Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
RSC Adv ; 11(27): 16510-16521, 2021 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-35479169

RESUMO

The valve replacement is the main treatment of heart valve disease. However, thrombus formation following valve replacement has always been a major clinical drawback. Accelerating the endothelialization of cardiac valve prosthesis is the main approach to reduce thrombus. In the current study, a titanium nanotube was biofunctionalized with a chitosan/genipin heparin hydrogel and the controlled release of interleukin-4 (IL-4), and its regulation of macrophages was investigated to see if it could influence endothelial cells to eventually accelerate endothelialization. TNT60 (titanium dioxide nanotubes, 60 V) with nanoarray was obtained by anodic oxidation of 60 V, and IL-4 was loaded into the nanotube by vacuum drying. The hydrogel (chitosan : genipin = 4 : 1) was applied to the surface of the nanotubes following drying, and the heparin drops were placed on the hydrogel surface with chitosan as the polycation and heparin as the polyanion. A TNT/IL-4/G (G = gel, chitosan/genipin heparin) delivery system was prepared. Our results demonstrated that the biofunctionalization of titanium nanotube with chitosan/genipin heparin hydrogel and the controlled release of IL-4 had a significant regulatory effect on macrophage M2 polarization, reducing the inflammatory factor release and higher secretion of VEGF (vascular endothelial growth factor), which can accelerate the endothelialization of the implant.

2.
Am J Transl Res ; 12(7): 3329-3345, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32774703

RESUMO

Calcific aortic valve disease (CAVD) currently lacks a highly effective in vitro model. The presence of high concentrations of serum inorganic phosphate in patients with end-stage renal disease leads to calcification of vascular and aortic valves. Therefore, we applied inorganic phosphate to induce the osteogenic differentiation of valvular interstitial cells (VICs) and mimic its in vivo pathophysiological effects. Calcification and inflammatory response assays determined that inorganic phosphate-osteogenic induction medium (IP-OIM) was more efficient than classic osteogenic induction medium (OIM) containing organic glycerophosphate. Levels of BMP-2, RhoA, and ROCK-1 were significantly increased in IP-OIM cells. Knockdown efficiency of BMP-2- and RhoA-siRNA in VICs was evaluated, and expression of RhoA and its downstream target ROCK-1 was decreased after BMP-2-siRNA transfection. Moreover, ROCK-1 was significantly downregulated after RhoA knockdown, whereas expression of BMP-2 was unchanged. Interference of BMP-2 had a stronger anti-calcification effect than RhoA, further identifying BMP-2 as an upstream regulator of RhoA/ROCK-1. Stimulation of VICs by IP-OIM led to increased Smad1/5/9 phosphorylation, which peaked at 60 min, while pre-treatment of VICs with the Smad1/5/9 inhibitor Compound C attenuated VICs calcification. These results suggest that IP-OIM induced VICs osteogenic differentiation via Smad1/5/9 signaling. Knockdown of BMP-2 or RhoA also decreased Smad1/5/9 phosphorylation also decreased. We conclude that the RhoA/ROCK-1 axis participates in VICs osteogenic differentiation as a "bypass mediator" of the BMP-2/Smad1/5/9 signaling pathway.

3.
Int J Nanomedicine ; 14: 441-455, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30666106

RESUMO

Background: Macrophages play important roles in the immune response to, and successful implantation of, biomaterials. Titanium nanotubes are considered promising heart valve stent materials owing to their effects on modulation of macrophage behavior. However, the effects of nanotube-regulated macrophages on endothelial cells, which are essential for stent endothelialization, are unknown. Therefore, in this study we evaluated the inflammatory responses of endothelial cells to titanium nanotubes prepared at different voltages. Methods and results: In this study we used three different voltages (20, 40, and 60 V) to produce titania nanotubes with three different diameters by anodic oxidation. The state of macrophages on the samples was assessed, and the supernatants were collected as conditioned media (CM) to stimulate human umbilical vein endothelial cells (HUVECs), with pure titanium as a control group. The results indicated that titanium dioxide (TiO2) nanotubes induced macrophage polarization toward the anti-inflammatory M2 state and increased the expression of arginase-1, mannose receptor, and interleukin 10. Further mechanistic analysis revealed that M2 macrophage polarization controlled by the TiO2 nanotube surface activated the phosphatidylinositol 3-kinase/AKT and extracellular signal-regulated kinase 1/2 pathways through release of vascular endothelial growth factor to influence endothelialization. Conclusion: Our findings expanded our understanding of the complex influence of nanotubes in implants and the macrophage inflammatory response. Furthermore, CM generated from culture on the TiO2 nanotube surface may represent an integrated research model for studying the interactions of two different cell types and may be a promising approach for accelerating stent endothelialization through immunoregulation.


Assuntos
Biomarcadores/análise , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Nanotubos/química , Neovascularização Fisiológica/efeitos dos fármacos , Titânio/farmacologia , Células Cultivadas , Humanos , Macrófagos/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/farmacologia , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Titânio/química , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
4.
RSC Adv ; 9(21): 11882-11893, 2019 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-35517024

RESUMO

Decellularized valve stents are widely used in tissue-engineered heart valves because they maintain the morphological structure of natural valves, have good histocompatibility and low immunogenicity. However, the surface of the cell valve loses the original endothelial cell coverage, exposing collagen and causing calcification and decay of the valve in advance. In this study, poly ε-caprolactone (PCL) nanoparticles loaded with osteoprotegerin (OPG) were bridged to a decellularized valve using a nanoparticle drug delivery system and tissue engineering technology to construct a new anti-calcification composite valve with sustained release function. The PCL nanoparticles loaded with OPG were prepared via an emulsion solvent evaporation method, which had a particle size of 133 nm and zeta potential of -27.8 mV. Transmission electron microscopy demonstrated that the prepared nanoparticles were round in shape, regular in size, and uniformly distributed, with an encapsulation efficiency of 75%, slow release in vitro, no burst release, no cytotoxicity to BMSCs, and contained OPG nanoparticles in vitro. There was a delay in the differentiation of BMSCs into osteoblasts. The decellularized valve modified by nanoparticles remained intact and its collagen fibers were continuous. After 8 weeks of subcutaneous implantation in rats, the morphological structure of the valve was almost complete, and the composite valve showed anti-calcification ability to a certain extent.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...