首页> 外文会议>World biomaterials congress >Enhancing hepatic functions of human induced pluripotent stem cells-derived hepatocytes and their engraftment in immunocompetent mice via 3D co-aggregation and encapsulation
【24h】

Enhancing hepatic functions of human induced pluripotent stem cells-derived hepatocytes and their engraftment in immunocompetent mice via 3D co-aggregation and encapsulation

机译:通过3D共聚集和包囊增强人类诱导的多能干细胞衍生的肝细胞的肝功能及其在免疫活性小鼠中的移植

获取原文

摘要

Introduction: Liver diseases affect over 600 million people worldwide. Currently, liver transplantation is the only therapy to prevent mortality of end-stage liver diseases. However, the situation is deteriorating given the severe shortage of donor livers/hepatocytes. Human induced pluripotent stem cells (iPSCs) are promising cell source to provide sufficient hepatocytes for transplantation. To generate hepatocytes from human iPSCs, various approaches have been energetically pursued in the past decade. Although hepatocyte-like cells were derived to some extent, their functions were similar to fetal rather than adult human hepatocytes. Moreover, previous transplantations were performed in immunocompromised animals without immunoisolation of hepatocyte-like cells; whether these cells can engraft, mature and function in immunocompetent animals and eventually humans remains unknown. In this study, we combined the 2D monolayer culture and differentiation with the 3D microwell co-culture with stromal cells (SCs) to enhance the maturation and function of human iPSCs-derived hepatocytes (iPS-H) in vitro. Furthermore, we encapsulated the iPS-H/SCs aggregates in recently developed, non-fibrotic alginate capsules and evaluated the in vivo function of iPS-H in an immunocompetent mouse model. Materials and Methods: PDMS microwells were fabricated using standard soft lithography. Undifferentiated iPSCs were maintained and differentiated into iPS-H on 2D monolayer culture. To enhance the maturation and function of iPS-H, 3D co-aggregates of iPS-H/SCs (Cell ratio: iPS-H:SCs=2:1) were formed in PDMS microwells. Cell aggregates of iPS-H/SCs in PDMS microwells were analyzed for albumin secretion and cytochrome P450 activity. Cell aggregates of iPS-H/SCs were encapsulated in recently developed, non-fibrotic alginate capsules and transplanted into the intraperitoneal cavity of immunocompetent C57BL/6 mice. Mouse blood was collected and human albumin in mouse serum was measured for 24 days. Results and Discussion: iPS-H were differentiated from iPSCs on 2D monolayer and then placed into PDMS microwells with SCs in 3D to recapitulate the vital cell-cell and cell-matrix interactions in vivo. Numerous size-controllable iPS-H/SCs aggregates were formed in microwells (Fig 1 a). Compared with cell aggregates of iPS-H alone, iPS-H/SCs aggregates generated more mature and functional hepatocytes as indicated by enhanced albumin secretion (Fig 1b) and cytochrome P450 activities (Fig 1c). Cell aggregates of iPS-H alone consistently secreted albumin at low level. In contrast, the amount of secreted albumin gradually increased as culture time prolonged in iPS-H/SCs aggregates. It is well-known that cytochrome P450 enzymes are of great importance in phase Ⅰ xenobiotic biotransformation. CYP3A4 and CYP2C9 activities of iPS-H/SCs aggregates increased approximately 1.4 ~ 3.0 folds when compared to cell aggregates of iPS-H alone. The physical contacts, deposited matrix proteins, and paracrine signaling from SCs in 3D may play important roles in formation of cell aggregates as well as maturation and function of iPS-H. The iPS-H/SCs aggregates were collected from microwells (Fig 2a) and then encapsulated into alginate capsules (Fig 2b). The capsules containing cell aggregates were transplanted into immunocompetent mice to evaluate the engraftment and function of iPS-H in vivo. The beneficial effect of alginate encapsulation was obvious as human albumin secreted from iPS-H/SCs aggregates was detected in mouse sera for 24 days in vivo (Fig. 2c). The level of secretion was generally comparable to Hum-H/SCs aggregates and previous reports using immunocompromised mice. Conclusion: The in vitro hepatic functions of iPS-H were enhanced by 3D co-aggregation of iPS-H and SCs in microwells. Engraftment of iPS-H in immunocompetent mice was demonstrated through encapsulation of iPS-H/SCs aggregates in biocompatible alginate capsules. Human albumin in mouse sera secreted by encapsulated iPS-H/SCs aggregates reached a level comparable to the primary human hepatocyte control. This proof-of-concept study provides a simple yet robust approach to improve the engraftment of iPS-H and may be applicable to other stem cell-based therapies.
机译:简介:肝病影响全球超过6亿人。当前,肝移植是防止终末期肝病死亡的唯一疗法。但是,由于供体肝脏/肝细胞严重短缺,这种情况正在恶化。人类诱导的多能干细胞(iPSC)是有希望的细胞来源,可为移植提供足够的肝细胞。为了从人iPSC产生肝细胞,在过去的十年中一直在大力追求各种方法。尽管肝细胞样细胞在某种程度上是衍生的,但它们的功能与胎儿的相似,而不是成年的人类肝细胞。而且,先前的移植是在免疫受损的动物中进行的,没有免疫分离肝细胞样细胞。这些细胞是否可以在具有免疫能力的动物中以及最终在人类中移植,成熟并发挥功能,仍然未知。在这项研究中,我们将2D单层培养和分化与3D微孔共培养与基质细胞(SCs)结合在一起,以增强人iPSCs衍生的肝细胞(iPS-H)的体外成熟和功能。此外,我们将iPS-H / SCs聚集体封装在最近开发的非纤维化藻酸盐胶囊中,并在具有免疫功能的小鼠模型中评估了iPS-H的体内功能。材料和方法:PDMS微孔是使用标准的软光刻技术制造的。维持未分化的iPSC,并在2D单层培养上将其分化为iPS-H。为了增强iPS-H的成熟度和功能,在PDMS微孔中形成了iPS-H / SCs的3D共聚集体(细胞比例:iPS-H:SCs = 2:1)。分析PDMS微孔中iPS-H / SCs的细胞聚集体的白蛋白分泌和细胞色素P450活性。将iPS-H / SCs的细胞聚集体封装在最近开发的非纤维化藻酸盐胶囊中,然后移植到具有免疫功能的C57BL / 6小鼠的腹膜腔中。收集小鼠血液,并测量小鼠血清中的人白蛋白24天。结果与讨论:在2D单层上将iPS-H与iPSC区别开,然后将其放入3D SC的PDMS微孔中,以概括体内重要的细胞-细胞和细胞-基质相互作用。在微孔中形成了许多大小可控的iPS-H / SCs聚集体(图1a)。与单独的iPS-H细胞聚集体相比,iPS-H / SCs聚集体产生更多成熟和功能性的肝细胞,如白蛋白分泌增强(图1b)和细胞色素P450活性(图1c)所示。单独的iPS-H的细胞聚集体持续分泌低水平的白蛋白。相反,随着培养时间延长,iPS-H / SCs聚集物中分泌白蛋白的量逐渐增加。众所周知,细胞色素P450酶在Ⅰ期异源生物转化中非常重要。与单独的iPS-H细胞聚集体相比,iPS-H / SCs聚集体的CYP3A4和CYP2C9活性增加约1.4〜3.0倍。 3D中来自SC的物理接触,沉积的基质蛋白和旁分泌信号可能在细胞聚集体的形成以及iPS-H的成熟和功能中发挥重要作用。从微孔中收集iPS-H / SCs聚集体(图2a),然后将其封装到藻酸盐胶囊中(图2b)。将含有细胞聚集体的胶囊移植到具有免疫能力的小鼠中,以评估iPS-H在体内的植入和功能。藻酸盐封装的有益作用是显而易见的,因为在体内检测了小鼠血清中iPS-H / SCs聚集体分泌的人白蛋白已有24天(图2c)。分泌水平通常与Hum-H / SCs聚集体和使用免疫受损小鼠的先前报道相当。结论:iPS-H和SC在微孔中的3D共聚集可增强iPS-H的体外肝功能。通过将iPS-H / SCs聚集体封装在生物相容性藻酸盐胶囊中,证明了iPS-H在免疫活性小鼠中的植入。封装的iPS-H / SCs聚集体分泌的小鼠血清中的人白蛋白达到了与原代人肝细胞对照相当的水平。这项概念验证研究提供了一种简单而强大的方法来改善iPS-H的植入,并且可能适用于其他基于干细胞的疗法。

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