Abstract

Loss of hepatic fructose-1, 6-bisphosphate aldolase B (Aldob) leads to a paradoxical up-regulation of glucose metabolism to favor hepatocellular carcinogenesis (HCC), but the upstream signaling events remain poorly defined. Akt is highly activated in HCC, and targeting Akt is being explored as a potential therapy for HCC. Herein, we demonstrate that Aldob suppresses Akt activity and tumor growth through a protein complex containing Aldob, Akt, and protein phosphatase 2A (PP2A), leading to inhibition of cell viability, cell cycle progression, glucose uptake, and metabolism. Interestingly, Aldob directly interacts with phosphorylated Akt (p-Akt) and promotes the recruitment of PP2A to dephosphorylate p-Akt, and this scaffolding effect of Aldob is independent of its enzymatic activity. Loss of Aldob or disruption of Aldob/Akt interaction in Aldob R304A mutant restores Akt activity and tumor-promoting effects. Consistently, Aldob and p-Akt expression are inversely correlated in human HCC tissues, and Aldob down-regulation coupled with p-Akt up-regulation predicts a poor prognosis for HCC. We have further discovered that Akt inhibition or a specific small-molecule activator of PP2A (SMAP) efficiently attenuates HCC tumorigenesis in xenograft mouse models. Our work reveals a novel nonenzymatic role of Aldob in negative regulation of Akt activation, suggesting that directly inhibiting Akt activity or through reactivating PP2A may be a potential therapeutic approach for HCC treatment.

Highlights

  • Hepatocellular carcinoma (HCC) is the most common primary liver malignancy and the fourth leading cause of cancer-related deaths worldwide [1]

  • Our study uncovers a novel inverse correlation between aldolase B (Aldob) and phosphorylated Akt (p-Akt) expression in human HCC and low Aldob expression with high p-Akt predicts the worst prognosis for HCC patients, suggesting an important role of Akt signaling in HCC in the context of Aldob downregulation

  • This tumor promoting effect due to the loss of Aldob is achieved by releasing the inhibition on glucose-6-phosphate dehydrogenase (G6PD) and phosphate pathway (PPP) metabolism as a result of destabilizing Aldob/G6PD/p53 protein complex [18]

Read more

Summary

Introduction

Hepatocellular carcinoma (HCC) is the most common primary liver malignancy and the fourth leading cause of cancer-related deaths worldwide [1]. Abnormal activation of the PI3K/Akt signaling pathway is a hallmark of many human malignancies, including HCC [3]. Activated Akt triggers the subsequent cellular response through the phosphorylation of various downstream substrates or induction of multiple gene expressions. Inhibitory phosphorylation of glycogen synthase kinase 3β (GSK3β) by active Akt leads to increased stability and accumulation of CyclinD1, thereby promoting cell cycle progression [5]. A delicate balance between protein kinase-catalyzed phosphorylation and protein phosphatase-mediated dephosphorylation is vital for Akt kinase activity in cellular homeostasis; dysregulation of this balance may lead to tumorigenesis. The upstream signaling networks involved in the regulation of Akt activity remain to be fully characterized

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