American Association for Cancer Research
Browse
00085472can170686-sup-180105_2_supp_4276710_xwhx7x.png (288.51 kB)

Figure S1 from Loss of FOXO1 Cooperates with TMPRSS2–ERG Overexpression to Promote Prostate Tumorigenesis and Cell Invasion

Download (288.51 kB)
figure
posted on 2023-03-31, 01:02 authored by Yinhui Yang, Alexandra M. Blee, Dejie Wang, Jian An, Yunqian Pan, Yuqian Yan, Tao Ma, Yundong He, Joseph Dugdale, Xiaonan Hou, Jun Zhang, S. John Weroha, Wei-Guo Zhu, Y. Alan Wang, Ronald A. DePinho, Wanhai Xu, Haojie Huang

Supplementary Figure S1. In panel A, ERG pull-down in HUVEC cells showing no interaction between FOXO1 and ERG. In contrast, AR binding with ERG is detected as expected. In panel B, bioinformatic analysis of 492 prostate cancer cases from TCGA to show the profiles of FOXO1, PTEN and ERG alterations. PTEN and FOXO1 deletions largely do not overlap.

Funding

NIH

DOD

Natural Science Foundation of China

Natural Science Foundation of Heilongjiang Province

History

ARTICLE ABSTRACT

E26 transformation-specific transcription factor ERG is aberrantly overexpressed in approximately 50% of all human prostate cancer due to TMPRSS2-ERG gene rearrangements. However, mice with prostate-specific transgenic expression of prostate cancer–associated ERG alone fail to develop prostate cancer, highlighting that ERG requires other lesions to drive prostate tumorigenesis. Forkhead box (FOXO) transcription factor FOXO1 is a tumor suppressor that is frequently inactivated in human prostate cancer. Here, we demonstrate that FOXO1, but not other FOXO proteins (FOXO3 and FOXO4), binds and inhibits the transcriptional activity of prostate cancer–associated ERG independently of FOXO1 transcriptional activity. Knockdown of endogenous FOXO1 increased invasion of TMPRSS2–ERG fusion–positive VCaP cells, an effect completely abolished by ERG knockdown. Patient specimen analysis demonstrated that FOXO1 and ERG protein expression inversely correlated in a subset of human prostate cancer. Although human ERG transgene expression or homozygous deletion of Foxo1 alone in the mouse prostate failed to promote tumorigenesis, concomitant ERG transgene expression and Foxo1 deletion resulted in upregulation of ERG target genes, increased cell proliferation, and formation of high-grade prostatic intraepithelial neoplasia. Overall, we provide biochemical and genetic evidence that aberrantly activated ERG cooperates with FOXO1 deficiency to promote prostate tumorigenesis and cell invasion. Our findings enhance understanding of prostate cancer etiology and suggest that the FOXO1–ERG signaling axis can be a potential target for treatment of prostate cancer. Cancer Res; 77(23); 6524–37. ©2017 AACR.

Usage metrics

    Cancer Research

    Licence

    Exports

    RefWorks
    BibTeX
    Ref. manager
    Endnote
    DataCite
    NLM
    DC