4.6 Article

RNA Aptamer Targeting of Adam8 in Cancer Growth and Metastasis

期刊

CANCERS
卷 15, 期 12, 页码 -

出版社

MDPI
DOI: 10.3390/cancers15123254

关键词

aptamer; Adam8; cancer; metastasis; myCAF

类别

向作者/读者索取更多资源

An RNA aptamer was developed to target and inhibit the extracellular sheddase domain of Adam8 in both in vitro and in vivo studies. This aptamer effectively blocked the activities of extracellular Adam8, leading to the reversal of myofibroblast cancer-associated fibroblast phenotype induced by cancer-derived osteopontin. The findings suggest that the signal pathways involved in the development and maintenance of the myCAF phenotype may differ, and that targeting extracellular Adam8 sheddase activity could be a potential therapeutic approach.
Simple Summary An RNA aptamer targeting the extracellular sheddase domain of Adam8 was isolated and characterized both in vitro and in vivo. In co-cultures of human mesenchymal stem cells with human MDA-MB-231 breast cancer or Hep G2 cell liver cancer cells, the aptamer blocked extracellular Adam8 activities with associated reversal of the previously established cancer-derived osteopontin-induced myofibroblast cancer-associated fibroblast phenotype (myCAF). Our results suggest that the signal pathways that initiate the development of the myCAF phenotype may be distinct from those required for maintenance, that extracellular Adam8 sheddase activity is required for maintenance of the myCAF phenotype, and that this aptamer may serve as a vehicle for the further investigation of this new pathway. Cancer progression depends on an accumulation of metastasis-supporting physiological changes, which are regulated by cell-signaling molecules. In this regard, a disintegrin and metalloproteinase 8 (Adam8) is a transmembrane glycoprotein that is selectively expressed and induced by a variety of inflammatory stimuli. In this study, we identified Adam8 as a sox2-dependent protein expressed in MDA-MB-231 breast cancer cells when cocultured with mesenchymal-stem-cell-derived myofibroblast-like cancer-associated fibroblasts (myCAF). We have previously found that myCAF-induced cancer stemness is required for the maintenance of the myCAF phenotype, suggesting that the initiation and maintenance of the myCAF phenotype require distinct cell-signaling crosstalk pathways between cancer cells and myCAF. Adam8 was identified as a candidate secreted protein induced by myCAF-mediated cancer stemness. Adam8 has a known sheddase function against which we developed an RNA aptamer, namely, Adam8-Apt1-26nt. The Adam8-Apt1-26nt-mediated blockade of the extracellular soluble Adam8 metalloproteinase domain abolishes the previously initiated myCAF phenotype, or, termed differently, blocks the maintenance of the myCAF phenotype. Consequently, cancer stemness is significantly decreased. Xenograft models show that Adam8-Apt-1-26nt administration is associated with decreased tumor growth and metastasis, while flow cytometric analyses demonstrate a significantly decreased fraction of myCAF after Adam8-Apt-1-26nt treatment. The role of soluble Adam8 in the maintenance of the myCAF phenotype has not been previously characterized. Our study suggests that the signal pathways for the induction or initiation of the myCAF phenotype may be distinct from those involved with the maintenance of the myCAF phenotype.

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

4.6
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