Abstract

The vacuolar H+-adenosine triphosphatase (ATPase) subunit V0C (ATP6V0C), a proton-conducting, pore-forming subunit of vacuolar ATPase, maintains pH homeostasis and induces organelle acidification. The intracellular and extracellular pH of cancer cells affects their growth; however, the role of ATP6V0C in highly invasive esophageal cancer cells (ECCs) remains unclear. In this study, we examined the role of ATP6V0C in glucose metabolism in ECCs. The ATP6V0C depletion attenuated ECC proliferation, invasion, and suppressed glucose metabolism, as indicated by reduced glucose uptake and decreased lactate and adenosine triphosphate (ATP) production in cells. Consistent with this, expression of glycolytic enzyme and the extracellular acidification rate (ECAR) were also decreased by ATP6V0C knockdown. Mechanistically, ATP6V0C interacted with pyruvate kinase isoform M2 (PKM2), a key regulator of glycolysis in ECCs. The ATP6V0C depletion reduced PKM2 phosphorylation at tyrosine residue 105 (Tyr105), leading to inhibition of nuclear translocation of PKM2. In addition, ATP6V0C was recruited at hypoxia response element (HRE) sites in the lactate dehydrogenase A (LDHA) gene for glycolysis. Thus, our data suggest that ATP6V0C enhances aerobic glycolysis and motility in ECCs.

Highlights

  • IntroductionIn the majority of cases, esophageal cancer is a squamous cell carcinoma and carries a poor prognosis due to the late diagnosis after metastasis [1]

  • Esophageal cancer is one of the most lethal malignancies in the world [1]

  • The pyruvate kinase isoform M2 (PKM2) interacts with hypoxia inducible factor 1 (HIF-1), leading to an increase in aerobic glycolysis; we examined whether ATP6V0C interacts with

Read more

Summary

Introduction

In the majority of cases, esophageal cancer is a squamous cell carcinoma and carries a poor prognosis due to the late diagnosis after metastasis [1]. Despite recent advances in diagnosis and treatment using chemotherapy or radiation, the five-year survival rate in esophageal squamous cell carcinoma (ESCC) patients is low (up to 30–45%) because of high mortality [2]. Aerobic glycolysis is inefficient in generating ATP, cancer cells compensate for this inefficiency by increasing glucose uptake and lactate production [5]. Consistent with this result, positron emission tomography scans with

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