BRN4 Is a Novel Driver of Neuroendocrine Differentiation in Castration-Resistant Prostate Cancer and Is Selectively Released in Extracellular Vesicles with BRN2

Clin Cancer Res. 2019 Nov 1;25(21):6532-6545. doi: 10.1158/1078-0432.CCR-19-0498. Epub 2019 Aug 1.

Abstract

Purpose: Neuroendocrine prostate cancer (NEPC), an aggressive variant of castration-resistant prostate cancer (CRPC), often emerges after androgen receptor-targeted therapies such as enzalutamide or de novo, via trans-differentiation process of neuroendocrine differentiation. The mechanistic basis of neuroendocrine differentiation is poorly understood, contributing to lack of effective predictive biomarkers and late disease recognition. The purpose of this study was to examine the role of novel proneural Pit-Oct-Unc-domain transcription factors (TF) in NEPC and examine their potential as noninvasive predictive biomarkers.Experimental Design: Prostate cancer patient-derived xenograft models, clinical samples, and cellular neuroendocrine differentiation models were employed to determine the expression of TFs BRN1 and BRN4. BRN4 levels were modulated in prostate cancer cell lines followed by functional assays. Furthermore, extracellular vesicles (EV) were isolated from patient samples and cell culture models, characterized by nanoparticle tracking analyses, Western blotting, and real-time PCR.

Results: We identify for the first time that: (i) BRN4 is amplified and overexpressed in NEPC clinical samples and that BRN4 overexpression drives neuroendocrine differentiation via its interplay with BRN2, a TF that was previously implicated in NEPC; (ii) BRN4 and BRN2 mRNA are actively released in prostate cancer EVs upon neuroendocrine differentiation induction; and (iii) enzalutamide treatment augments release of BRN4 and BRN2 in prostate cancer EVs, promoting neuroendocrine differentiation induction.

Conclusions: Our study identifies a novel TF that drives NEPC and suggests that as adaptive mechanism to enzalutamide treatment, prostate cancer cells express and secrete BRN4 and BRN2 in EVs that drive oncogenic reprogramming of prostate cancer cells to NEPC. Importantly, EV-associated BRN4 and BRN2 are potential novel noninvasive biomarkers to predict neuroendocrine differentiation in CRPC.

Publication types

  • Research Support, N.I.H., Extramural
  • Research Support, U.S. Gov't, Non-P.H.S.

MeSH terms

  • Aged
  • Aged, 80 and over
  • Animals
  • Carcinogenesis / genetics
  • Carcinoma, Neuroendocrine / genetics*
  • Carcinoma, Neuroendocrine / pathology
  • Cell Differentiation / genetics
  • Cell Line, Tumor
  • Disease Progression
  • Extracellular Vesicles / genetics
  • Gene Expression Regulation, Neoplastic / genetics
  • Heterografts
  • Homeodomain Proteins / genetics*
  • Humans
  • Male
  • Mice
  • Middle Aged
  • POU Domain Factors / genetics*
  • Prostate / metabolism
  • Prostate / pathology
  • Prostatic Neoplasms, Castration-Resistant / epidemiology
  • Prostatic Neoplasms, Castration-Resistant / genetics*
  • Prostatic Neoplasms, Castration-Resistant / pathology
  • Receptors, Androgen / genetics

Substances

  • AR protein, human
  • Homeodomain Proteins
  • POU Domain Factors
  • POU3F4 protein, human
  • Receptors, Androgen
  • transcription factor Brn-2