[HTML][HTML] RABL6A inhibits tumor-suppressive PP2A/AKT signaling to drive pancreatic neuroendocrine tumor growth

S Umesalma, CA Kaemmer… - The Journal of …, 2019 - Am Soc Clin Investig
S Umesalma, CA Kaemmer, JL Kohlmeyer, B Letney, AM Schab, JA Reilly, RM Sheehy…
The Journal of clinical investigation, 2019Am Soc Clin Investig
Hyperactivated AKT/mTOR signaling is a hallmark of pancreatic neuroendocrine tumors
(PNETs). Drugs targeting this pathway are used clinically, but tumor resistance invariably
develops. A better understanding of factors regulating AKT/mTOR signaling and PNET
pathogenesis is needed to improve current therapies. We discovered that RABL6A, a new
oncogenic driver of PNET proliferation, is required for AKT activity. Silencing RABL6A
caused PNET cell-cycle arrest that coincided with selective loss of AKT-S473 (not T308) …
Hyperactivated AKT/mTOR signaling is a hallmark of pancreatic neuroendocrine tumors (PNETs). Drugs targeting this pathway are used clinically, but tumor resistance invariably develops. A better understanding of factors regulating AKT/mTOR signaling and PNET pathogenesis is needed to improve current therapies. We discovered that RABL6A, a new oncogenic driver of PNET proliferation, is required for AKT activity. Silencing RABL6A caused PNET cell-cycle arrest that coincided with selective loss of AKT-S473 (not T308) phosphorylation and AKT/mTOR inactivation. Restoration of AKT phosphorylation rescued the G1 phase block triggered by RABL6A silencing. Mechanistically, loss of AKT-S473 phosphorylation in RABL6A-depleted cells was the result of increased protein phosphatase 2A (PP2A) activity. Inhibition of PP2A restored phosphorylation of AKT-S473 in RABL6A-depleted cells, whereas PP2A reactivation using a specific small-molecule activator of PP2A (SMAP) abolished that phosphorylation. Moreover, SMAP treatment effectively killed PNET cells in a RABL6A-dependent manner and suppressed PNET growth in vivo. The present work identifies RABL6A as a new inhibitor of the PP2A tumor suppressor and an essential activator of AKT in PNET cells. Our findings offer what we believe is a novel strategy of PP2A reactivation for treatment of PNETs as well as other human cancers driven by RABL6A overexpression and PP2A inactivation.
The Journal of Clinical Investigation