Loss of FOXP3 and TSC1 accelerates prostate cancer progression through synergistic transcriptional and posttranslational regulation of c-MYC

L Wu, B Yi, S Wei, D Rao, Y He, G Naik, S Bae, XM Liu… - Cancer research, 2019 - AACR
L Wu, B Yi, S Wei, D Rao, Y He, G Naik, S Bae, XM Liu, WH Yang, G Sonpavde, R Liu…
Cancer research, 2019AACR
Although c-MYC and mTOR are frequently activated proteins in prostate cancer, any
interaction between the two is largely untested. Here, we characterize the functional cross-
talk between FOXP3–c-MYC and TSC1–mTOR signaling during tumor progression. Deletion
of Tsc1 in mouse embryonic fibroblasts (MEF) decreased phosphorylation of c-MYC at
threonine 58 (pT58) and increased phosphorylation at serine 62 (pS62), an observation
validated in prostate cancer cells. Conversely, inhibition of mTOR increased pT58 but …
Abstract
Although c-MYC and mTOR are frequently activated proteins in prostate cancer, any interaction between the two is largely untested. Here, we characterize the functional cross-talk between FOXP3–c-MYC and TSC1–mTOR signaling during tumor progression. Deletion of Tsc1 in mouse embryonic fibroblasts (MEF) decreased phosphorylation of c-MYC at threonine 58 (pT58) and increased phosphorylation at serine 62 (pS62), an observation validated in prostate cancer cells. Conversely, inhibition of mTOR increased pT58 but decreased pS62. Loss of both FOXP3 and TSC1 in prostate cancer cells synergistically enhanced c-MYC expression via regulation of c-Myc transcription and protein phosphorylation. This crosstalk between FOXP3 and TSC1 appeared to be mediated by both the mTOR–4EBP1–c-MYC and FOXP3–c-MYC pathways. In mice, Tsc1 and Foxp3 double deletions in the prostate led to prostate carcinomas at an early age; this did not occur in these mice with an added c-Myc deletion. In addition, we observed synergistic antitumor effects of cotreating mice with inhibitors of mTOR and c-MYC in prostate cancer cells and in Foxp3 and Tsc1 double-mutant mice. In human prostate cancer, loss of nuclear FOXP3 is often accompanied by low expression of TSC1. Because loss of FOXP3 transcriptionally induces c-Myc expression and loss of TSC1 activates mTOR signaling, these data suggest cross-talk between FOXP3–c-MYC and TSC1–mTOR signaling that converges on c-MYC to regulate tumor progression. Coadministration of c-MYC and mTOR inhibitors may overcome the resistance to mTOR inhibition commonly observed in prostate cancer cells.
Significance
These results establish the principle of a synergistic action of TSC1 and FOXP3 during prostate cancer progression and provide new therapeutic targets for patients who have prostate cancer with two signaling defects.
AACR