Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 12(1): 7349, 2021 12 21.
Article in English | MEDLINE | ID: mdl-34934057

ABSTRACT

Neuroendocrine (NE) prostate cancer (NEPC) is a lethal subtype of castration-resistant prostate cancer (PCa) arising either de novo or from transdifferentiated prostate adenocarcinoma following androgen deprivation therapy (ADT). Extensive computational analysis has identified a high degree of association between the long noncoding RNA (lncRNA) H19 and NEPC, with the longest isoform highly expressed in NEPC. H19 regulates PCa lineage plasticity by driving a bidirectional cell identity of NE phenotype (H19 overexpression) or luminal phenotype (H19 knockdown). It contributes to treatment resistance, with the knockdown of H19 re-sensitizing PCa to ADT. It is also essential for the proliferation and invasion of NEPC. H19 levels are negatively regulated by androgen signaling via androgen receptor (AR). When androgen is absent SOX2 levels increase, driving H19 transcription and facilitating transdifferentiation. H19 facilitates the PRC2 complex in regulating methylation changes at H3K27me3/H3K4me3 histone sites of AR-driven and NEPC-related genes. Additionally, this lncRNA induces alterations in genome-wide DNA methylation on CpG sites, further regulating genes associated with the NEPC phenotype. Our clinical data identify H19 as a candidate diagnostic marker and predictive marker of NEPC with elevated H19 levels associated with an increased probability of biochemical recurrence and metastatic disease in patients receiving ADT. Here we report H19 as an early upstream regulator of cell fate, plasticity, and treatment resistance in NEPC that can reverse/transform cells to a treatable form of PCa once therapeutically deactivated.


Subject(s)
Carcinoma, Neuroendocrine/genetics , Carcinoma, Neuroendocrine/pathology , Cell Plasticity/genetics , Gene Expression Regulation, Neoplastic , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , RNA, Long Noncoding/metabolism , Androgen Antagonists/therapeutic use , Animals , Benzamides/pharmacology , Benzamides/therapeutic use , Biomarkers, Tumor/metabolism , Carcinoma, Neuroendocrine/diagnosis , Carcinoma, Neuroendocrine/drug therapy , Cell Line, Tumor , Cell Lineage/genetics , Cell Nucleus/metabolism , Cell Proliferation/genetics , Cohort Studies , DNA Methylation/genetics , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , Epigenesis, Genetic/drug effects , Genome, Human , Histones/metabolism , Humans , Male , Neoplasm Grading , Neoplasm Invasiveness , Neoplastic Stem Cells/metabolism , Nitriles/pharmacology , Nitriles/therapeutic use , Organoids/metabolism , Organoids/pathology , Phenylthiohydantoin/pharmacology , Phenylthiohydantoin/therapeutic use , Phylogeny , Polycomb Repressive Complex 2/metabolism , Promoter Regions, Genetic/genetics , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/drug therapy , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Long Noncoding/genetics , Receptors, Androgen/metabolism , SOXB1 Transcription Factors/metabolism , Transcription, Genetic/drug effects
2.
Gigascience ; 7(6)2018 06 01.
Article in English | MEDLINE | ID: mdl-29757368

ABSTRACT

Background: Treatment-induced neuroendocrine prostate cancer (tNEPC) is an aggressive variant of late-stage metastatic castrate-resistant prostate cancer that commonly arises through neuroendocrine transdifferentiation (NEtD). Treatment options are limited, ineffective, and, for most patients, result in death in less than a year. We previously developed a first-in-field patient-derived xenograft (PDX) model of NEtD. Longitudinal deep transcriptome profiling of this model enabled monitoring of dynamic transcriptional changes during NEtD and in the context of androgen deprivation. Long non-coding RNA (lncRNA) are implicated in cancer where they can control gene regulation. Until now, the expression of lncRNAs during NEtD and their clinical associations were unexplored. Results: We implemented a next-generation sequence analysis pipeline that can detect transcripts at low expression levels and built a genome-wide catalogue (n = 37,749) of lncRNAs. We applied this pipeline to 927 clinical samples and our high-fidelity NEtD model LTL331 and identified 821 lncRNAs in NEPC. Among these are 122 lncRNAs that robustly distinguish NEPC from prostate adenocarcinoma (AD) patient tumours. The highest expressed lncRNAs within this signature are H19, LINC00617, and SSTR5-AS1. Another 742 are associated with the NEtD process and fall into four distinct patterns of expression (NEtD lncRNA Class I, II, III, and IV) in our PDX model and clinical samples. Each class has significant (z-scores >2) and unique enrichment for transcription factor binding site (TFBS) motifs in their sequences. Enriched TFBS include (1) TP53 and BRN1 in Class I, (2) ELF5, SPIC, and HOXD1 in Class II, (3) SPDEF in Class III, (4) HSF1 and FOXA1 in Class IV, and (5) TWIST1 when merging Class III with IV. Common TFBS in all NEtD lncRNA were also identified and include E2F, REST, PAX5, PAX9, and STAF. Interrogation of the top deregulated candidates (n = 100) in radical prostatectomy adenocarcinoma samples with long-term follow-up (median 18 years) revealed significant clinicopathological associations. Specifically, we identified 25 that are associated with rapid metastasis following androgen deprivation therapy (ADT). Two of these lncRNAs (SSTR5-AS1 and LINC00514) stratified patients undergoing ADT based on patient outcome. Discussion: To date, a comprehensive characterization of the dynamic landscape of lncRNAs during the NEtD process has not been performed. A temporal analysis of the PDX-based NEtD model has for the first time provided this dynamic landscape. TFBS analysis identified NEPC-related TF motifs present within the NEtD lncRNA sequences, suggesting functional roles for these lncRNAs in NEPC pathogenesis. Furthermore, select NEtD lncRNAs appear to be associated with metastasis and patients receiving ADT. Treatment-related metastasis is a clinical consequence of NEPC tumours. Top candidate lncRNAs FENDRR, H19, LINC00514, LINC00617, and SSTR5-AS1 identified in this study are implicated in the development of NEPC. We present here for the first time a genome-wide catalogue of NEtD lncRNAs that characterize the transdifferentiation process and a robust NEPC lncRNA patient expression signature. To accomplish this, we carried out the largest integrative study that applied a PDX NEtD model to clinical samples. These NEtD and NEPC lncRNAs are strong candidates for clinical biomarkers and therapeutic targets and warrant further investigation.


Subject(s)
Neuroendocrine Tumors/genetics , Prostatic Neoplasms/genetics , RNA, Long Noncoding/genetics , Animals , Binding Sites , Cell Transdifferentiation/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Male , Mice , Neoplasm Metastasis , Neuroendocrine Tumors/pathology , Nucleotide Motifs/genetics , Phenotype , Prostatic Neoplasms/pathology , RNA, Long Noncoding/metabolism , Transcription Factors/metabolism , Transcriptome/genetics , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...