Skip to main content

Lund University Publications

LUND UNIVERSITY LIBRARIES

Proteogenomic characterization of patient-derived xenografts highlights the role of REST in neuroendocrine differentiation of castration-resistant prostate cancer

Flores-Morales, Amilcar ; Bergmann, Tobias B. ; Lavallee, Charlotte ; Batth, Tanveer S. ; Lin, Dong ; Lerdrup, Mads ; Friis, Stine ; Bartels, Anette ; Kristensen, Gitte and Krzyzanowska, Agnieszka LU , et al. (2019) In Clinical Cancer Research 25(2). p.595-608
Abstract

Purpose: An increasing number of castration-resistant prostate cancer (CRPC) tumors exhibit neuroendocrine (NE) features. NE prostate cancer (NEPC) has poor prognosis, and its development is poorly understood. Experimental Design: We applied mass spectrometry–based proteomics to a unique set of 17 prostate cancer patient–derived xenografts (PDX) to characterize the effects of castration in vivo, and the proteome differences between NEPC and prostate adenocarcinomas. Genome-wide profiling of REST-occupied regions in prostate cancer cells was correlated to the expression changes in vivo to investigate the role of the transcriptional repressor REST in castration-induced NEPC differentiation. Results: An average of 4,881 proteins were... (More)

Purpose: An increasing number of castration-resistant prostate cancer (CRPC) tumors exhibit neuroendocrine (NE) features. NE prostate cancer (NEPC) has poor prognosis, and its development is poorly understood. Experimental Design: We applied mass spectrometry–based proteomics to a unique set of 17 prostate cancer patient–derived xenografts (PDX) to characterize the effects of castration in vivo, and the proteome differences between NEPC and prostate adenocarcinomas. Genome-wide profiling of REST-occupied regions in prostate cancer cells was correlated to the expression changes in vivo to investigate the role of the transcriptional repressor REST in castration-induced NEPC differentiation. Results: An average of 4,881 proteins were identified and quantified from each PDX. Proteins related to neurogenesis, cell-cycle regulation, and DNA repair were found upregulated and elevated in NEPC, while the reduced levels of proteins involved in mitochondrial functions suggested a prevalent glycolytic metabolism of NEPC tumors. Integration of the REST chromatin bound regions with expression changes indicated a direct role of REST in regulating neuronal gene expression in prostate cancer cells. Mechanistically, depletion of REST led to cell-cycle arrest in G1, which could be rescued by p53 knockdown. Finally, the expression of the REST-regulated gene secretagogin (SCGN) correlated with an increased risk of suffering disease relapse after radical prostatectomy. Conclusions: This study presents the first deep characterization of the proteome of NEPC and suggests that concomitant inhibition of REST and the p53 pathway would promote NEPC. We also identify SCGN as a novel prognostic marker in prostate cancer.

(Less)
Please use this url to cite or link to this publication:
author
; ; ; ; ; ; ; ; and , et al. (More)
; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; and (Less)
organization
publishing date
type
Contribution to journal
publication status
published
subject
in
Clinical Cancer Research
volume
25
issue
2
pages
14 pages
publisher
American Association for Cancer Research
external identifiers
  • pmid:30274982
  • scopus:85060009188
ISSN
1078-0432
DOI
10.1158/1078-0432.CCR-18-0729
language
English
LU publication?
yes
id
238e540e-0579-498b-a525-a68b9ae67912
date added to LUP
2019-01-29 12:56:39
date last changed
2024-04-15 22:47:36
@article{238e540e-0579-498b-a525-a68b9ae67912,
  abstract     = {{<p>Purpose: An increasing number of castration-resistant prostate cancer (CRPC) tumors exhibit neuroendocrine (NE) features. NE prostate cancer (NEPC) has poor prognosis, and its development is poorly understood. Experimental Design: We applied mass spectrometry–based proteomics to a unique set of 17 prostate cancer patient–derived xenografts (PDX) to characterize the effects of castration in vivo, and the proteome differences between NEPC and prostate adenocarcinomas. Genome-wide profiling of REST-occupied regions in prostate cancer cells was correlated to the expression changes in vivo to investigate the role of the transcriptional repressor REST in castration-induced NEPC differentiation. Results: An average of 4,881 proteins were identified and quantified from each PDX. Proteins related to neurogenesis, cell-cycle regulation, and DNA repair were found upregulated and elevated in NEPC, while the reduced levels of proteins involved in mitochondrial functions suggested a prevalent glycolytic metabolism of NEPC tumors. Integration of the REST chromatin bound regions with expression changes indicated a direct role of REST in regulating neuronal gene expression in prostate cancer cells. Mechanistically, depletion of REST led to cell-cycle arrest in G<sub>1</sub>, which could be rescued by p53 knockdown. Finally, the expression of the REST-regulated gene secretagogin (SCGN) correlated with an increased risk of suffering disease relapse after radical prostatectomy. Conclusions: This study presents the first deep characterization of the proteome of NEPC and suggests that concomitant inhibition of REST and the p53 pathway would promote NEPC. We also identify SCGN as a novel prognostic marker in prostate cancer.</p>}},
  author       = {{Flores-Morales, Amilcar and Bergmann, Tobias B. and Lavallee, Charlotte and Batth, Tanveer S. and Lin, Dong and Lerdrup, Mads and Friis, Stine and Bartels, Anette and Kristensen, Gitte and Krzyzanowska, Agnieszka and Xue, Hui and Fazli, Ladan and Hansen, Klaus H. and Røder, Martin A. and Brasso, Klaus and Moreira, Jose M. and Bjartell, Anders and Wang, Yuzhuo and Olsen, Jesper V. and Collins, Colin C. and Iglesias-Gato, Diego}},
  issn         = {{1078-0432}},
  language     = {{eng}},
  number       = {{2}},
  pages        = {{595--608}},
  publisher    = {{American Association for Cancer Research}},
  series       = {{Clinical Cancer Research}},
  title        = {{Proteogenomic characterization of patient-derived xenografts highlights the role of REST in neuroendocrine differentiation of castration-resistant prostate cancer}},
  url          = {{http://dx.doi.org/10.1158/1078-0432.CCR-18-0729}},
  doi          = {{10.1158/1078-0432.CCR-18-0729}},
  volume       = {{25}},
  year         = {{2019}},
}