Communications Biology (Jan 2024)

DPYSL5 is highly expressed in treatment-induced neuroendocrine prostate cancer and promotes lineage plasticity via EZH2/PRC2

  • Roosa Kaarijärvi,
  • Heidi Kaljunen,
  • Lucia Nappi,
  • Ladan Fazli,
  • Sonia H. Y. Kung,
  • Jaana M. Hartikainen,
  • Ville Paakinaho,
  • Janne Capra,
  • Kirsi Rilla,
  • Marjo Malinen,
  • Petri I. Mäkinen,
  • Seppo Ylä-Herttuala,
  • Amina Zoubeidi,
  • Yuzhuo Wang,
  • Martin E. Gleave,
  • Mikko Hiltunen,
  • Kirsi Ketola

DOI
https://doi.org/10.1038/s42003-023-05741-x
Journal volume & issue
Vol. 7, no. 1
pp. 1 – 13

Abstract

Read online

Abstract Treatment-induced neuroendocrine prostate cancer (t-NEPC) is a lethal subtype of castration-resistant prostate cancer resistant to androgen receptor (AR) inhibitors. Our study unveils that AR suppresses the neuronal development protein dihydropyrimidinase-related protein 5 (DPYSL5), providing a mechanism for neuroendocrine transformation under androgen deprivation therapy. Our unique CRPC-NEPC cohort, comprising 135 patient tumor samples, including 55 t-NEPC patient samples, exhibits a high expression of DPYSL5 in t-NEPC patient tumors. DPYSL5 correlates with neuroendocrine-related markers and inversely with AR and PSA. DPYSL5 overexpression in prostate cancer cells induces a neuron-like phenotype, enhances invasion, proliferation, and upregulates stemness and neuroendocrine-related markers. Mechanistically, DPYSL5 promotes prostate cancer cell plasticity via EZH2-mediated PRC2 activation. Depletion of DPYSL5 decreases proliferation, induces G1 phase cell cycle arrest, reverses neuroendocrine phenotype, and upregulates luminal genes. In conclusion, DPYSL5 plays a critical role in regulating prostate cancer cell plasticity, and we propose the AR/DPYSL5/EZH2/PRC2 axis as a driver of t-NEPC progression.