4.7 Article

HIF-1α/YAP Signaling Rewrites Glucose/Iodine Metabolism Program to Promote Papillary Thyroid Cancer Progression

期刊

出版社

IVYSPRING INT PUBL
DOI: 10.7150/ijbs.75459

关键词

Papillary thyroid cancer; Glucose/Iodine metabolism; HIF-1 alpha; YAP; Progression.

向作者/读者索取更多资源

This study revealed a novel regulatory mechanism of glucose/iodine metabolic reprogramming in papillary thyroid cancer (PTC). Under hypoxic conditions, YAP activation accelerated glycolysis and reduced the expression of NIS. In addition, the HIF-1 alpha/YAP signaling indirectly reduced NIS expression. These findings have important implications for understanding the development of aggressive PTC.
Background: The management of aggressive and progressive metastatic papillary thyroid cancer (PTC) is very difficult. An inverse relationship between radioiodine and F-18 fluorodeoxyglucose (FDG) uptake (``flip-flop'' phenomenon) is described for invasive PTC during dedifferentiation. However, no satisfactory biologic explanation for this phenomenon. Hypoxia is an important microenvironmental factor that promotes cancer progression and glycolysis. The Hippo-YAP is a highly conserved tumor suppressor pathway and contributes to cancer metabolic reprogramming. Thus, we investigated the underlying molecular mechanisms of glucose/iodine metabolic reprogramming in PTC, focusing on the tumor hypoxia microenvironment and Hippo-YAP signaling. Methods: Immunohistochemistry staining was conducted to evaluate the expressions of hypoxia-inducible factor 1 alpha (HIF-1 alpha), yes-associated protein (YAP), glucose transporters 1 (GLUT1) and sodium iodine symporter (NIS) in matched PTC and the adjacent noncancerous tissues. PTC cell lines were cultured under normoxic (20% O-2) and hypoxic (1% O-2) conditions and the glycolysis level and NIS expression were measured. Further, we characterized the molecular mechanism of glucose/iodine metabolic reprogramming in PTC cell. Finally, we validated the results in vivo by establishing subcutaneous xenografts in nude mice. Results: The expression levels of HIF1-alpha, YAP and GLUT1 were upregulated in PTC tissues and YAP expression was positively associated with HIF-1 alpha, GLUT1 and TNM stages. Meanwhile, the expression of NIS was negatively correlated with YAP. Further, in vitro studies indicated that hypoxia-induced YAP activation was critical for accelerating glycolysis and reducing NIS expression in PTC cells. Inhibition of YAP had the opposite effects in vitro and tumorigenicity in vivo. Hypoxia inhibited the Hippo signaling pathway resulting in the inactivation of YAP phosphorylation, further promoting the nuclear localization of YAP in PTC cells. The mechanism is that hypoxic stress promoted YAP binding to HIF-1 alpha in the nucleus and maintained HIF-1 alpha protein stability. The YAP/HIF-1 alpha complex bound and directly activated the GLUT1 transcription to accelerate glycolysis. Meanwhile, HIF-1 alpha/YAP signaling might indirectly reduce the expression of NIS by promoting the output of MAPK signaling. In vivo studies confirmed the YAP-mediated reprogramming of glucose/iodine metabolism promoted PTC progression. Conclusions: Collectively, our data revealed a novel regulatory mechanism of the glucose/iodine metabolic program rewritten by HIF-1 alpha/YAP signaling in PTC. Inhibition of HIF-1 alpha/YAP signaling alone or in combination with other potential markers may effectively combat aggressive PTC.

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