Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Int J Mol Sci ; 23(10)2022 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-35628424

RESUMO

Cardiovascular diseases are considered one of the worldwide causes of death, with atherosclerosis being the most predominant. Nowadays, the gold standard treatment is blood vessel replacement by bypass surgery; however, autologous source is not always possible. Thereby, tissue-engineered blood vessels (TEBVs) are emerging as a potential alternative source. In terms of composition, collagen has been selected in many occasions to develop TEBVs as it is one of the main extracellular matrix components of arteries. However, it requires specific support or additional processing to maintain the tubular structure and appropriate mechanical properties. Here, we present a method to develop support-free collagen TEBVs with co-axial extrusion in a one-step procedure with high concentrated collagen. The highest concentration of collagen of 20 mg/mL presented a burst pressure of 619.55 ± 48.77 mmHg, being able to withstand perfusion of 10 dynes/cm2. Viability results showed a high percentage of viability (86.1 and 85.8% with 10 and 20 mg/mL, respectively) of human aortic smooth muscle cells (HASMCs) and human umbilical vein endothelial cells (HUVEC) after 24 h extrusion. Additionally, HUVEC and HASMCs were mainly localized in their respective layers, mimicking the native distribution. All in all, this approach allows the direct extrusion of collagen TEBVs in a one-step procedure with enough mechanical properties to be perfused.


Assuntos
Colágeno , Engenharia Tecidual , Artérias/fisiologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Miócitos de Músculo Liso , Engenharia Tecidual/métodos
2.
Adv Funct Mater ; 30(37)2020 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-33708027

RESUMO

From micro-scaled capillaries to millimeter-sized arteries and veins, human vasculature spans multiple scales and cell types. The convergence of bioengineering, materials science, and stem cell biology has enabled tissue engineers to recreate the structure and function of different hierarchical levels of the vascular tree. Engineering large-scale vessels has been pursued over the past thirty years to replace or bypass damaged arteries, arterioles, and venules, and their routine application in the clinic may become a reality in the near future. Strategies to engineer meso- and microvasculature have been extensively explored to generate models to study vascular biology, drug transport, and disease progression, as well as for vascularizing engineered tissues for regenerative medicine. However, bioengineering of large-scale tissues and whole organs for transplantation, have failed to result in clinical translation due to the lack of proper integrated vasculature for effective oxygen and nutrient delivery. The development of strategies to generate multi-scale vascular networks and their direct anastomosis to host vasculature would greatly benefit this formidable goal. In this review, we discuss design considerations and technologies for engineering millimeter-, meso-, and micro-scale vessels. We further provide examples of recent state-of-the-art strategies to engineer multi-scale vasculature. Finally, we identify key challenges limiting the translation of vascularized tissues and offer our perspective on future directions for exploration.

3.
J Surg Res ; 221: 143-151, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29229120

RESUMO

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.


Assuntos
Prótese Vascular , Infecções/etiologia , Politetrafluoretileno , Infecções Relacionadas à Prótese/etiologia , Enxerto Vascular/efeitos adversos , Animais , Escherichia coli , Masculino , Ratos Sprague-Dawley , Staphylococcus aureus
4.
Adv Exp Med Biol ; 1064: 123-144, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30471030

RESUMO

Cardiovascular disease is the number one cause of death in the U.S and results in the loss of approximately one million lives and more than 400 billion U.S. dollars for treatments every year. Recently, tissue engineered blood vessels have been studied and developed as promising replacements for treatment with autologous veins. Here, we summarize the cell sources and methods to make tissue-engineered blood vessels (TEBVs), the recent progress in TEBV related research, and also the recent progress in TEBV related clinical studies.


Assuntos
Vasos Sanguíneos , Doenças Cardiovasculares/terapia , Engenharia Tecidual , Humanos
5.
Biochem Biophys Res Commun ; 473(3): 733-42, 2016 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-26427871

RESUMO

Proper blood vessel networks are necessary for constructing and re-constructing tissues, promoting wound healing, and delivering metabolic necessities throughout the body. Conversely, an understanding of vascular dysfunction has provided insight into the pathogenesis and progression of diseases both common and rare. Recent advances in stem cell-based regenerative medicine - including advances in stem cell technologies and related progress in bioscaffold design and complex tissue engineering - have allowed rapid advances in the field of vascular biology, leading in turn to more advanced modeling of vascular pathophysiology and improved engineering of vascularized tissue constructs. In this review we examine recent advances in the field of stem cell-derived vasculature, providing an overview of stem cell technologies as a source for vascular cell types and then focusing on their use in three primary areas: studies of vascular development and angiogenesis, improved disease modeling, and the engineering of vascularized constructs for tissue-level modeling and cell-based therapies.


Assuntos
Materiais Biocompatíveis/química , Vasos Sanguíneos/citologia , Imageamento Tridimensional/métodos , Células-Tronco/citologia , Engenharia Tecidual/métodos , Animais , Terapia Baseada em Transplante de Células e Tecidos , Modelos Animais de Doenças , Células-Tronco Embrionárias/citologia , Células Endoteliais/citologia , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Microcirculação , Músculo Liso Vascular/citologia , Alicerces Teciduais , Cicatrização/fisiologia
6.
ACS Biomater Sci Eng ; 10(8): 5154-5167, 2024 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-39079153

RESUMO

There remains a lack of small-caliber tissue-engineered blood vessels (TEBVs) with wide clinical use. Biotubes were developed by electrospinning and in-body tissue architecture (iBTA) technology to prepare small-caliber TEBVs with promising applications. Different ratios of hybrid fibers of poly(l-lactic-co-ε-caprolactone) (PLCL) and polyurethane (PU) were obtained by electrospinning, and the electrospun tubes were then implanted subcutaneously in the abdominal area of a rabbit (as an in vivo bioreactor). The biotubes were harvested after 4 weeks. They were then decellularized and cross-linked with heparin. PLCL/PU electrospun vascular tubes, decellularized biotubes (D-biotubes), and heparinized combined decellularized biotubes (H + D-biotubes) underwent carotid artery allograft transplantation in a rabbit model. Vascular ultrasound follow-up and histological observation revealed that the biotubes developed based on electrospinning and iBTA technology, after decellularization and heparinization cross-linking, showed a better patency rate, adequate mechanical properties, and remodeling ability in the rabbit model. IBTA technology caused a higher patency, and the heparinization cross-linking process gave the biotubes stronger mechanical properties.


Assuntos
Prótese Vascular , Heparina , Poliésteres , Engenharia Tecidual , Animais , Coelhos , Engenharia Tecidual/métodos , Heparina/química , Poliésteres/química , Aloenxertos , Poliuretanos/química , Alicerces Teciduais/química , Artérias Carótidas
7.
JVS Vasc Sci ; 4: 100120, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37662589

RESUMO

Objective: Synthetic expanded polytetrafluoroethylene (ePTFE) grafts are known to be susceptible to bacterial infection. Results from preclinical and clinical studies of bioengineered human acellular vessels (HAVs) have shown relatively low rates of infection. This study evaluates the interactions of human neutrophils and bacteria with ePTFE and HAV vascular conduits to determine whether there is a correlation between neutrophil-conduit interactions and observed differences of their infectivity in vivo. Methods: A phase III comparative clinical study between investigational HAVs (n = 177) and commercial ePTFE grafts (n = 178) used for hemodialysis access (ClinicalTrials.gov Identifier: NCT02644941) was evaluated for conduit infection rates followed by histological analyses of HAV and ePTFE tissue explants. The clinical histopathology of HAV and ePTFE conduits reported to be infected was compared with immunohistochemistry of explanted materials from a preclinical model of bacterial contamination. Mechanistic in vitro studies were then conducted using isolated human neutrophils seeded directly onto HAV and ePTFE materials to analyze neutrophil viability, morphology, and function. Results: Clinical trial results showed that the HAV had a significantly lower (0.93%; P = .0413) infection rate than that of ePTFE (4.54%). Histological analysis of sections from infected grafts explanted approximately 1 year after implantation revealed gram-positive bacteria near cannulation sites. Immunohistochemistry of HAV and ePTFE implanted in a well-controlled rodent infection model suggested that the ePTFE matrix permitted bacterial infiltration and colonization but may be inaccessible to neutrophils. In the same model, the HAV showed host recellularization and lacked detectable bacteria at the 2-week explant. In vitro results demonstrated that the viability of human neutrophils decreased significantly upon exposure to ePTFE, which was associated with neutrophil elastase release in the absence of bacteria. In contrast, neutrophils exposed to the HAV material retained high viability and native morphology. Cocultures of neutrophils and Staphylococcus aureus on the conduit materials demonstrated that neutrophils were more effective at ensnaring and degrading bacteria on the HAV than on ePTFE. Conclusions: The HAV material seems to demonstrate a resistance to bacterial infection. This infection resistance is likely due to the HAV's native-like material composition, which may be more biocompatible with host neutrophils than synthetic vascular graft material.

8.
Int J Biol Macromol ; 209(Pt B): 2070-2083, 2022 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-35500770

RESUMO

Cardiovascular diseases and vascular trauma can be commonly found in the population. Scholars worldwide hope to develop small-diameter vascular grafts that can replace autologous vessels for clinical use. Decellularized blood vessels can retain the original morphology, structure, and physical properties of blood vessels, which is conducive to cell growth, proliferation, and differentiation. In this study, porcine coronary arteries (PCAs) were decellularized to prepare decellularized porcine coronary artery (DPCA), and bilayer hybrid scaffolds were prepared by coating gelatin and sodium alginate mixed hydrogel of seven different proportions and combined with mouse fibroblasts (L929 cells) to study the construction of tissue engineering vessels in vitro. The obtained bilayer hybrid scaffolds were 3-7 cm in length, 5 mm in external diameter, and 1 mm in average wall thickness. All seven bilayer hybrid scaffolds showed good biocompatibility after cell inoculation. Compared with 2D culture, cells on 3D scaffolds grew relatively slowly in the first 4 days, and the number of cells proliferated rapidly at 7 days. In the same culture days, different concentrations of hydrogel also had an impact on cell proliferation. With the increase of hydrogel content, cells on the 3D scaffold formed cell colonies faster. The results showed that the scaffold had good biocompatibility and could meet the needs of artificial blood vessel construction.


Assuntos
Gelatina , Hidrogéis , Alginatos , Animais , Vasos Coronários , Gelatina/química , Hidrogéis/farmacologia , Camundongos , Suínos , Engenharia Tecidual/métodos , Alicerces Teciduais/química
9.
Regen Ther ; 21: 277-281, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36092503

RESUMO

At present, the vascular grafts used in clinic are mainly autologous blood vessels, but they often face the dilemma of no blood vessels available due to limited sources. However, synthetic blood vessels made of polytetrafluoroethylene (ePTFE), which is commonly used in clinic, are prone to thrombosis and intimal hyperplasia, and the long-term patency rate is poor, so its effectiveness is severely limited, which is far from meeting the clinical needs. With the development of nano-materials, stem cells and 3D bio-printing technology, people began to explore the preparation of new endothelialized vascular grafts through this technology. Nano-peptide materials have excellent biocompatibility, can be compounded with different bioactive molecules, and have unique advantages in cultivating stem cells. It has been reported that self-assembled nano-polypeptide hydrogel was successfully constructed, mesenchymal stem cells were correctly isolated and cultured, and their transformation into blood vessels was studied. It was confirmed that the 3D bio-printed nano-polypeptide hydrogel tissue ADMSCs still had strong vascular endothelial differentiation ability. The application of mesenchymal stem cells and nano-polypeptide hydrogel in tissue engineering blood vessels has gradually become a research hotspot, and it is expected to develop a new type of transplanted blood vessel that meets the physiological functions of human body in terms of vascular endothelialization, cell compatibility and histocompatibility, so as to realize the customized and personalized printing of the endothelialized transplanted blood vessel according to the shape of the target blood vessel, which has attractive prospects and far-reaching social and economic benefits.

10.
Biomaterials ; 280: 121286, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34871879

RESUMO

A functional multi-scale vascular network can promote 3D engineered tissue growth and improve transplantation outcome. In this work, by using a combination of living cells, biological hydrogel, and biodegradable synthetic polymer we fabricated a biocompatible, multi-scale vascular network (MSVT) within thick, implantable engineered tissues. Using a templating technique, macro-vessels were patterned in a 3D biodegradable polymeric scaffold seeded with endothelial and support cells within a collagen gel. The lumen of the macro-vessel was lined with endothelial cells, which further sprouted and anastomosed with the surrounding self-assembled capillaries. Anastomoses between the two-scaled vascular systems displayed tightly bonded cell junctions, as indicated by vascular endothelial cadherin expression. Moreover, MSVT functionality and patency were demonstrated by dextran passage through the interconnected multi-scale vasculature. Additionally, physiological flow conditions were applied with home-designed flow bioreactors, to achieve a MSVT with a natural endothelium structure. Finally, implantation of a multi-scale-vascularized graft in a mouse model resulted in extensive host vessel penetration into the graft and a significant increase in blood perfusion via the engineered vessels compared to control micro-scale-vascularized graft. Designing and fabricating such multi-scale vascular architectures within 3D engineered tissues may benefit both in vitro models and therapeutic translation research.


Assuntos
Vasos Sanguíneos , Capilares , Células Endoteliais , Engenharia Tecidual , Animais , Materiais Biocompatíveis , Colágeno/química , Hidrogéis , Camundongos , Engenharia Tecidual/métodos , Alicerces Teciduais
11.
ACS Appl Mater Interfaces ; 14(25): 28591-28603, 2022 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-35696386

RESUMO

The development of a well-designed tissue-engineered blood vessel (TEBV) still remains a challenge. In recent years, approaches in which the host response to implanted biomaterials is used to generate vascular constructs within the patient's body have gained increasing interest. The delivery of growth factors to these in situ-engineered vascular grafts might enhance myofibroblast recruitment and the secretion of essential extracellular matrix proteins, thereby optimizing their functional properties. Layer-by-layer (LbL) coating has emerged as an innovative technology for the controlled delivery of growth factors in tissue engineering applications. In this study, we combined the use of surface-etched polymeric rods with LbL coatings to control the delivery of TGF-ß1, PDGF-BB, and IGF-1 and steer the foreign body response toward the formation of a functional vascular graft. Results showed that the regenerated tissue is composed of elastin, glycosaminoglycans, and circumferentially oriented collagen fibers, without calcification or systemic spill of the released growth factors. Functional controlled delivery was observed, whereas myofibroblast-rich tissue capsules were formed with enhanced collagen and elastin syntheses using TGF-ß1 and TGF-ß1/PDGF-BB releasing rods, when compared to control rods that were solely surface-engineered by chloroform etching. By combining our optimized LbL method and surface-engineered rods in an in vivo bioreactor approach, we could regulate the fate and ECM composition of in situ-engineered vascular grafts to create a successful in vivo vascular tissue-engineered replacement.


Assuntos
Elastina , Fator de Crescimento Transformador beta1 , Becaplermina , Prótese Vascular , Colágeno , Humanos , Engenharia Tecidual/métodos
12.
Front Cardiovasc Med ; 9: 847554, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35310996

RESUMO

Despite the efforts devoted to drug discovery and development, the number of new drug approvals have been decreasing. Specifically, cardiovascular developments have been showing amongst the lowest levels of approvals. In addition, concerns over the adverse effects of drugs to the cardiovascular system have been increasing and resulting in failure at the preclinical level as well as withdrawal of drugs post-marketing. Besides factors such as the increased cost of clinical trials and increases in the requirements and the complexity of the regulatory processes, there is also a gap between the currently existing pre-clinical screening methods and the clinical studies in humans. This gap is mainly caused by the lack of complexity in the currently used 2D cell culture-based screening systems, which do not accurately reflect human physiological conditions. Cell-based drug screening is widely accepted and extensively used and can provide an initial indication of the drugs' therapeutic efficacy and potential cytotoxicity. However, in vitro cell-based evaluation could in many instances provide contradictory findings to the in vivo testing in animal models and clinical trials. This drawback is related to the failure of these 2D cell culture systems to recapitulate the human physiological microenvironment in which the cells reside. In the body, cells reside within a complex physiological setting, where they interact with and respond to neighboring cells, extracellular matrix, mechanical stress, blood shear stress, and many other factors. These factors in sum affect the cellular response and the specific pathways that regulate variable vital functions such as proliferation, apoptosis, and differentiation. Although pre-clinical in vivo animal models provide this level of complexity, cross species differences can also cause contradictory results from that seen when the drug enters clinical trials. Thus, there is a need to better mimic human physiological conditions in pre-clinical studies to improve the efficiency of drug screening. A novel approach is to develop 3D tissue engineered miniaturized constructs in vitro that are based on human cells. In this review, we discuss the factors that should be considered to produce a successful vascular construct that is derived from human cells and is both reliable and reproducible.

13.
Adv Biol (Weinh) ; 5(4): e2000428, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33852179

RESUMO

Atherosclerosis begins with the accumulation of cholesterol-carrying lipoproteins on blood vessel walls and progresses to endothelial cell dysfunction, monocyte adhesion, and foam cell formation. Endothelialized tissue-engineered blood vessels (TEBVs) have previously been fabricated to recapitulate artery functionalities, including vasoconstriction, vasodilation, and endothelium activation. Here, the initiation of atherosclerosis is emulated by designing branched TEBVs (brTEBVs) of various geometries treated with enzyme-modified low-density-lipoprotein (eLDL) and TNF-α to induce endothelial cell dysfunction and adhesion of perfused human monocytes. Locations of monocyte adhesion under pulsatile flow are identified, and the hemodynamics in the brTEBVs are characterized using particle image velocimetry (PIV) and computational fluid dynamics (CFD). Monocyte adhesion is greater at the side outlets than at the main outlets or inlets, and is greatest at larger side outlet branching angles (60° or 80° vs 45°). In PIV experiments, the branched side outlets are identified as atherosclerosis-prone areas where fluorescent particles show a transient swirling motion following flow pulses; in CFD simulations, side outlets with larger branching angles show higher vorticity magnitude and greater flow disturbance than other areas. These results suggest that the branched TEBVs with eLDL/TNF-α treatment provide a physiologically relevant model of early atherosclerosis for preclinical studies.


Assuntos
Aterosclerose , Artérias , Células Espumosas , Humanos , Lipoproteínas LDL , Fluxo Pulsátil
14.
Front Cardiovasc Med ; 8: 790529, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35155603

RESUMO

Atherosclerosis is the primary cause of hardening and narrowing arteries, leading to cardiovascular disease accounting for the high mortality in the United States. For developing effective treatments for atherosclerosis, considerable efforts have been devoted to developing in vitro models. Compared to animal models, in vitro models can provide great opportunities to obtain data more efficiently, economically. Therefore, this review discusses the recent progress in in vitro models for atherosclerosis studies, including traditional two-dimensional (2D) systems cultured on the tissue culture plate, 2D cell sheets, and recently emerged microfluidic chip models with 2D culture. In addition, advanced in vitro three-dimensional models such as spheroids, cell-laden hydrogel constructs, tissue-engineered blood vessels, and vessel-on-a-chip will also be covered. Moreover, the functions of these models are also summarized along with model discussion. Lastly, the future perspectives of this field are discussed.

15.
Adv Healthc Mater ; 7(11): e1800026, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29637716

RESUMO

Small-diameter (<6 mm) tissue-engineered blood vessels (TEBVs) have a low patency rate due to chronic inflammation mediated intimal hyperplasia. Functional coating with drug release is a promising solution, but preventing the released drug from being rushed away by blood flow remains a great challenge. A single-walled carboxylic acid functionalized carbon nanotube (C-SWCNT) is used to build an irregular mesh for TEBV coating. However, an interaction between the released drug and the cells is still insufficient due to the blood flow. Thus, an intracellular drug delivery system mediated by macrophage cellular uptake is designed. Resveratrol (RSV) modified CNT is used for macrophage uptake. M1 macrophage uptakes CNT-RSV and then converts to the M2 phenotype upon intracellular RSV release. Prohealing M2 macrophage inhibits the chronic inflammation thus maintains the contractile phenotype of the vascular smooth muscle cell (VSMC), which reduces intimal hyperplasia. Additionally, RSV released from the mesh coating also directly protects the contractile VSMCs from being converted to a secretory phenotype. Through antishear stress coating and macrophage-based intracellular drug delivery, CNT-RSV TEBVs exhibit a long-term anti-intimal hyperplasia function. Animal transplantation studies show that the patency rate remains high until day 90 after grafting in rat carotid arteries.


Assuntos
Prótese Vascular , Materiais Revestidos Biocompatíveis , Nanotubos/química , Resveratrol , Estresse Mecânico , Engenharia Tecidual , Animais , Materiais Revestidos Biocompatíveis/química , Materiais Revestidos Biocompatíveis/farmacologia , Preparações de Ação Retardada/química , Preparações de Ação Retardada/farmacologia , Implantação de Prótese , Ratos , Ratos Sprague-Dawley , Resveratrol/química , Resveratrol/farmacologia
16.
Sci China Life Sci ; 61(10): 1178-1188, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30159681

RESUMO

The rapid endothelialization of tissue-engineered blood vessels (TEBVs) can effectively prevent thrombosis and inhibit intimal hyperplasia. The traditional Chinese medicine ingredient icariin is highly promising for the treatment of cardiovascular diseases. ß-cyclodextrin sulfate is a type of hollow molecule that has good biocompatibility and anticoagulation properties and exhibits a sustained release of icariin. We studied whether icariin-loaded ß-cyclodextrin sulfate can promote the endothelialization of TEBVs. The experimental results showed that icariin could significantly promote the proliferation and migration of endothelial progenitor cells; at the same time, icariin could promote the migration of rat vascular endothelial cells (RAVECs). Subsequently, we used an electrostatic force to modify the surface of the TEBVs with icariin-loaded ß-cyclodextrin sulfate, and these vessels were implanted into the rat common carotid artery. After 3 months, micro-CT results showed that the TEBVs modified using icariin-loaded ß-cyclodextrin sulfate had a greater patency rate. Scanning electron microscopy (SEM) and CD31 immunofluorescence results showed a better degree of endothelialization. Taken together, icariin-loaded ß-cyclodextrin sulfate can achieve anticoagulation and rapid endothelialization of TEBVs to ensure their long-term patency.


Assuntos
Coagulação Sanguínea/efeitos dos fármacos , Vasos Sanguíneos/efeitos dos fármacos , Células Progenitoras Endoteliais/efeitos dos fármacos , Flavonoides/farmacologia , beta-Ciclodextrinas/farmacologia , Animais , Prótese Vascular , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/fisiologia , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Medicamentos de Ervas Chinesas/química , Medicamentos de Ervas Chinesas/farmacologia , Células Progenitoras Endoteliais/metabolismo , Células Progenitoras Endoteliais/fisiologia , Flavonoides/química , Ratos Sprague-Dawley , Sulfatos/metabolismo , Engenharia Tecidual/métodos , beta-Ciclodextrinas/química , beta-Ciclodextrinas/metabolismo
17.
J Biomed Mater Res A ; 106(10): 2634-2642, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29790247

RESUMO

The mobilization and homing of endothelial progenitor cells (EPCs) contribute to the rapid endothelialization of tissue engineering blood vessel (TEBV). Inflammation can affect TEBV patency, and monocytes/macrophages (MM) are the main effector cells. But it is not clear how EPCs interact with MM after TEBV transplantation. Our results showed acellular materials would not directly cause acute and severe inflammatory responses but activate E-selectin expression in homing EPCs, gradually promoting the polarization of MM to the M1. Adenosine A2a receptor agonist CGS21680 promoted the secretion of more proangiogenic factors from MM, inducing EPC migration and mobilization. CGS21680 could inhibit MM polarization to the M1 type through the down-regulation of EPC proinflammatory molecules, such as E-selectin. Chitosan/(2-hydroxypropyl)-ß-cyclodextrin nanoparticles were prepared to control the release of CGS-21680 and then modified to TEBVs through layer-by-layer assembly. Animal experiments showed that this TEBV can maintain patency for 6 months and good endothelialization was observed. In summary, our results showed the regulation of EPC pro-inflammatory activities is a new approach to enhance TEBV patency. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A: 2634-2642, 2018.


Assuntos
Adenosina/análogos & derivados , Células Progenitoras Endoteliais/patologia , Inflamação/patologia , Fenetilaminas/farmacologia , Engenharia Tecidual/métodos , Grau de Desobstrução Vascular/efeitos dos fármacos , 2-Hidroxipropil-beta-Ciclodextrina/química , Adenosina/química , Adenosina/farmacologia , Animais , Prótese Vascular , Contagem de Células , Movimento Celular/efeitos dos fármacos , Angiografia por Tomografia Computadorizada , Preparações de Ação Retardada , Células Progenitoras Endoteliais/efeitos dos fármacos , Células Progenitoras Endoteliais/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Monócitos/citologia , Monócitos/efeitos dos fármacos , Nanopartículas/química , Fenetilaminas/química , Ratos Wistar
18.
ACS Nano ; 11(11): 10964-10973, 2017 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-29035553

RESUMO

Thrombosis is one of the biggest obstacles in the clinical application of small-diameter tissue-engineered blood vessels (TEBVs). The implantation of an unmodified TEBV will lead to platelet aggregation and further activation of the coagulation cascade, in which the high concentration of adenosine diphosphate (ADP) that is released by platelets plays an important role. Inspired by the phenomenon that endothelial cells continuously generate endogenous antiplatelet substances via enzymatic reactions, we designed a reduced graphene oxide (RGO) based dual-enzyme biomimetic cascade to successively convert ADP into adenosine monophosphate (AMP) and AMP into adenosine. We used RGO as a support and bound apyrase and 5'-nucleotidase (5'-NT) on the surface of RGO through covalent bonds, and then, we modified the surface of the collagen-coated decellularized vascular matrix with the RGO-enzyme complexes, in which RGO functions as a platform with a large open surface area and minimal diffusion barriers for substrates/products to integrate two catalytic systems for cascading reactions. The experimental results demonstrate that the two enzymes can synergistically catalyze procoagulant ADP into anticoagulant AMP and adenosine successively under physiological conditions, thus reducing the concentration of ADP. AMP and adenosine can weaken or even reverse the platelet aggregation induced by ADP, thereby inhibiting thrombosis. Adenosine can also accelerate the endothelialization of TEBVs by regulating cellular energy metabolism and optimizing the microenvironment, thus ensuring the antithrombotic function and patency of TEBVs even after the RGO-enzyme complex loses its activity.


Assuntos
Vasos Sanguíneos/química , Fibrinolíticos/uso terapêutico , Trombose/terapia , Engenharia Tecidual , Biomimética , Plaquetas/efeitos dos fármacos , Vasos Sanguíneos/crescimento & desenvolvimento , Vasos Sanguíneos/transplante , Fibrinolíticos/química , Grafite/química , Humanos , Agregação Plaquetária/efeitos dos fármacos , Trombose/patologia
19.
Adv Healthc Mater ; 4(7): 1004-8, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25694105

RESUMO

Regulation of cellular response pattern to phosphorus ion (PI) is a new target for the design of tissue-engineered materials. Changing cellular response pattern to high PI can maintain monocyte/macrophage survival in TEBV and the signal of increasing PI can be converted by klotho to the adenosine signals through the regulation of energy metabolism in monocytes/macrophages.


Assuntos
Vasos Sanguíneos/efeitos dos fármacos , Vasos Sanguíneos/fisiologia , Fósforo/farmacologia , Animais , Macrófagos/efeitos dos fármacos , Macrófagos/fisiologia , Monócitos/efeitos dos fármacos , Monócitos/fisiologia , Ratos , Ratos Wistar , Engenharia Tecidual/métodos
20.
Expert Opin Drug Metab Toxicol ; 11(7): 1021-4, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26028128

RESUMO

Drug-induced vascular injury (DIVI) is a serious problem in preclinical studies of vasoactive molecules and for survivors of pediatric cancers. DIVI is often observed in rodents and some larger animals, primarily with drugs affecting vascular tone, but not in humans; however, DIVI observed in animal studies often precludes a drug candidate from continuing along the development pipeline. Thus, there is great interest by the pharmaceutical industry to identify quantifiable human biomarkers of DIVI. Small-scale endothelialized tissue-engineered blood vessels using human cells represent a promising approach to screen drug candidates and develop alternatives to cancer therapeutics in vitro. We identify several technical challenges that remain to be addressed, including high-throughput systems to screen large numbers of candidates, identification of suitable cell sources and establishing and maintaining a differentiated state of the vessel wall cells. Adequately addressing these challenges should yield novel platforms to screen drugs and develop new therapeutics to treat cardiovascular disease.


Assuntos
Vasos Sanguíneos/efeitos dos fármacos , Engenharia Tecidual/métodos , Doenças Vasculares/induzido quimicamente , Animais , Biomarcadores/metabolismo , Prótese Vascular , Desenho de Fármacos , Avaliação Pré-Clínica de Medicamentos/métodos , Ensaios de Triagem em Larga Escala , Humanos , Especificidade da Espécie , Testes de Toxicidade/métodos , Doenças Vasculares/diagnóstico
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa