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共聚合和包封增强人诱导的多能干细胞衍生肝细胞的肝细胞及其在免疫活性小鼠中的肝细胞功能

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 encaps
机译:介绍:肝病在全球范围内影响超过6亿人。目前,肝移植是唯一可防止终末期肝病死亡率的疗法。然而,鉴于供体/肝细胞严重短缺的情况恶化。人诱导多能干细胞(IPSC)是有前途的细胞源,以提供足够的肝细胞进行移植。为了产生来自人体IPSC的肝细胞,在过去十年中,各种方法都在充满活力地追求。尽管在一定程度上得出肝细胞样细胞,但它们的功能与胎儿相似而不是成人的人肝细胞。此外,先前的移植在免疫环化动物中进行,而不免疫肝细胞样细胞;这些细胞是否可以在免疫活性动物中植入,成熟和功能,最终人类仍然未知。在这项研究中,我们将2D单层培养物与三维微孔共同培养物的分化与基质细胞(SCS)组合,以增强体外人IPSCS衍生的肝细胞(IPS-H)的成熟和功能。此外,我们封装了最近开发的非纤维分离藻酸盐胶囊中的IPS-H / SCs骨料,并评估了免疫活性小鼠模型中IPS-H的体内功能。材料和方法:使用标准软光刻制造PDMS微孔。未分化的IPSC在2D单层培养物上维持和分化为IPS-H.为了增强IPS-H的成熟和功能,在PDMS微孔中形成IPS-H / SCS的3D共聚合(单元比:IPS-H:SCS = 2:1)。针对白蛋白分泌和细胞色素P450活性分析了PDMS微孔中IPS-H / SCs的细胞聚集体。 IPS-H / SCS的细胞聚集在最近开发的非纤维分离藻酸盐胶囊中包封,并移植到免疫活性C57BL / 6小鼠的腹腔内腔中。收集小鼠血液,并测量小鼠血清中的人白蛋白24天。结果和讨论:IPS-H与2D单层的IPSC分化,然后用3D中的SCS置于PDMS微孔中,以重新延长体内重要细胞 - 细胞和细胞基质相互作用。在微孔中形成了许多尺寸可控的IPS-H / SCS聚集体(图1A)。与单独的IPS-H的细胞聚集体相比,IPS-H / SCS聚集在增强白蛋白分泌(图1B)和细胞色素P450活性(图1C)中产生的产生更成熟和功能性肝细胞。 IPS-H的细胞聚集在低水平下始终分泌白蛋白。相反,分泌的白蛋白的量随着培养时间延长IPS-H / SCS聚集体逐渐增加。众所周知,细胞色素P450酶在Ⅰ期血管生物转化中具有重要意义。与单独的IPS-H的细胞聚集体相比,CYP3A4和CYP2C9的IPS-H / SCS聚集体的活动增加了大约1.4〜3.0折。从3D中的SCS的物理接触,沉积基质蛋白和旁静脉信号传导可能在形成细胞聚集体以及IPS-H的成熟和功能中起重要作用。从微孔(图2A)收集IPS-H / SCS骨料,然后包封成藻酸盐胶囊(图2B)。将含有细胞聚集体的胶囊移植到免疫活性小鼠中以评估体内IPS-H的植入和功能。藻酸盐包封的有益效果明显,因为在小鼠血清中检测到从IPS-H / SCS聚集体中分泌的人白蛋白在体内24天(图2C)。分泌水平通常与使用免疫血肿小鼠的嗡嗡声-H / SCs聚集体和先前的报道相当。结论:通过微孔的IPS-H和SCS的3D共聚合增强了IPS-H的体外肝功能。通过在生物相容性藻酸盐胶囊中的IPS-H / SCS聚集体的包封来证明IPS-H在免疫活性小鼠中的植入。在小鼠血清中分泌的人类白蛋白由encaps分泌

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