4.7 Article

Patient-derived small intestinal myofibroblasts direct perfused, physiologically responsive capillary development in a microfluidic Gut-on-a-Chip Model

期刊

SCIENTIFIC REPORTS
卷 10, 期 1, 页码 -

出版社

NATURE PUBLISHING GROUP
DOI: 10.1038/s41598-020-60672-5

关键词

-

资金

  1. Children's Discovery Institute of Washington University in St. Louis
  2. St. Louis Children's Hospital [CDI-CORE-2015-505, MI-FR2017-596, MI-F-2017-629]
  3. National Institutes of Health [R01 DK-104698, R03DK111473, R01DK118568, R01 DK-106382, R01DK-112378, R01DK109384, K01DK109081, K08DK101608, 4T32HD043010-14, 3T32DK007130-45S1]
  4. Digestive Diseases Research Core Center Grant NIDDK [P30 DK052574]
  5. Children's Surgical Sciences Research Institute of the St. Louis Children's Hospital
  6. Association for Academic Surgery Foundation
  7. March of Dimes Foundation [5-FY17-79]
  8. Department of Pediatrics at Washington University School of Medicine, St. Louis
  9. Lawrence C. Pakula MD IBD Research, Innovation, and Education Fund
  10. Washington University School of Medicine
  11. Children's Discovery Institute of Washington University
  12. Foundation for Barnes-Jewish Hospital [3770]

向作者/读者索取更多资源

The development and physiologic role of small intestine (SI) vasculature is poorly studied. This is partly due to a lack of targetable, organ-specific markers for in vivo studies of two critical tissue components: endothelium and stroma. This challenge is exacerbated by limitations of traditional cell culture techniques, which fail to recapitulate mechanobiologic stimuli known to affect vessel development. Here, we construct and characterize a 3D in vitro microfluidic model that supports the growth of patient-derived intestinal subepithelial myofibroblasts (ISEMFs) and endothelial cells (ECs) into perfused capillary networks. We report how ISEMF and EC-derived vasculature responds to physiologic parameters such as oxygen tension, cell density, growth factors, and pharmacotherapy with an antineoplastic agent (Erlotinib). Finally, we demonstrate effects of ISEMF and EC co-culture on patient-derived human intestinal epithelial cells (HIECs), and incorporate perfused vasculature into a gut-on-a-chip (GOC) model that includes HIECs. Overall, we demonstrate that ISEMFs possess angiogenic properties as evidenced by their ability to reliably, reproducibly, and quantifiably facilitate development of perfused vasculature in a microfluidic system. We furthermore demonstrate the feasibility of including perfused vasculature, including ISEMFs, as critical components of a novel, patient-derived, GOC system with translational relevance as a platform for precision and personalized medicine research.

作者

我是这篇论文的作者
点击您的名字以认领此论文并将其添加到您的个人资料中。

评论

主要评分

4.7
评分不足

次要评分

新颖性
-
重要性
-
科学严谨性
-
评价这篇论文

推荐

暂无数据
暂无数据