterized at days 14 and 42 and found to depend on the initial hydrogel composition. All gel formulations had similar levels of collagen, elastin and chondroitin sulphate deposited as the porcine aortic valve. However, the composition of collagen deposited by VICs in VGVAPG-functionalized gels had a significantly higher collagen-X:collagen-1 ratio, which is associated with stenotic valves. Taken together, these data suggest that peptide-functionalized PEG hydrogels are a useful system for culturing VICs three-dimensionally and, with the ability to systematically alter biochemical and biophysical properties, this platform may prove useful in manipulating VIC function for valve regeneration. Copyright © 2013 John Wiley & Sons, Ltd.


Assuntos
Colágeno/química , Matriz Extracelular/química , Valvas Cardíacas/metabolismo , Hidrogéis/química , Oligopeptídeos/química , Fragmentos de Peptídeos/química , Polietilenoglicóis/química , Animais , Células Cultivadas , Valvas Cardíacas/citologia , Suínos
2.
Am J Physiol Heart Circ Physiol ; 306(9): H1302-13, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24610917

RESUMO

Risk factors for fibrocalcific aortic valve disease (FCAVD) are associated with systemic decreases in bioavailability of endothelium-derived nitric oxide (EDNO). In patients with bicuspid aortic valve (BAV), vascular expression of endothelial nitric oxide synthase (eNOS) is decreased, and eNOS(-/-) mice have increased prevalence of BAV. The goal of this study was to test the hypotheses that EDNO attenuates profibrotic actions of valve interstitial cells (VICs) in vitro and that EDNO deficiency accelerates development of FCAVD in vivo. As a result of the study, coculture of VICs with aortic valve endothelial cells (vlvECs) significantly decreased VIC activation, a critical early phase of FCAVD. Inhibition of VIC activation by vlvECs was attenuated by N(G)-nitro-l-arginine methyl ester or indomethacin. Coculture with vlvECs attenuated VIC expression of matrix metalloproteinase-9, which depended on stiffness of the culture matrix. Coculture with vlvECs preferentially inhibited collagen-3, compared with collagen-1, gene expression. BAV occurred in 30% of eNOS(-/-) mice. At age 6 mo, collagen was increased in both bicuspid and trileaflet eNOS(-/-) aortic valves, compared with wild-type valves. At 18 mo, total collagen was similar in eNOS(-/-) and wild-type mice, but collagen-3 was preferentially increased in eNOS(-/-) mice. Calcification and apoptosis were significantly increased in BAV of eNOS(-/-) mice at ages 6 and 18 mo. Remarkably, these histological changes were not accompanied by physiologically significant valve stenosis or regurgitation. In conclusion, coculture with vlvECs inhibits specific profibrotic VIC processes. In vivo, eNOS deficiency produces fibrosis in both trileaflet and BAVs but produces calcification only in BAVs.


Assuntos
Valva Aórtica/patologia , Calcinose/metabolismo , Cardiopatias Congênitas/metabolismo , Doenças das Valvas Cardíacas/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Animais , Valva Aórtica/metabolismo , Valva Aórtica/fisiopatologia , Apoptose , Doença da Válvula Aórtica Bicúspide , Calcinose/patologia , Calcinose/fisiopatologia , Células Cultivadas , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Colágeno Tipo III/genética , Colágeno Tipo III/metabolismo , Cardiopatias Congênitas/patologia , Cardiopatias Congênitas/fisiopatologia , Doenças das Valvas Cardíacas/patologia , Doenças das Valvas Cardíacas/fisiopatologia , Células Intersticiais de Cajal/metabolismo , Células Intersticiais de Cajal/patologia , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Óxido Nítrico Sintase Tipo III/deficiência , Óxido Nítrico Sintase Tipo III/genética , Esclerose/metabolismo , Esclerose/patologia , Esclerose/fisiopatologia , Suínos
3.
Adv Healthc Mater ; 3(5): 649-57, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24459068

RESUMO

Biophysical cues are widely recognized to influence cell phenotype. While this evidence was established using static substrates, there is growing interest in creating stimulus-responsive biomaterials that better recapitulate the dynamic extracellular matrix. Here, a clickable, photodegradable hydrogel substrate that allows the user to precisely control substrate elasticity and topography in situ is presented. The hydrogels are synthesized by reacting an 8-arm poly(ethylene glycol) alkyne with an azide-functionalized photodegradable crosslinker. The utility of this platform by exploiting its photoresponsive properties to modulate the phenotype of porcine aortic valvular interstitial cells (VICs) is demonstrated. First, VIC phenotype is monitored, in response to initial substratum modulus and static topographic cues. Higher modulus (E ≈ 15 kPa) substrates induce higher levels of activation (≈70% myofibroblasts) versus soft (E ≈ 3 kPa) substrates (≈20% myofibroblasts). Microtopographies that induce VIC alignment and elongation on low modulus substrates also stimulate activation. Finally, VIC phenotype is monitored in response to sequential in situ manipulations. The results illustrate that VIC activation on stiff surfaces (≈70% myofibroblasts) can be partially reversed by reducing surface modulus (≈30% myofibroblats) and subsequently re-activated by anisotropic topographies (≈60% myofibroblasts). Such dynamic substrates afford unique opportunities to decipher the complex role of matrix cues on the plasticity of VIC activation.


Assuntos
Valva Aórtica/citologia , Técnicas de Cultura de Células/instrumentação , Técnicas de Cultura de Células/métodos , Hidrogéis/química , Animais , Células Cultivadas , Módulo de Elasticidade , Fenótipo , Fotólise , Suínos , Alicerces Teciduais
4.
Biomaterials ; 35(11): 3596-606, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24462357

RESUMO

The effects of valvular endothelial cell (VlvEC) paracrine signaling on VIC phenotype and nodule formation were tested using a co-culture platform with physiologically relevant matrix elasticities and diffusion distance. 100 µm thin poly(ethylene glycol) (PEG) hydrogels of 3-27 kPa Young's moduli were fabricated in transwell inserts. VICs were cultured on the gels, as VIC phenotype is known to change significantly within this range, while VlvECs lined the underside of the membrane. Co-culture with VlvECs significantly reduced VIC activation to the myofibroblast phenotype on all gels with the largest percent decrease on the 3 kPa gels (~70%), while stiffer gels resulted in approximately 20-30% decrease. Additionally, VlvECs significantly reduced αSMA protein expression (~2 fold lower) on both 3 and 27 kPa gels, as well as the number (~2 fold lower) of nodules formed on the 27 kPa gels. Effects of VlvECs were prevented when nitric oxide (NO) release was inhibited with l-NAME, suggesting that VlvEC produced NO inhibits VIC activation. Withdrawal of l-NAME after 3, 5, and 7 days with restoration of VlvEC NO production for 2 additional days led to a partial reversal of VIC activation (~25% decrease). A potential mechanism by which VlvEC produced NO reduced VIC activation was studied by inhibiting initial and mid-stage cGMP pathway molecules. Inhibition of soluble guanylyl cyclase (sGC) with ODQ or protein kinase G (PKG) with RBrcGMP or stimulation of Rho kinase (ROCK) with LPA, abolished VlvEC effects on VIC activation. This work contributes substantially to the understanding of the valve endothelium's role in preventing VIC functions associated with aortic valve stenosis initiation and progression.


Assuntos
Valva Aórtica/citologia , Elasticidade , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Comunicação Parácrina , Transdução de Sinais , Animais , Células Cultivadas , Técnicas de Cocultura , Módulo de Elasticidade/efeitos dos fármacos , Elasticidade/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/metabolismo , Peso Molecular , Miofibroblastos/citologia , Miofibroblastos/efeitos dos fármacos , Miofibroblastos/metabolismo , Comunicação Parácrina/efeitos dos fármacos , Fenótipo , Polietilenoglicóis/farmacologia , Poliestirenos/farmacologia , Transdução de Sinais/efeitos dos fármacos , Sus scrofa
5.
Circ Res ; 113(2): 186-97, 2013 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-23833293

RESUMO

This review highlights aspects of calcific aortic valve disease that encompass the entire range of aortic valve disease progression from initial cellular changes to aortic valve sclerosis and stenosis, which can be initiated by changes in blood flow (hemodynamics) and pressure across the aortic valve. Appropriate hemodynamics is important for normal valve function and maintenance, but pathological blood velocities and pressure can have profound consequences at the macroscopic to microscopic scales. At the macroscopic scale, hemodynamic forces impart shear stresses on the surface of the valve leaflets and cause deformation of the leaflet tissue. As discussed in this review, these macroscale forces are transduced to the microscale, where they influence the functions of the valvular endothelial cells that line the leaflet surface and the valvular interstitial cells that populate the valve extracellular matrix. For example, pathological changes in blood flow-induced shear stress can cause dysfunction, impairing their homeostatic functions, and pathological stretching of valve tissue caused by elevated transvalvular pressure can activate valvular interstitial cells and latent paracrine signaling cytokines (eg, transforming growth factor-ß1) to promote maladaptive tissue remodeling. Collectively, these coordinated and complex interactions adversely impact bulk valve tissue properties, feeding back to further deteriorate valve function and propagate valve cell pathological responses. Here, we review the role of hemodynamic forces in calcific aortic valve disease initiation and progression, with focus on cellular responses and how they feed back to exacerbate aortic valve dysfunction.


Assuntos
Valva Aórtica/fisiologia , Calcinose/patologia , Cardiomiopatias/patologia , Cardiopatias Congênitas/patologia , Doenças das Valvas Cardíacas/patologia , Hemodinâmica/fisiologia , Miócitos Cardíacos/fisiologia , Animais , Valva Aórtica/citologia , Valva Aórtica/patologia , Valva Aórtica/fisiopatologia , Doença da Válvula Aórtica Bicúspide , Calcinose/fisiopatologia , Cardiomiopatias/fisiopatologia , Cardiopatias Congênitas/fisiopatologia , Doenças das Valvas Cardíacas/fisiopatologia , Humanos , Miócitos Cardíacos/patologia
6.
Acta Biomater ; 8(9): 3201-9, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22609448

RESUMO

A thiol-ene polymerization platform was used to synthesize peptide functionalized poly(ethylene glycol) hydrogels, which were initially characterized and compared to theoretical predictions of Young's modulus via a theoretical crosslinking density equation presented herein. After thorough characterization, this material system's utility for answering specific biological hypotheses was demonstrated with the culture and observation of aortic valvular interstitial cells (VICs). Specifically, these materials were used to better understand the role of substrate elasticity and biochemical functionality on VIC α-smooth muscle (αSMA) expression and secretory properties (i.e. de novo extracellular matrix (ECM)). The Young's moduli of the hydrogels varied from 28kPa (activating, 90% myofibroblasts) to 4kPa (non-activating, 15% myofibroblast), and the biochemical functionality was tailored by incorporating three small adhesive peptide sequences, RGDS, VGVAPG and P15. To promote VIC adhesion, a basal [RGDS] of 0.8mM was used in all formulations, while the [VGVAPG] or [P15] were varied to be lower than, equal to or higher than 0.8mM. The substrates with 1.2mM VGVAPG and all gels with P15 led to significantly higher αSMA expression for both stiff and soft substrates, as compared to 0.8mM RGDS alone. Importantly, all gel conditions αSMA expression were significantly lower than tissue culture poly(styrene) (TCPS; ∼4- to 10-fold difference). The ECM produced decreased significantly as the total integrin-binding peptide concentration increased, but was significantly higher than that produced on TCPS. This easily tailored material system provides a useful culture platform to improve the fundamental understanding of VIC biology through isolating specific biological cues and observing VIC function.


Assuntos
Hidrogéis , Peptídeos/química , Compostos de Sulfidrila/química , Sequência de Aminoácidos , Adesão Celular , Matriz Extracelular/química , Dados de Sequência Molecular
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...