Heterochromatin protein 1α mediates development and aggressiveness of neuroendocrine prostate cancer

X Ci, J Hao, X Dong, SY Choi, H Xue, R Wu, S Qu… - Cancer research, 2018 - AACR
X Ci, J Hao, X Dong, SY Choi, H Xue, R Wu, S Qu, PW Gout, F Zhang, AM Haegert, L Fazli
Cancer research, 2018AACR
Neuroendocrine prostate cancer (NEPC) is a lethal subtype of prostate cancer arising mostly
from adenocarcinoma via neuroendocrine transdifferentiation following androgen
deprivation therapy. Mechanisms contributing to both NEPC development and its
aggressiveness remain elusive. In light of the fact that hyperchromatic nuclei are a
distinguishing histopathologic feature of NEPC, we utilized transcriptomic analyses of our
patient-derived xenograft (PDX) models, multiple clinical cohorts, and genetically …
Abstract
Neuroendocrine prostate cancer (NEPC) is a lethal subtype of prostate cancer arising mostly from adenocarcinoma via neuroendocrine transdifferentiation following androgen deprivation therapy. Mechanisms contributing to both NEPC development and its aggressiveness remain elusive. In light of the fact that hyperchromatic nuclei are a distinguishing histopathologic feature of NEPC, we utilized transcriptomic analyses of our patient-derived xenograft (PDX) models, multiple clinical cohorts, and genetically engineered mouse models to identify 36 heterochromatin-related genes that are significantly enriched in NEPC. Longitudinal analysis using our unique, first-in-field PDX model of adenocarcinoma-to-NEPC transdifferentiation revealed that, among those 36 heterochromatin-related genes, heterochromatin protein 1α (HP1α) expression increased early and steadily during NEPC development and remained elevated in the developed NEPC tumor. Its elevated expression was further confirmed in multiple PDX and clinical NEPC samples. HP1α knockdown in the NCI-H660 NEPC cell line inhibited proliferation, ablated colony formation, and induced apoptotic cell death, ultimately leading to tumor growth arrest. Its ectopic expression significantly promoted NE transdifferentiation in adenocarcinoma cells subjected to androgen deprivation treatment. Mechanistically, HP1α reduced expression of androgen receptor and RE1 silencing transcription factor and enriched the repressive trimethylated histone H3 at Lys9 mark on their respective gene promoters. These observations indicate a novel mechanism underlying NEPC development mediated by abnormally expressed heterochromatin genes, with HP1α as an early functional mediator and a potential therapeutic target for NEPC prevention and management.
Significance: Heterochromatin proteins play a fundamental role in NEPC, illuminating new therapeutic targets for this aggressive disease. Cancer Res; 78(10); 2691–704. ©2018 AACR.
AACR