Abstract

Autophagy favors both cell survival and cancer suppression, and increasing evidence reveals that microRNAs (MIRs) regulate autophagy. Previously we reported that MIR126 is downregulated in malignant mesothelioma (MM). Therefore, we investigated the role of MIR126 in the regulation of cell metabolism and autophagy in MM models. We report that MIR126 induces autophagic flux in MM cells by downregulating insulin receptor substrate-1 (IRS1) and disrupting the IRS1 signaling pathway. This was specific to MM cells, and was not observed in non-malignant cells of mesothelial origin or in MM cells expressing MIR126-insensitive IRS1 transcript. The MIR126 effect on autophagy in MM cells was recapitulated by IRS1 silencing, and antagonized by IRS1 overexpression or antisense MIR126 treatment. The MIR126-induced loss of IRS1 suppressed glucose uptake, leading to energy deprivation and AMPK-dependent phosphorylation of ULK1. In addition, MIR126 stimulated lipid droplet accumulation in a hypoxia-inducible factor-1α (HIF1α)-dependent manner. MIR126 also reduced pyruvate dehydrogenase kinase (PDK) and acetyl-CoA-citrate lyase (ACL) expression, leading to the accumulation of cytosolic citrate and paradoxical inhibition of pyruvate dehydrogenase (PDH) activity. Simultaneous pharmacological and genetic intervention with PDK and ACL activity phenocopied the effects of MIR126. This suggests that in MM MIR126 initiates a metabolic program leading to high autophagic flux and HIF1α stabilization, incompatible with tumor progression of MM. Consistently, MIR126-expressing MM cells injected into immunocompromised mice failed to progress beyond the initial stage of tumor formation, showing that increased autophagy has a protective role in MM.

Highlights

  • Metabolic reprogramming of cancer cells is essential for their adaptation to tumor microenvironment and for maintenance of tumor growth [1]

  • We report that MIR126 induces autophagic flux in MM cells by downregulating insulin receptor substrate-1 (IRS1) and disrupting the IRS1 signaling pathway

  • We show that MIR126 overexpression was accompanied by accumulation of intracellular lipid droplets (LDs) in MM cells due to alteration of mitochondrial function in a hypoxia-inducible factor-1α (HIF1α)-dependent manner

Read more

Summary

Introduction

Metabolic reprogramming of cancer cells is essential for their adaptation to tumor microenvironment and for maintenance of tumor growth [1]. Autophagy is a catabolic pathway that has a fundamental role in this adaptation [2]. Recent implication of tumor suppressors like beclin-1 (BECN1) in autophagic pathways indicates a causative role for autophagy deficiencies in cancer formation. It has been documented that autophagy is strictly controlled to maintain homeostatic balance of energy metabolism www.impactjournals.com/oncotarget and turnover of proteins as well as cellular organelles [4,5]. This process is post-transcriptionally regulated by small non-coding microRNAs (MIRs) that regulate gene expression via complementary base-pairing with mRNAs [6]

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