Abstract

Hepatocellular carcinoma (HCC) remains the primary cause of cancer-related death. Metabolic change is the major characteristic of cancer. The present study attempted to investigate the regulatory mechanisms of HCC energy metabolism from the perspective of noncoding RNA regulation of HIF1A and LDHA. The expression of miR-100-5p expression was significantly suppressed in HCC tissue samples and HCC cell lines under 1% O2-induced hypoxia. miR-100-5p overexpression significantly suppressed hypoxia-induced increases in lactate concentration and glucose uptake. Exposure to 1% O2 induced HIF1A protein and reduced miR-100-5p expression, while HIF1A silencing dramatically rescued miR-100-5p expression upon 1% O2 exposure. In addition, 1% O2-induced increases in lactate concentration and glucose uptake were also suppressed by HIF1A silencing. Next, by analyzing available data in TCGA, we found that lncRNA RAET1K was correlated with HIF1A and miR-100-5p.LncRNA RAET1K could downregulate the expression of miR-100-5p by acting as a sponge, while HIF1A bound the lncRNA RAET1K promoter region to activate its transcription. LncRNA RAET1K silencing significantly suppressed HCC cell proliferation and invasion and also suppressed hypoxia-induced increases in lactate concentration and glucose uptake, while miR-100-5p inhibition reversed the effects of lncRNA RAET1K silencing on hypoxia-induced glycolysis in HCC cells. Finally, the expression of HIF1A, lncRNA RAET1K, and LDHA was upregulated in HCC tissue specimens; the expression of miR-100-5p was negatively related to HIF1A, lncRNA RAET1K, and LDHA; and HIF1A, lncRNA RAET1K, and LDHA were positively correlated with each other. In conclusion, the HIF1A/lncRNA RAET1K/miR-100-5p axis modulates hypoxia-induced glycolysis in HCC cells and might affect HCC progression.

Highlights

  • Hepatocellular carcinoma (HCC) is the most common type of digestive system tumor and the primary cause of death from cancer[1]

  • The lactate concentration and glucose uptake were significantly induced by hypoxia but inhibited by miR-100-5p overexpression (Fig. 1f, g), suggesting that miR-100-5p overexpression might block glycolysis in cancer cells in a hypoxic microenvironment

  • These data suggest that miR-100-5p might modulate hepatocellular carcinoma (HCC) cell phenotypes by affecting glycolysis under hypoxia

Read more

Summary

Introduction

Hepatocellular carcinoma (HCC) is the most common type of digestive system tumor and the primary cause of death from cancer[1]. It is urgent to fully understand the molecular mechanisms of HCC development. One of the characteristic features of tumor cells is metabolic change[3]. Official journal of the Cell Death Differentiation Association. Zhou et al Cell Death and Disease (2020)11:176. Lactate dehydrogenase A (LDHA) is a member of the tetrameric enzyme lactate dehydrogenase (LDH) family[10] and an essential component of the last step of the glycolytic pathway and exerts critical effects on tumor maintenance and could be regulated by HIF111–13. We attempted to investigate the regulatory mechanisms of HCC energy metabolism from the perspective of HIF1A and LDHA regulation

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call