首页> 外文会议>World biomaterials congress >Creating tissue-engineered blood vessels as disease models and drug screening platforms
【24h】

Creating tissue-engineered blood vessels as disease models and drug screening platforms

机译:创建组织工程血管作为疾病模型和药物筛选平台

获取原文

摘要

Introduction: Existing models for drug efficacy and toxicity tests, including human cell lines and animal models, cannot accurately predict how the drugs will behave in people -9 out of 10 experimental drugs fail in clinical studies, even though they pass tests in cell and animal models [FDA news]. Conceivably, in vitro drugs may cause significant functional changes at tissue or organ level but not in conventional 2D cell culture; in vivo, rodent and human response is different. Therefore, physiologically relevant and functional human tissue models have been increasingly valued as alternatives for drug screening. In this work, we report the fabrication of functional tissue-engineered blood vessels (TEBVs) with human cells. Materials and Methods: TEBV fabrication: TEBVs with an inner diameter <0.80 mm were fabricated from: ⅰ) sheets of aligned human mesenchymal stem cells (hMSC) (Fig A) and ⅱ) human cells embedded in dense collagen gel. TEBV functional test: Vessel diameters were measured before and after perfusion with vasoconstrictor phenylephrine (1 MM), vasodilators acetylcholine (1 pM), or buffer alone with or without changes in flow rate. Media after perfusion were harvested to measure nitric oxide (NO) release from endothelial cells. Imaging of immune cell adhesion and transendothelium migration: Monocytes and cells to construct TEBVs were labeled with fluorescent dyes. TEBVs were treated with 20ng/mL TNF-a for two hours and then perfused with medium containing HL-60 cells (2x10A5 cell/mL) at 0.5 dynes shear stress for one hour. Confocal images were taken at both the side and the topview of the TEBV lumen to track immune cell perfusion, attachment, and transendothelial migration (TEM) events. 3D images and movies showing cell migration and TEM were reconstructed with Imaris 7.7. Significances indicated as * p<0.05," p<0.01 by t-test. Results and Discussion: Confluent aligned hMSC sheets were harvested from nano-grooved PDMS substrate (350nm width, 350nm height, and 700nm periodicity), wrapped circumferentially around a Teflon rod and matured in a rotating wall bioreactor. After further maturation under perfusion for 3 weeks and endothelial progenitor cell coating, vessels increased in strength and exhibited excellent handling and suturing characteristics (Fig A). Flow increased the vessel diameter and nitric oxide (NO) release (Fig B), indicating a functional endothelium. TEBVs exhibited vasoconstriction that increased with the dose of phenylephrine, and vasodilation induced by acetylcholine. To facilitate disease modeling of atherosclerosis, branched TEBVs were fabricated using 3D-printed molds, in which cells were mixed with collagen (1.5mg/mL) to form collagen-gel-TEBV of different sizes (Fig C). Perfusion of monocytes in TEBV with activated endothelium showed that monocytes could attach to the lumen of the TEBV and migrate transendothelially (Fig D, E). With these TEBV models, we were able to monitor different functional changes at tissue level. These models may be applied for drug efficacy and toxicity evaluation as well as disease modeling, like atherosclerosis. Ongoing work includes screening of drugs that have vascular toxicity and fabrication of TEBVs with cells from patients afflicted with genetic disorders. Figure 1. Fabrication of TEBVs and functional tests. A: Schematic diagram of cell-sheet TEBV formation; B: NO release under different perfusion speed and comparison with native vessel; C: Fabrication of branched TEBV using 3D printed molds, made at different sizes (1mm-20mm) with a resolution at 0.1 um; D: Perfusion of monocytes in TEBV; arrows indicate monocytes in perfusion or attached to the lumen of TEBV (Blue: Collagen fibers from second harmonic image; Red: HL-60). E: Left panel is the cross-section view of a TEBV (Blue: cell nucleus; Red: HL-60; Green: cells in TEBV), right panel is the top view of the TEBV lumen (Blue: collagen fiber; Red: cells in TEBV; Green: HL-60). Monocyte adhesion and trans-endothelial migration were observed in real-time. Scale bar in C is 20mm, in D and E are 50μm.
机译:简介:现有的药物功效和毒性测试模型,包括人类细胞系和动物模型,即使通过了细胞和动物测试,也无法准确预测药物在人中的行为-10种实验药物中有-9种在临床研究中失败型号[FDA新闻]。可以想象,体外药物可能会在组织或器官水平上引起明显的功能变化,而在常规2D细胞培养中却不会。在体内,啮齿动物和人类的反应是不同的。因此,生理上相关且功能正常的人体组织模型已被越来越多地视为药物筛选的替代方法。在这项工作中,我们报告了具有人类细胞的功能性组织工程血管(TEBV)的制造。材料和方法:TEBV的制造:内径<0.80 mm的TEBV的制造方法是:ⅰ)对齐的人间充质干细胞(hMSC)片(图A)和ⅱ)嵌入致密胶原凝胶的人细胞。 TEBV功能测试:分别在血管收缩剂去氧肾上腺素(1 MM),血管扩张剂乙酰胆碱(1 pM)或单独缓冲液(有或无流速变化)的灌注前后测量血管直径。收集灌注后的培养基以测量内皮细胞释放的一氧化氮(NO)。免疫细胞粘附和内皮细胞迁移的成像:用荧光染料标记单核细胞和构建TEBV的细胞。将TEBV用20ng / mL TNF-a处理2小时,然后在0.5达因剪切应力下用含有HL-60细胞(2x10A5细胞/ mL)的培养基灌注1小时。共聚焦图像是在TEBV腔的侧面和顶视图上拍摄的,以跟踪免疫细胞的灌注,附着和跨内皮迁移(TEM)事件。使用Imaris 7.7重建了显示细胞迁移和TEM的3D图像和电影。通过t检验表明* * p <0.05,p <0.01。结果与讨论:从纳米沟槽PDMS基板(350nm宽度,350nm高度和700nm周期性)中收集汇合的对齐的hMSC片,将其周向缠绕在特氟隆上杆并在旋转壁生物反应器中成熟,在灌注下进一步成熟3周和内皮祖细胞涂层后,血管强度增加,并具有出色的处理和缝合特性(图A),流量增加了血管直径和一氧化氮(NO)。释放(图B),表明内皮功能正常TEBVs的血管收缩随去氧肾上腺素的剂量增加而增加,乙酰胆碱引起的血管舒张为了促进动脉粥样硬化的疾病建模,使用3D打印的模具制造了分支的TEBVs,其中混合了细胞用胶原蛋白(1.5mg / mL)形成不同大小的胶原蛋白凝胶TEBV(图C)。结婚单核细胞可以附着到TEBV的内腔和跨内皮迁移(图D,E)。使用这些TEBV模型,我们能够在组织水平上监视不同的功能变化。这些模型可用于药物功效和毒性评估以及疾病建模,例如动脉粥样硬化。正在进行的工作包括筛选具有血管毒性的药物,以及使用患有遗传性疾病的患者的细胞制造TEBV。图1. TEBV的制造和功能测试。 A:细胞片TEBV形成示意图; B:在不同的灌注速度下和与天然血管比较NO释放; C:使用3D打印模具制造支化TEBV,以不同尺寸(1mm-20mm)制作,分辨率为0.1 um; D:在TEBV中灌注单核细胞;箭头指示灌注或附着于TEBV腔的单核细胞(蓝色:二次谐波图像中的胶原纤维;红色:HL-60)。 E:左图是TEBV的截面图(蓝色:细胞核;红色:HL-60;绿色:TEBV中的细胞),右图是TEBV管腔的俯视图(蓝色:胶原纤维;红色: TEBV中的细胞;绿色:HL-60)。实时观察到单核细胞粘附和跨内皮迁移。 C中的比例尺为20mm,D和E中的比例尺为50μm。

著录项

相似文献

  • 外文文献
  • 中文文献
  • 专利
获取原文

客服邮箱:kefu@zhangqiaokeyan.com

京公网安备:11010802029741号 ICP备案号:京ICP备15016152号-6 六维联合信息科技 (北京) 有限公司©版权所有
  • 客服微信

  • 服务号