Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 14(1): 7040, 2024 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-38575597

RESUMO

Whole lung engineering and the transplantation of its products is an ambitious goal and ultimately a viable solution for alleviating the donor-shortage crisis for lung transplants. There are several limitations currently impeding progress in the field with a major obstacle being efficient revascularization of decellularized scaffolds, which requires an extremely large number of cells when using larger pre-clinical animal models. Here, we developed a simple but effective experimental pulmonary bioengineering platform by utilizing the lung as a scaffold. Revascularization of pulmonary vasculature using human umbilical cord vein endothelial cells was feasible using a novel in-house developed perfusion-based bioreactor. The endothelial lumens formed in the peripheral alveolar area were confirmed using a transmission electron microscope. The quality of engineered lung vasculature was evaluated using box-counting analysis of histological images. The engineered mouse lungs were successfully transplanted into the orthotopic thoracic cavity. The engineered vasculature in the lung scaffold showed blood perfusion after transplantation without significant hemorrhage. The mouse-based lung bioengineering system can be utilized as an efficient ex-vivo screening platform for lung tissue engineering.


Assuntos
Células Endoteliais , Transplante de Pulmão , Animais , Humanos , Alicerces Teciduais , Pulmão/irrigação sanguínea , Engenharia Tecidual/métodos , Transplante de Pulmão/métodos , Perfusão , Reatores Biológicos , Matriz Extracelular
2.
Adv Healthc Mater ; 13(19): e2400192, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38518808

RESUMO

Microphysiological and organ-on-chip platforms seek to address critical gaps in human disease models and drug development that underlie poor rates of clinical success for novel interventions. While the fabrication technology and model cells used to synthesize organs-on-chip have advanced considerably, most platforms rely on animal-derived or synthetic extracellular matrix as a cell substrate, limiting mimicry of human physiology and precluding use in modeling diseases in which matrix dynamics play a role in pathogenesis. Here, the development of human cell-derived matrix (hCDM) composite hydrogels for use in 3D microphysiologic models of the vasculature is reported. hCDM composite hydrogels are derived from human donor fibroblasts and maintain a complex milieu of basement membrane, proteoglycans, and nonfibrillar matrix components. The use of hCDM composite hydrogels as 2D and 3D cell culture substrates is demonstrated, and hCDM composite hydrogels are patterned to form engineered human microvessels. Interestingly, hCDM composite hydrogels are enriched in proteins associated with vascular morphogenesis as determined by mass spectrometry, and functional analysis demonstrates proangiogenic signatures in human endothelial cells cultured in these hydrogels. In conclusion, this study suggests that human donor-derived hCDM composite hydrogels could address technical gaps in human organs-on-chip development and serve as substrates to promote vascularization.


Assuntos
Matriz Extracelular , Hidrogéis , Humanos , Hidrogéis/química , Matriz Extracelular/metabolismo , Matriz Extracelular/química , Dispositivos Lab-On-A-Chip , Engenharia Tecidual/métodos , Fibroblastos/metabolismo , Fibroblastos/citologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Neovascularização Fisiológica/efeitos dos fármacos
3.
Adv Healthc Mater ; 13(14): e2302830, 2024 06.
Artigo em Inglês | MEDLINE | ID: mdl-38366136

RESUMO

Tissue engineering holds great promise for regenerative medicine, drug discovery, and as an alternative to animal models. However, as soon as the dimensions of engineered tissue exceed the diffusion limit of oxygen and nutriments, a necrotic core forms leading to irreversible damage. To overcome this constraint, the establishment of a functional perfusion network is essential. In this work, digital light processing bioprinting is used to encapsulate endothelial progenitor cells (EPCs) in 3D light-cured hydrogel scaffolds to guide them toward vascular network formation. In these scaffolds, EPCs proliferate and self-organize within a few days into branched tubular structures with predefined geometry, forming capillary-like vascular tubes or trees of diameters in the range of 10 to 100 µm. Presenting a confluent monolayer wall of cells strongly connect by tight junctions around a central lumen-like space, these structures can be microinjected with a fluorescent dye and are stable for several weeks in vitro. These endothelial structures can be recovered and manipulated in an alginate patch without altering their shape or viability. This approach opens new opportunities for future applications, such as stacking with other cell sheets or multicellular constructs to yield bioengineered tissue with higher complexity and functionality.


Assuntos
Bioimpressão , Células Progenitoras Endoteliais , Engenharia Tecidual , Alicerces Teciduais , Humanos , Bioimpressão/métodos , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Células Progenitoras Endoteliais/citologia , Células Progenitoras Endoteliais/metabolismo , Hidrogéis/química , Capilares/fisiologia , Alginatos/química , Impressão Tridimensional
4.
Trends Mol Med ; 29(1): 35-47, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36371337

RESUMO

Blood vessels are crucial for tissue development, functionality, and homeostasis and are typically a determinant in the progression of healing and regeneration. The tissue microenvironment provides physicochemical cues that affect cellular function, and the study of the microenvironment can be accelerated by the engineering of approaches capable of mimicking various aspects of the microenvironment. In this review, we introduce the major components of the vascular niche and focus on the roles of oxygen and the extracellular matrix (ECM). We demonstrate how vascular engineering approaches enhance our understanding of the microenvironment's impact on the vasculature towards vascular regeneration and describe the current limitations and future directions towards clinical utilization.


Assuntos
Matriz Extracelular , Engenharia Tecidual , Humanos , Cicatrização
6.
Trends Parasitol ; 37(5): 401-413, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33485788

RESUMO

Plasmodium falciparum pathogenesis is complex and intimately connected to vascular physiology. This is exemplified by cerebral malaria (CM), a neurovascular complication that accounts for most of the malaria deaths worldwide. P. falciparum sequestration in the brain microvasculature is a hallmark of CM and is not replicated in animal models. Numerous aspects of the disease are challenging to fully understand from clinical studies, such as parasite binding tropism or causal pathways in blood-brain barrier breakdown. Recent bioengineering approaches allow for the generation of 3D microvessels and organ-specific vasculature that provide precise control of vessel architecture and flow dynamics, and hold great promise for malaria research. Here, we discuss recent and future applications of bioengineered microvessels in malaria pathogenesis research.


Assuntos
Bioengenharia , Microvasos , Parasitologia , Plasmodium falciparum , Animais , Bioengenharia/tendências , Encéfalo/parasitologia , Humanos , Microvasos/química , Microvasos/parasitologia , Parasitologia/métodos , Plasmodium falciparum/fisiologia
7.
Regen Med ; 15(11): 2273-2283, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33325258

RESUMO

Introduction: Resurfacing complex full thickness wounds requires free tissue transfer which creates donor site morbidity. We describe a method to fabricate a skin flap equivalent with a hierarchical microvascular network. Materials & methods: We fabricated a flap of skin-like tissue containing a hierarchical vascular network by sacrificing Pluronic® F127 macrofibers and interwoven microfibers within collagen encapsulating human pericytes and fibroblasts. Channels were seeded with smooth muscle and endothelial cells. Constructs were topically seeded with keratinocytes. Results: After 28 days in culture, multiphoton microscopy revealed a hierarchical interconnected network of macro- and micro-vessels; larger vessels (>100 µm) were lined with a monolayer endothelial neointima and a subendothelial smooth muscle neomedia. Neoangiogenic sprouts formed in the collagen protodermis and pericytes self-assembled around both fabricated vessels and neoangiogenic sprouts. Conclusion: We fabricated a prevascularized scaffold containing a hierarchical 3D network of interconnected macro- and microchannels within a collagen protodermis subjacent to an overlying protoepidermis with the potential for recipient microvascular anastomosis.


Assuntos
Colágeno , Células Endoteliais , Epiderme , Alicerces Teciduais , Fibroblastos , Humanos , Queratinócitos , Pele , Engenharia Tecidual
8.
Elife ; 92020 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-33044167

RESUMO

High levels of proteins called proteoglycans in the walls of umbilical arteries enable these arteries to close rapidly after birth and thus prevent blood loss in newborns.


Assuntos
Proteoglicanas , Artérias Umbilicais , Divisão Celular , Humanos , Recém-Nascido , Caracteres Sexuais
9.
Elife ; 92020 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-32909945

RESUMO

The umbilical artery lumen closes rapidly at birth, preventing neonatal blood loss, whereas the umbilical vein remains patent longer. Here, analysis of umbilical cords from humans and other mammals identified differential arterial-venous proteoglycan dynamics as a determinant of these contrasting vascular responses. The umbilical artery, but not the vein, has an inner layer enriched in the hydrated proteoglycan aggrecan, external to which lie contraction-primed smooth muscle cells (SMC). At birth, SMC contraction drives inner layer buckling and centripetal displacement to occlude the arterial lumen, a mechanism revealed by biomechanical observations and confirmed by computational analyses. This vascular dimorphism arises from spatially regulated proteoglycan expression and breakdown. Mice lacking aggrecan or the metalloprotease ADAMTS1, which degrades proteoglycans, demonstrate their opposing roles in umbilical vascular dimorphism, including effects on SMC differentiation. Umbilical vessel dimorphism is conserved in mammals, suggesting that differential proteoglycan dynamics and inner layer buckling were positively selected during evolution.


Assuntos
Agrecanas/metabolismo , Miócitos de Músculo Liso , Artérias Umbilicais , Proteína ADAMTS1/metabolismo , Animais , Diferenciação Celular/fisiologia , Feminino , Humanos , Camundongos Transgênicos , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/fisiologia , Parto/fisiologia , Gravidez , Artérias Umbilicais/citologia , Artérias Umbilicais/metabolismo , Artérias Umbilicais/fisiologia
10.
Adv Funct Mater ; 30(48)2020 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-33613149

RESUMO

Formation of capillary blood vasculature is a critical requirement for native as well as engineered organs and can be induced in vitro by co-culturing endothelial cells with fibroblasts. However, whether these fibroblasts are required only in the initial morphogenesis of endothelial cells or needed throughout is unknown, and the ability to remove these stromal cells after assembly could be useful for clinical translation. In this study, we introduce a technique termed CAMEO (Controlled Apoptosis in Multicellular Tissues for Engineered Organogenesis), whereby fibroblasts are selectively ablated on demand, and utilize it to probe the dispensability of fibroblasts in vascular morphogenesis. The presence of fibroblasts is shown to be necessary only during the first few days of endothelial cell morphogenesis, after which they can be ablated without significantly affecting the structural and functional features of the developed vasculature. Furthermore, we demonstrate the use of CAMEO to vascularize a construct containing primary human hepatocytes that improved tissue function. In conclusion, this study suggests that transient, initial support from fibroblasts is sufficient to drive vascular morphogenesis in engineered tissues, and this strategy of engineering-via-elimination may provide a new general approach for achieving desired functions and cell compositions in engineered organs.

11.
Stem Cells Dev ; 29(4): 249-256, 2020 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-31701817

RESUMO

The neural crest stem cells derived from human induced pluripotent stem cells (iPSC-NCSCs) are a valuable autologous cell source for tissue engineering and regenerative medicine. In this study, we investigated how iPSC-NCSCs could be regulated to regenerate arteries by microenvironmental factors, including the physical factor of matrix stiffness, and the chemical factor of transforming growth factor beta-1 (TGF-ß1). We found that, compared to soft substrate, stiff substrate drove iPSC-NCSCs differentiation into smooth muscle cells, which was further enhanced by TGF-ß1. To investigate the regulatory role of TGF-ß1 in vivo, we fabricated vascular grafts composed of electrospun nanofibrous scaffolds, collagen gel, iPSC-NCSCs, and TGF-ß1, and implanted them into athymic rats. The results showed that TGF-ß1 significantly promoted extracellular matrix synthesis and increased mechanical strength of vascular grafts. This study presents a proof of concept that iPSC-NCSCs can be used as a promising autologous cell source for vascular regeneration when combined with physical and chemical engineering.


Assuntos
Prótese Vascular , Artérias Carótidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Neurais/efeitos dos fármacos , Alicerces Teciduais , Fator de Crescimento Transformador beta1/farmacologia , Animais , Fenômenos Biomecânicos , Artérias Carótidas/citologia , Artérias Carótidas/fisiologia , Diferenciação Celular/efeitos dos fármacos , Colágeno/química , Colágeno/farmacologia , Géis , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/fisiologia , Nanofibras/química , Nanofibras/ultraestrutura , Crista Neural/citologia , Crista Neural/efeitos dos fármacos , Crista Neural/fisiologia , Células-Tronco Neurais/citologia , Células-Tronco Neurais/fisiologia , Poliésteres/química , Ratos , Ratos Nus , Regeneração/efeitos dos fármacos , Regeneração/fisiologia , Engenharia Tecidual/métodos
12.
Acta Biomater ; 95: 152-164, 2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31271883

RESUMO

3D bioprinted vascular constructs have gained increased interest due to their significant potential for creating customizable alternatives to autologous vessel grafts. In this study, we developed a new approach for biofabricating fibrin-based vascular constructs using a novel rotary 3D bioprinter developed in our lab. We formulated a new bioink by incorporating fibrinogen with gelatin to achieve a desired shear-thinning property for rotary bioprinting. The blending of heat-treated gelatin with fibrinogen turned unprintable fibrinogen into a printable biomaterial for vessel bioprinting by leveraging the favorable rheological properties of gelatin. We discovered that the heat-treatment of gelatin remarkably affects the rheological properties of a gelatin-fibrinogen blended bioink, which in turn influences the printability of the ink. Further characterizations revealed that not only concentration of the gelatin but the heat treatment also affects cell viability during printing. Notably, the density of cells included in the bioinks also influenced printability and tissue volumetric changes of the printed vessel constructs during cultures. We observed increased collagen deposition and construct mechanical strength during two months of the cultures. The burst pressure of the vessel constructs reached 1110 mmHg, which is about 52% of the value of the human saphenous vein. An analysis of the tensile mechanical properties of the printed vessel constructs unveiled an increase in both the circumferential and axial elastic moduli during cultures. This study highlights important considerations for bioink formulation when bioprinting vessel constructs. STATEMENT OF SIGNIFICANCE: There has been an increased demand for small-diameter tissue-engineered vascular grafts. Vascular 3D bioprinting holds the potential to create equivalent vascular grafts but with the ability to tailor them to meet patient's needs. Here, we presented a new and innovative 3D rotary bioprinter and a new bioink formulation for printing vascular constructs using fibrinogen, a favorable biomaterial for vascular tissue engineering. The bioink was formulated by blending fibrinogen with a more printable biomaterial, gelatin. The systematic characterization of the effects of heat treatment and gelatin concentration as well as bioink cell concentration on the printability of the bioink offers new insight into the development of printable biomaterials for tissue biofabrication.


Assuntos
Bioimpressão , Prótese Vascular , Tinta , Impressão Tridimensional , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Animais , Humanos
13.
Microcirculation ; 26(8): e12547, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-30946511

RESUMO

Immense progress in microscale engineering technologies has significantly expanded the capabilities of in vitro cell culture systems for reconstituting physiological microenvironments that are mediated by biomolecular gradients, fluid transport, and mechanical forces. Here, we examine the innovative approaches based on microfabricated vessels for studying lymphatic biology. To help understand the necessary design requirements for microfluidic models, we first summarize lymphatic vessel structure and function. Next, we provide an overview of the molecular and biomechanical mediators of lymphatic vessel function. Then we discuss the past achievements and new opportunities for microfluidic culture models to a broad range of applications pertaining to lymphatic vessel physiology. We emphasize the unique attributes of microfluidic systems that enable the recapitulation of multiple physicochemical cues in vitro for studying lymphatic pathophysiology. Current challenges and future outlooks of microscale technology for studying lymphatics are also discussed. Collectively, we make the assertion that further progress in the development of microscale models will continue to enrich our mechanistic understanding of lymphatic biology and physiology to help realize the promise of the lymphatic vasculature as a therapeutic target for a broad spectrum of diseases.


Assuntos
Dispositivos Lab-On-A-Chip , Vasos Linfáticos/fisiologia , Técnicas Analíticas Microfluídicas , Animais , Humanos , Técnicas de Cultura de Órgãos/instrumentação , Técnicas de Cultura de Órgãos/métodos
14.
Trends Biotechnol ; 37(5): 451-453, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30773221

RESUMO

Vascular engineering requires integrating dimensional flexibility, strength, and bioactivity to fabricate materials that enable diffusive exchange of oxygen and nutrients between cells and their environment. A recent publication (Biomaterials 2019;192:334-345) has described a new method of creating freestanding, tailorable, and biocompatible vascular constructs by coating ice scaffolds with natural or synthetic polymers.


Assuntos
Vasos Sanguíneos , Gelo , Impressão Tridimensional , Engenharia Tecidual , Materiais Biocompatíveis , Vasos Sanguíneos/citologia , Vasos Sanguíneos/fisiologia , Células Cultivadas , Humanos , Alicerces Teciduais
15.
ACS Biomater Sci Eng ; 5(2): 633-643, 2019 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-33405827

RESUMO

Numerous approaches have been employed to improve the efficacy of drug and gene delivery systems, but their strategic development is hindered by a lack of mechanistic understanding and assessment of drug transport and action. Optimizing the efficiency of a drug delivery system requires a detailed understanding of the pharmacokinetics, transendothelial transport, distribution at the tumor site, and uptake in target cells. Elucidating transport kinetics and rate-limiting steps in animal models can be extremely challenging, while in vitro platforms often fail to recapitulate the complexities of drug transport in vivo. To recapitulate the critical aspects of delivery of anticancer agents, we have developed a 3D tissue-engineered microvessel model of the tumor microenvironment. Our model consists of single MDA-MB-231 breast cancer cells embedded within a collagen matrix that surrounds a perfusable cylindrical microvessel lined with human endothelial cells. Here we compare transport and action of free doxorubicin and Doxil, a liposomal formulation of doxorubicin. We show that the mode of drug delivery influences uptake in the vessel endothelium and tumor cells. Through quantification of endothelial and tumor cell proliferation, apoptosis, and motility, we profile the kinetics of drug action with mechanisms of drug transport across the vessel lumen and into the surrounding matrix. Our model can be customized to mimic specific tumor microenvironments and disease states within a physiologically relevant microfluidic platform and provides a basis for characterizing and optimizing drug delivery systems.

16.
Int J Clin Exp Pathol ; 12(7): 2405-2424, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31934068

RESUMO

BACKGROUND: The methods used to rebuild tumour vascular structure and function are called vascular normalization. Vascular normalization methods often block a single angiogenic molecular pathway, but tumor molecular pathways are interconnected and unstable. Since the vascular structure is not repaired, vascularity can be normalized only within a limited time. Amniotic epithelial cells (AECs) are used in tissue engineering to increase blood perfusion and promote wound healing. There have been no reports on the use of AECs in treatment to promote tumor vascular restoration. METHODS: The multipotential stem cell features of AECs were detected by immunofluorescence (IF), RT-PCR, and western blot. A nude rat in situ endometrial carcinoma model was developed. AECs were transfected with lentivirus-green fluorescent protein (GFP)-luciferase (Luc). The vascular formation abilities of AECs were monitored in vitro and in vivo under different conditions. AECs were injected by the rat tail vein, tumour vascular structural and perfusion changes were monitored, and the synergistic effects of AECs with cisplatin (DDP) chemotherapy were evaluated. RESULTS: AECs expressed the stem cell markers OCT4, Nanog, and CK19 at high levels. AECs could differentiate into adipocytes, chondrocytes, and osteocytes. Lentiviral GFP-Luc was successfully transfected into AECs, and GFP-labelled AECs formed vascular tube-like structures and invaded tumor tissue to form vascular structures in vitro. Kinetic luciferase imaging confirmed that AECs homed to rat uterine tumor tissues after injection by the tail vein. After AEC injection, tumour vascular α-SMA/CD31 labelling increased in vascular pericytes, while detection of VEGF-A expression by ELISA decreased. Cadherin labelling showed that basement membrane integrity improved distinctly in the AEC group compared with that in the corresponding control group. Hoechst 33342 and ultrasound Doppler detection showed that tumor vascular perfusion was ameliorated; pimonidazole perfusion showed reduced tumour tissue anoxia, and FITC-dextran perfusion confirmed that vascular leakage was obviously reduced in the AEC group compared with that in the control group. Tumor apoptosis and the rat survival rate in the AEC + DDP group were further enhanced, as demonstrated by CD31 (or α-SMA) IF and GFP colocalization, as well as GFP western blot. AECs differentiated into tumor vascular endotheliocytes or pericytes and enhanced tumor vascular integrity. CONCLUSION: AECs had the characteristics of pluripotent stem cells, and they could vascularize tissues under different conditions. AECs integrated into endometrial cancer vascular structures in nude rats, reduced dysregulated tumour angiogenesis, improved the efficiency of tumour vascular perfusion, and enhanced the cytotoxic effects of DDP. These findings provide a new method for the reconstruction of tumor vessels.

17.
Adv Healthc Mater ; 7(15): e1701461, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29732735

RESUMO

Tissue engineered vascular grafts (TEVGs) are beginning to achieve clinical success and hold promise as a source of grafting material when donor grafts are unsuitable or unavailable. Significant technological advances have generated small-diameter TEVGs that are mechanically stable and promote functional remodeling by regenerating host cells. However, developing a biocompatible blood-contacting surface remains a major challenge. The TEVG luminal surface must avoid negative inflammatory responses and thrombogenesis immediately upon implantation and promote endothelialization. The surface has therefore become a primary focus for research and development efforts. The current state of TEVGs is herein reviewed with an emphasis on the blood-contacting surface. General vascular physiology and developmental challenges and strategies are briefly described, followed by an overview of the materials currently employed in TEVGs. The use of biodegradable materials and stem cells requires careful control of graft composition, degradation behavior, and cell recruitment ability to ensure that a physiologically relevant vessel structure is ultimately achieved. The establishment of a stable monolayer of endothelial cells and the quiescence of smooth muscle cells are critical to the maintenance of patency. Several strategies to modify blood-contacting surfaces to resist thrombosis and control cellular recruitment are reviewed, including coatings of biomimetic peptides and heparin.


Assuntos
Engenharia Tecidual/métodos , Animais , Prótese Vascular , Implante de Prótese Vascular , Colágeno/química , Humanos , Suínos , Alicerces Teciduais/química
18.
Adv Mater ; 29(37)2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28737278

RESUMO

A photodegradable material-based approach to generate endothelialized 3D vascular networks within cell-laden hydrogel biomaterials is introduced. Exploiting multiphoton lithography, microchannel networks spanning nearly all size scales of native human vasculature are readily generated with unprecedented user-defined 4D control. Intraluminal channel architectures of synthetic vessels are fully customizable, providing new opportunities for next-generation microfluidics and directed cell function.


Assuntos
Engenharia Tecidual , Materiais Biocompatíveis , Humanos , Hidrogéis , Microfluídica , Fotólise
19.
Biomaterials ; 35(1): 83-91, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24099708

RESUMO

Over the past 30 years, silk has been proposed for numerous biomedical applications that go beyond its traditional use as a suture material. Silk sutures are well tolerated in humans, but the use of silk for vascular engineering applications still requires extensive biocompatibility testing. Some studies have indicated a need to modify silk to yield a hemocompatible surface. This study examined the potential of low molecular weight heparin as a material for refining silk properties by acting as a carrier for vascular endothelial growth factor (VEGF) and improving silk hemocompatibility. Heparinized silk showed a controlled VEGF release over 6 days; the released VEGF was bioactive and supported the growth of human endothelial cells. Silk samples were then assessed using a humanized hemocompatibility system that employs whole blood and endothelial cells. The overall thrombogenic response for silk was very low and similar to the clinical reference material polytetrafluoroethylene. Despite an initial inflammatory response to silk, apparent as complement and leukocyte activation, the endothelium was maintained in a resting, anticoagulant state. The low thrombogenic response and the ability to control VEGF release support the further development of silk for vascular applications.


Assuntos
Materiais Biocompatíveis , Vasos Sanguíneos/citologia , Heparina , Seda , Engenharia Tecidual , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA