4.8 Article

A synthetic mechanogenetic gene circuit for autonomous drug delivery in engineered tissues

期刊

SCIENCE ADVANCES
卷 7, 期 5, 页码 -

出版社

AMER ASSOC ADVANCEMENT SCIENCE
DOI: 10.1126/sciadv.abd9858

关键词

-

资金

  1. Shriners Hospitals for Children
  2. NIH [AR76665, AG46927, AG15768, AR072999, AR74240, AR73752, AR074992, AR073221, AG28716]
  3. Nancy Taylor Foundation
  4. Arthritis Foundation
  5. NSF Graduate Research Fellowship Program [DGE-1745038]
  6. Phillip and Sima Needleman Fellowship
  7. Duke School of Medicine
  8. Duke Clinical and Translational Science Award (CTSA) [UL1TR001117]
  9. NSF EAGER Award

向作者/读者索取更多资源

This study presents a novel approach using synthetic biology and tissue engineering to develop a mechanically responsive bioartificial tissue that produces therapeutic biologic drug in response to mechanical loading. By deconstructing signaling networks induced by TRPV4 activation, synthetic TRPV4-responsive genetic circuits were created in chondrocytes to produce anti-inflammatory drug in response to mechanical loading, protecting tissues from inflammatory degradation.
Mechanobiologic signals regulate cellular responses under physiologic and pathologic conditions. Using synthetic biology and tissue engineering, we developed a mechanically responsive bioartificial tissue that responds to mechanical loading to produce a preprogrammed therapeutic biologic drug. By deconstructing the signaling networks induced by activation of the mechanically sensitive ion channel transient receptor potential vanilloid 4 (TRPV4), we created synthetic TRPV4-responsive genetic circuits in chondrocytes. We engineered these cells into living tissues that respond to mechanical loading by producing the anti-inflammatory biologic drug interleukin-1 receptor antagonist. Chondrocyte TRPV4 is activated by osmotic loading and not by direct cellular deformation, suggesting that tissue loading is transduced into an osmotic signal that activates TRPV4. Either osmotic or mechanical loading of tissues transduced with TRPV4-responsive circuits protected constructs from inflammatory degradation by interleukin-1 alpha. This synthetic mechanobiology approach was used to develop a mechanogenetic system to enable long-term, autonomously regulated drug delivery driven by physiologically relevant loading.

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

4.8
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