SlideShare a Scribd company logo
1 of 9
Download to read offline
The emerging role of noncoding RNA in prostate cancer
progression and its implication on diagnosis and
treatment
Ken-ichi Takay , MuungoLungwani-Tysonama and Satoshi Inoue
Corresponding author: Satoshi Inoue, Bunkyo-ku, Tokyo 113-8655, Japan. E-mail: INOUE-GER@h.u-tokyo.ac.jp
Abstract
Recent transcriptome studies using next-generation sequencing have detected aberrant changes in the expression of non-
coding RNAs (ncRNAs) associated with cancer. For prostate cancer, the expression levels of ncRNAs including microRNAs
and long noncoding RNAs are strongly associated with diagnosis, carcinogenesis and tumor growth. Moreover, androgen
and its cognate receptor, androgen receptor (AR), regulate various signaling pathways for prostate tumor growth. In add-
ition, progression to lethal castration-resistant prostate cancer (CRPC) is also owing to AR function. Systematic analysis of
AR-binding sites and their regulated transcripts revealed that many ncRNAs are widely regulated at the transcriptional
level. Thus, recent studies provide new insight into the complicated molecular mechanism of prostate cancer progression.
This review focused on the role of various ncRNAs in prostate cancer and the association between their expression and
CRPC.
Key words: CRPC; AR; cap analysis of gene expression (CAGE); miRNA; lncRNA; epigenome
The role of noncoding RNAs in prostate cancer
biology
Recent advances in transcriptome technology have revealed
that >90% of the human genome is actively transcribed [1]. The
encyclopedia of DNA elements (ENCODE) project has shown
that only 2% of these transcripts are translated into proteins [2].
The noncoding RNAs (ncRNAs), which occupy the majority of
transcripts in the nucleus, were initially thought as the ‘dark
matter’. Generally, ncRNAs are broadly divided into short
(<200 nt) and long (>200 nt) transcripts [3]. In this review, we
summarize the function and clinical significance of long non-
coding RNAs (lncRNAs) and microRNAs (miRNAs) in prostate
cancer and their regulation by the androgen receptor (AR).
miRNAs play important roles in cancer by posttranscriptional
modification of target mRNA or protein expression. LncRNAs
represent most of the transcribed ncRNA in the human genome.
GENCODE v19 includes 13 870 human lncRNA-related genes,
which produce 23 898 lncRNAs [4]. These lncRNAs exhibit simi-
lar structure and biogenesis as mRNAs. They are polyadeny-
lated and may function in either nuclear or cytoplasmic
compartments. Evidence showing that lncRNAs are aberrantly
expressed in numerous human diseases including cancer sup-
ports their importance [5–7].
The function of AR in castration resistance
AR is a member of the nuclear receptor superfamily [8, 9] and is
a key molecule for androgen signaling in its target organ, the
prostate. Testosterone that is produced in the testes is the most
abundant circulating androgen (90%). Testosterone diffuses
into prostate cells and is converted to dihydrotestosterone
(DHT) by the enzyme 5a-reductase [10]. DHT directly binds to
and activates AR even more tightly than testosterone [11].
Before ligand binding, AR is found in the cytoplasm in a com-
plex that includes molecular chaperones and co-chaperones
from the heat shock protein (Hsp) family. After binding to an-
drogen, a conformational change in the complex leads to nu-
clear translocation of AR. In the nucleus, AR binds as a dimer to
specific genomic sequences called androgen-responsive elem-
ents (AREs), which are found in the promoter and enhancer
Ken-ichi Takayama is a researcher of Geriatric Medicine, Graduate School of Medicine, The University of Tokyo.
Satoshi Inoue is a professor of Anti-Aging Medicine, Graduate School of Medicine, The University of Tokyo.
VC The Author 2015. Published by Oxford University Press. All rights reserved. For permissions, please email: journals.permissions@oup.com
257
Briefings in Functional Genomics, 15(3), 2016, 257–265
doi: 10.1093/bfgp/elv057
Advance Access Publication Date: 17 December 2015
Review paper
Downloadedfromhttps://academic.oup.com/bfg/article-abstract/15/3/257/1742432bygueston10March2020
regions of target genes (Figure 1) [12]. AR regulates its enhancer
action by modifying the epigenetic condition of AR-binding sites
(ARBSs) [13].
AR also induces prostate cancer development and progres-
sion. Despite favorable responses to initial hormone therapy,
most patients progress to lethal castration-resistant prostate
cancer (CRPC) with elevated AR expression [14], hypersensitivity
to androgens [15], intra-tumoral steroidogenesis [16] and devel-
opment of AR variants [17]. Thus, identification of signaling
events downstream of AR is critical for understanding the pro-
gression of hormone-responsive prostate cancer to CRPC.
Although AR overexpression is commonly observed in CRPC,
the mechanism underlying altered AR expression is not com-
pletely understood. However, recent studies showed that both
transcriptional and epigenetic changes are important for AR
upregulation in prostate cancer. A recent study analyzing an an-
drogen-dependent prostate cancer cell line identified ARBSs in
the introns of the AR gene [18]. Androgen treatment causes AR
to bind the enhancer and recruit lysine-specific demethylase 1
(LSD1), which represses transcription by inhibiting histone
H3K4 methylation, thus demonstrating a negative feedback
loop that limits endogenous AR expression. However, if cells are
incubated in castration levels of androgens, AR expression in-
creases. Furthermore, low levels of androgens in CRPC are suffi-
cient to activate AR target genes, but are insufficient to
suppress AR itself.
Identification of androgen-regulated mIRNAs
miRNAs are a class of naturally occurring, short noncoding
RNAs that negatively regulate gene expression by binding to the
30
untranslated region (UTR) of mRNAs and inhibiting their
translation. Recent research demonstrates that dysregulation of
miRNA expression profiles contributes to pathogenesis of
human malignancies. In prostate cancer, miRNAs regulated by
androgen and AR have been identified using short RNA se-
quence studies or miRNA microarray, and their functions for
prostate cancer progression have been investigated.
miR-21
miR-21 is an oncogene encoded at a fragile genomic location,
17q23.2, which is amplified in several types of tumors [19]. miR-
21 is an androgen-regulated miRNA and is induced by AR re-
cruitment to its promoter in the presence of androgens (Figure
1). Expression analysis studies have shown that miR-21 expres-
sion is associated with prostate cancer aggressiveness. Elevated
miR-21 expression promotes androgen-dependent cell growth
and castration-resistant tumor growth [20]. The target gene of
miR-21 is MRCKS (myristoylated alanine rich protein kinase C
substrate), which regulates cell motility, mitogenesis and
plasma membrane trafficking [21]. Other factors repressed by
miR-21 are B-cell translocation gene 2 (BTG2) and phosphatase
and tensin homolog (PTEN) [22, 23].
miR-125b
This miRNA is induced by androgen and stimulates the growth
of prostate cancer cells (Figure 1). Expression of miR-125b is
increased in prostate cancer samples compared with normal
controls and correlates with Gleason scores, thus demonstrat-
ing that miR-125b is an oncogenic miRNA that participates in
the development of CRPC [24]. miR-125b targets the 30
UTR of
apoptosis regulators such as p53, p53 downstream modulator of
apoptosis (PUMA) and BCL2-antagonist/killer 1 (BAK1). Thus,
dysregulation of miR-125b results in an imbalance between pro-
and anti-apoptotic processes by targeting these pathways.
Other androgen-regulated miRNAs (miR-141, -148a
and -32)
We have previously performed short RNA sequence analysis in
LNCaP cells and reported several androgen-regulated miRNAs
that promote cell proliferation including miR-141, miR-200a and
miR-148a [25]. miR-148a is highly expressed in prostate cancer
cell line derived from the lymph node (LNCaP) cells and targets
cell cycle regulator cullin-associated and neddylation-
dissociated 1 to promote cell proliferation (Figure 1). Expression
of miR148a is upregulated in prostate cancer samples compared
with normal tissues [26].
miR-32 is an androgen-regulated miRNA, which confers a
growth advantage as demonstrated by reduction of apoptosis,
and this effect is mediated by targeting BCL2 interacting media-
tor (BIM), a pro-apoptotic member of the BCL2-antagonist/killer
1 (BCL2) family [27, 28]. In addition, miR-32 directly targets the 30
UTR of BTG2 to induce cell proliferation. Interestingly, both
miR-32 and miR-148a are reported to be upregulated in CRPC
samples, suggesting roles for both miRNAs in the emergence of
castration resistance [27].
miR-29a/b
Because the expression of the miR-29 family is reduced in sev-
eral cancer tissues than in normal tissues, their role in cancer is
controversial [29, 30]. While the precise role of the miR-29 fam-
ily members in cancer is still unclear, they are known to pro-
mote metastasis in breast cancer [31, 32]. Upregulated miR-29a
enhances hepatoma migration by targeting PTEN [33], suggest-
ing that miR-29 family may exhibit a dual function as a tumor
Testosterone
ARE
AR
Activation of transcriptionNuclear
translocation
Pri-miRNA
Androgen responsive element
Mature miRNAs
Processing
miR-21, miR-141, miR-148a
miR-32, miR-125b, miR-29a/b
Cell membrane
DHT
Anti-
apoptosis
Anti-
apoptosis
Epigenetic
regulation
Epigenetic
regulation
Cell cycleCell cycle
Cell
invasion
Cell
invasion
Figure 1. AR functions as a nuclear receptor to regulate its target genes including
miRNAs. AR translocates to the nucleus and binds AREs. AR epigenetically acti-
vates transcription by recruiting coactivators for histone modifications. Primary
miRNAs are induced by AR binding to its promoters. These transcripts are
cleaved and processed to form mature miRNAs. Some miRNAs have been docu-
mented to be regulated by AR and promote tumor growth. (A colour version of
this figure is available online at: http://bfg.oxfordjournals.org)
258 | Takayama and Inoue
Downloadedfromhttps://academic.oup.com/bfg/article-abstract/15/3/257/1742432bygueston10March2020
suppressing and oncogenic miRNAs depending on cellular con-
text and disease progression.
We found that miR-29a/b is androgen-inducible miRNA tar-
geting ten-eleven translocation 2 (TET2) and is highly expressed
in cancers with poor prognoses. In vitro, miR-29 family members
promote cell proliferation and migration of hormone refractory
prostate cancer cells by activating cell motility and cell cycle-
associated gene expression. In vivo, we showed that miR-29
family members enhance tumor growth using several AR antag-
onists- and castration-resistant prostate cancer models. TET2
repression is associated with decreased 5-hydroxymethylation
(5-hmC) in prostate cancer progression. Decreased 5-hmC pro-
motes AR collaborating factor, forkhead box A1 (FOXA1) tran-
scriptional activity and prostate cancer-associated expression
of genes such as mTOR (Mammalian target of rapamycin). These
results indicate a significant oncogenic role of miR-29 in pros-
tate cancer progression by regulating epigenetic status [34].
AR-regulated miRNAs such as miR-21-125b-141 and -148a
regulates apoptosis, cell cycle and cell proliferation by repress-
ing various factors for tumor progression as AR downstream. In
addition, our research highlights the epigenetic regulation by
androgen-mediated miRNAs for activating FOXA1 and AR tran-
scriptional activity and prostate cancer progression. These find-
ings showed the important role of androgen-regulated miRNAs
as tumor-promoting factors in prostate cancer.
CRPC-associated mIRNAs
In addition to androgen-induced miRNAs, oncogenic miRNAs
for CRPC development have been reported. miR-221 and miR-
222 are transcribed from a cluster on chromosome X and are
upregulated in CRPC tissues and cell lines [35]. Although andro-
gen regulation of these miRNAs is unclear, miR-221 is reported
to be repressed by androgen treatment [36]. Ectopic expression
of miR-221 and -222 resulted in decreased sensitivity to andro-
gen and induced androgen-independent growth. Both miRNAs
target the expression of the important tumor suppressor gene
p27kip1 [37]. HECT domain containing E3 ubiquitin ligase 2
(HECTD2) and Ras-related protein (RAB1A) are other miR-221
target genes. HECTD2 affected AR signaling, and its downregula-
tion enhances androgen-independent cell growth [38]. These
findings suggest the important roles of miR-221/222 in cell cycle
regulation and AR activation for CRPC development. However,
the functions of miR-221/22 remain to be determined because
another study has shown the tumor-suppressive function of
miR-221 and -222 by targeting oncogene Ecm29 [39].
Tumor-suppressive miRNAs in prostate
cancer
Several studies have revealed the importance of miRNAs as
tumor-suppressive factors targeting oncogenic signaling.
Recent advances of genomic biology have identified important
roles of miRNAs repressed in prostate cancer development by
genomic loss.
miR-101
miR-101 is significantly reduced in prostate cancer, and its ex-
pression is further decreased during cancer progression because
the genomic locus encoding miR-101 is found to be significantly
lost in metastatic prostate cancers. Interestingly, loss of miR-
101 is associated with overexpression of enhancer of zeste
homolog 2 (EZH2), a polycomb group protein, whose primary
function is to trimethylate histone H3 lysine 27 (H3K27) at gene
promoters [40].
miR-15/16
miR-15 and -16 are tumor suppressors likely involved in pros-
tate cancer development at multiple levels. Both miRNAs are
located at a fragile chromosomal locus, 13q14, in which gen-
omic loss is reportedly associated with clinically advanced pros-
tate cancer [41]. Delivery of miR-15 and -16 suppressed tumor
formation from nontumorigenic cells in severe combined
immunodeficiency (SCID) mice [42]. These miRNAs control cell
survival, proliferation and invasion via deregulation of onco-
genes such as BCL2, cyclinD1 and WNT3A (wingless type MMTV
integration site family).
Thus, in addition to AR-mediated pathways, these tumor-
suppressive miRNAs possesses other critical roles for progres-
sion to metastatic or advanced prostate cancer.
Integrative analysis to discover androgen-
regulated lncRNAs
Next-generation sequencing has been used to analyze the tran-
scriptome of prostate cancer cells. Combined analyses of gen-
ome-wide ARBSs obtained by chromatin immunoprecipitation
(ChIP)-based analyses such as ChIP-chip [43–48] or ChIP-seq
[49–51] and the androgen-regulated transcriptome have re-
cently been reported. A new technique, global nuclear run-on
sequencing (GRO-seq), has been used to analyze sequential
gene expression on androgen treatment [52]. For nuclear run-on
reactions, cell nuclei are isolated after treatment with andro-
gens for a specific amount of time. In the run-on step, RNA poly-
merases are allowed to run on about 100 bases in the presence
of a ribonucleotide analog [5-bromouridine 50
-triphosphate
(BrUTP)]. BrU-containing RNA is selected by purification with an
antibody specific for the nucleotide analog. A cDNA library is
then prepared for next-generation sequencing. Using this
method, the authors discovered the production of enhancer-
templated noncoding RNAs (enhancer RNA, eRNAs).
Cap analysis of gene expression (CAGE) [53] is a high-
throughput method for analysis of gene expression and to pro-
file transcription start sites (TSS), including those for promoter
usage. CAGE is based on sequencing of concatamers of DNA
tags derived from the initial 20 nucleotides at the 50
ends of
mRNAs. The frequency of CAGE tags is consistent with results
from other analyses, such as microarrays. This analysis is high-
throughput, and enables understanding of gene networks via
correlation between promoter usage and expression of gene
transcription factors. We performed CAGE to determine andro-
gen-regulated TSSs and ChIP-chip analysis to identify genome-
wide ARBSs and histone H3 acetylated sites in the complete
human genome [54]. Using CAGE, we identified 13,110 distinct,
androgen-regulated TSSs. Cross-referencing with the gene ex-
pression database for prostate cancer (Oncomine), the majority
of androgen-upregulated genes containing adjacent ARBSs and
CAGE tag clusters in our study were previously confirmed as
upregulated genes in prostate cancer. The integrated high-
throughput genome analyses of CAGE and ChIP-chip provide
useful information for elucidating the AR-mediated transcrip-
tional network that contributes to the development and pro-
gression of prostate cancer. ncRNAs, including miRNAs, were
also identified as androgen target transcripts in this study. We
found many androgen-dependent TSSs widely distributed
throughout the genome, including in the antisense (AS)
Emerging role of noncoding RNA in prostate cancer progression | 259
Downloadedfromhttps://academic.oup.com/bfg/article-abstract/15/3/257/1742432bygueston10March2020
direction of RefSeq genes. Several pairs of sense/AS promoters
were newly identified within single RefSeq gene regions, sug-
gesting the involvement of AS ncRNA in transcriptional
regulation.
Functional analysis of as ncRNA in prostate
cancer
Global transcriptome analysis revealed that the majority of the
mammalian genome can generate transcripts from both strands
of the DNA double helix, and identified paired sense/AS tran-
scripts, indicating that AS transcription is important for
gene regulation [55–57]. One example of AS transcripts is the
cyclin-dependent kinase (CDKN) 2B antisense RNA1 (CDKN2B-AS1)
[58], which is a natural AS transcript located within the
CDKN2A/CDKN2B tumor suppressor locus on 9q21.3. Cycline
dependent kinase (CDK) inhibitors, p14, p15(CDKN2B) and
p16(CDKN2A) are produced from this locus and regulate cell
cycle progression at G1/S. CDKN2B is specifically silenced by
CDKN2B-AS1 through heterochromatin formation, by increasing
dimethylation of H3K9 and demethylation of H3K4 at the gene
promoter. Another study showed the coordinated upregulation
of ANRIL (antisense noncoding RNA in the INK4 locus) and chromo-
box7 (CBX7), a member of Polycomb repressive complex 1 in
prostate cancer. CBX7 interacts with ANRIL at the promoters of
CDKN2A/2B for histone H3K27 methylation [59].
There are two potential mechanisms of sense-transcript re-
pression: (1) post-transcriptional repression [X-inactive specific
transcript (XIST) repression by XIST antisense RNA (TSIX)] by
degradation [60], and (2) epigenetic regulation by recruitment of
transcription factors. The interaction of lncRNAs with chroma-
tin remodeling complexes induces heterochromatin formation
in specific loci, leading to reduced target gene expression
(Figure 2). For example, HOX Antisense Intergenic RNA (HOTAIR)
[61], a 2.2 kb ncRNA, is transcribed in the AS direction from the
HOXC gene (homeobox gene) cluster. HOTAIR functions in trans
by interacting with and by recruiting the polycomb repressive
complex 2 to the HOXD locus, resulting in transcriptional silenc-
ing across a 40 kb region. In addition, HOTAIR interacts with a
second histone modification complex, the LSD1/corepressor for
Rest (CoREST)/RE1 silencing transcription factor (REST) complex,
which coordinates targeting of polycomb repressive complex2
(PRC2) and LSD1 to chromatin for coupled histone H3K27
methylation and K4 demethylation [62]. HOTAIR expression is
associated with hormone-dependent cancer such as breast and
prostate cancer. HOTAIR expression level was higher in breast
tumors than in normal breast epithelia, and high HOTAIR ex-
pression is associated with poor prognosis of patients. This clin-
ical finding also reflects the HOTAIR-mediated regulation of
hormone action. Although the regulation of HOTAIR by estrogen
is controversial [63, 64], HOTAIR enhanced estrogen receptor
(ER) protein level and enhanced downstream signaling [64].
Moreover, HOTAIR is repressed by androgen and upregulated by
hormone deprivation. In addition, HOTAIR binds to AR and
blocked E3-ubiquitine ligase murine double minute 2 (MDM2) to
stabilize AR and enhance the AR-mediated transcriptional ac-
tivity and drives CRPC development [65].
We investigated the functional role of the novel androgen-
responsive AS RNA, CTBP1-AS [66]. We focused on the mechan-
ism of androgen-mediated cancer proliferation by this lncRNA
and the clinical significance in prostate cancer progression. We
showed that C-terminal binding protein 1 (CTBP1) [67] is a tran-
scriptional corepressor of AR and negatively regulates androgen
signaling. CTBP1-AS is directly upregulated by AR and interacts
with RNA binding protein, PSF [68, 69], for transcriptional re-
pression using HDACs. Downstream signals of CTBP1-AS/PSF
(polypyrimidine tract binding protein associated splicing factor)
complex include cell cycle regulators such as p53 or SMAD3 in
addition to CTBP1. Castration-resistant tumor growth was pro-
moted by androgen-mediated repression of these cell cycle
regulators and activation of AR signaling.
Activation of AR transcription activity by
lncRNAs
In addition to CTBP1-AS and HOTAIR, several lncRNAs have been
reported to regulate AR activity for prostate cancer progression.
Steroid receptor RNA activator
Steroid receptor RNA activator (SRA) interacts with nuclear re-
ceptors, including AR, ER, progesterone receptor, glucocorticoid
receptor (GR) and thyroid hormone receptor, and regulates their
transcriptional activities. As a mechanism for activation, SRA
interacts with a nuclear receptor coactivator SRC-1 (steroid re-
ceptor coactivator). Mutation analysis revealed that six stem-
loop motifs are important for coactivation by SRA [70, 71].
Aberrant SRA transcription was observed in various tumors. It is
upregulated in tumors such as breast cancer, ovarian cancer
and prostate cancer, compared with normal tissues. In prostate
cancer, SRA and its alternative splicing form, steroid receptor
RNA activator protein (SRAP), were shown to be required for ex-
pression of AR target genes in the presence of androgens [72,
73].
Epigenetic and transcriptional regulation
SWI/
SNF
Epigenetic control
PSF
Deacethylation
Chromatin remodeling
Enhancer action
eRNA
Promoter enhancer
Loop formation
CTBP1-AS
Post-transcriptional regulation
miRNA synthesis
Regulation of mRNA
AAAAA
SChLAP1 PCGEM1
PNCNR1
SRA
MALAT1
miR-675
H19
TGFBI
AR
Inhibition of
AR binding
GAS5
AR
HOTAIR
MDM2
Enhancement
of AR binding
Figure 2. Representative mechanisms of lncRNAs in promoting prostate cancer.
(1) Transcriptional regulation by epigenetic mechanisms (interaction with epi-
genetic factors or accelerating AR transcriptional activity by direct interaction)
and (2) posttranscriptional regulation (modulating the target mRNA stability or
AR protein) have been reported. (A colour version of this figure is available
online at: http://bfg.oxfordjournals.org)
260 | Takayama and Inoue
Downloadedfromhttps://academic.oup.com/bfg/article-abstract/15/3/257/1742432bygueston10March2020
Enhancer RNA
In AR signals, AR-activated enhancers marked by increased
eRNA are required for activation of genes in their vicinity. eRNA
knockdown disrupts ligand-induced promoter–enhancer inter-
action. As a mechanism of enhancer–promoter interaction,
eRNA interacts with a cohesion complex and this interaction is
required for its recruitment to enhancers [74].
Prostate cancer gene expression marker 1 and Prostate
cancer noncoding RNA 1
Prostate cancer gene expression marker 1 (PCGEM1) was originally
identified as a prostate tissue-specific long ncRNA [75]. It is
involved in inhibition of apoptosis by delaying p53 and p21 in-
duction in an androgen-dependent manner [76]. PCGEM1 is
overexpressed in at least half of all prostate tumors. LncRNA
prostate cancer noncoding RNA 1 (PRNCR1) was found by a single
nucleotide polymorphism (SNP) associated with prostate cancer
susceptibility. Two SNPs, rs1456315 and rs7463708 in the
chromosome 8q24, which is a gene desert region, were shown
to be most significantly associated with prostate cancer suscep-
tibility. PCGEM1 and PRNCR1 are both involved in AR-mediated
gene transcription [77]. Knockdown of these lncRNAs inhibits
AR target gene regulation in a DHT-dependent manner and is
associated with tumor growth in CRPC xenograft model.
Interactions of PCGEM1 and PRNCR1 with AR were confirmed by
RNA pull down and RNA immunoprecipitation assay assays. For
the mechanism of AR activation, PCGEM1 was shown to interact
with pygopus homolog2 (Pygo2), and PRNCR1 interacts with
DOT1-like histone H3 methyltransferase for AR methylation. By
3C-ChIP experiments, these two lncRNAs were found to be
required for AR-bound enhancer promoter loop formation.
However, this mechanism is still controversial because another
study failed to reproduce the interaction of AR with these two
lncRNAs [78].
Growth arrest-specific 5
Growth arrest-specific 5 (GAS5) interacts with GR and sup-
presses GR-mediated transcriptional activity. The interaction of
GAS5 and GR reduces GR binding to GR-responsive elements
(GREs). The binding element of GAS5 with GR contains GRE-like
sequences, suggesting a competing role of GAS5 with GRE DNA
for GR binding. GAS5 also binds to other nuclear receptors such
as AR. GAS5 was found to be downregulated in breast cancer
and prostate cancer and then inhibit cell proliferation [79].
However, GAS5 inhibition of ERa transcriptional activity has not
been found. Noncoding multiple small nuclear RNAs (snoRNAs)
encoded in introns of GAS5, particularly U50, showed tumor
suppressor characteristics in breast cancer [80].
Functional lncRNA extends the insight of regulatory mech-
anism of AR. The major mechanism is the direct interaction
with AR and subsequent modulation of cofactors (SRA), stability
of AR protein itself (HOTAIR), inhibition of genomic binding
(GAS5) and enhancer promoter loop forming (eRNA, PCGEM1
and PRNCR1). Regulation of co-repressor or co-activator by epi-
genetic action of androgen-regulated lncRNA (CTBP1-AS) would
be another mechanism to enhance AR activity in prostate can-
cer. However, the number of lncRNAs described in detail is
small. We have to investigate the tissue-, spatial-specific func-
tion of lncRNAs or the secondary structure or high-level struc-
ture to determine the interaction and recruitment to specific
genomic regions.
Molecular mechanism of lncRNAs in prostate
cancer
Some lncRNAs are not related to the function of AR, but play an
important role in prostate cancer progression. These lncRNAs
could serve as cancer markers for clinical diagnosis.
Metastasis-associated lung adenocarcinoma transcript 1
Metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) is
involved in pre-mRNA processing and regulates the alternative
splicing of pre-mRNA by modulating levels of serine/arginine
splicing factors. Although the mechanism of MALAT1 upregula-
tion in cancer cells is still unclear, multiple copy-number gains
at the locus and chromosomal translocations have been re-
ported [81]. In prostate cancer, MALAT1 overexpression is corre-
lated with poor prognosis and cancer progression [82]. The
regulation of MALAT1 by androgen has not been reported, and
MALAT1 is upregulated in primary breast cancer and inhibited
by estrogen treatment. MALAT1 knockdown inhibits breast can-
cer cell proliferation, suggesting the tumor-promoting effect of
MALAT1 in breast cancer [83, 84]. Thus, MALAT1 may play a crit-
ical role in hormone-dependent cancer development.
Prostate cancer-associated ncRNA transcript-1
Prostate cancer-associated ncRNA transcript-1 (PCAT-1) is a rela-
tively small intergenic ncRNA located in the 8q24 gene desert,
and it was identified through global transcriptomic sequencing
of prostate tumors [85]. PCAT-1 expression is inversely corre-
lated with expression of the EZH2, a histone methyltransferase
that encodes components of PRC2 and a marker for prostate
cancer progression; thus, PRC2 represses PCAT-1 expression.
PCAT-1 induces cell proliferation in vitro and has a predomin-
antly repressive effect on gene expression, most notably on the
expression of the tumor suppressor gene breast cancer 2
(BRCA2) [86]. PCAT-1 also interacts with the SUZ12 component
of PRC2.
Second chromosome locus associated with prostate 1
Second chromosome locus associated with prostate 1 (SChLAP1) is an
lncRNA transcribed from within an intergenic gene desert on
2q31.1. It was originally identified in an analysis of intergenic
lncRNAs that are selectively upregulated in aggressive prostate
cancer samples. SChLAP1 is highly expressed in 25% of prostate
tumors and shows increased expression in metastatic cancer
cells. SChLAP1 is involved in the regulation of the switch/su-
crose nonfermenting (SWI/SNF) complex (Figure 2). This com-
plex canonically controls transcription by using ATP hydrolysis
to remodel chromatin and physically mobilize nucleosomes,
particularly at the gene promoters. SChLAP1 co-immunoprecipi-
tates with SNF5 and prevents its genomic binding, thus antago-
nizing tumor-suppressive SWI/SNF-mediated gene regulation
[87]. Using another cohort of 1008 patients, SChLAP1 was identi-
fied and validated as the highest-ranked overexpressed gene in
cancer with metastatic progression [88]. These findings demon-
strated the usefulness of SChLAP1 as a biomarker of prostate
cancer progression. Thus, both PCAT1-1 and SChLAP1 can be
useful biomarkers for clinically detecting advanced prostate
cancer.
Emerging role of noncoding RNA in prostate cancer progression | 261
Downloadedfromhttps://academic.oup.com/bfg/article-abstract/15/3/257/1742432bygueston10March2020
Prostate cancer gene 3
Prostate cancer gene 3 (PCA3) is an lncRNA associated with pros-
tate cancer and is a potentially useful biomarker [89, 90]. It was
originally discovered in 1999 by a differential display analysis of
prostate tissues and cell lines. Its expression in 95% of the pros-
tate tumors is up to 100-fold higher than that in adjacent
non-neoplastic tissues. PCA3 regulates the expression of its
overlapping gene PRUNE2, a tumor suppressor gene in prostate
cancer, by posttranscriptionally regulating the stability of
mRNA [91]. Urinary measurement of PCA3 RNA levels can be
helpful to detect prostate cancer with superior tumor specificity
to prostate-specific antigen [92, 93].
H19
This lncRNA is transcribed from the H19/Igf2 gene cluster on
chromosome 11p11.5. Some evidence suggests H19 plays a crit-
ical role in tumor progression by exhibiting oncogenic activity
in some types of tumors, such as bladder cancer. However, H19
was shown to have a tumor-suppressive role in metastatic pros-
tate cancer by repression of TGFb1 signals, which is involved in
metastasis. H19 and miR-675, derived from H19, were downre-
gulated in metastatic cancers. miR-675 inhibits tumor growth
factor beta 1 (TGFb1) signals by directly binding to the 30
UTR of
TGFb1. Overexpression of miR-675 and H19 repressed prostate
cancer cell migration [94]. In contrast, miR-675 enhances
tumorigenesis and metastasis of breast cancer cells [95]. H19
mediates estrogen-dependent cell proliferation in breast cancer
cell, suggesting the oncogenic role of H19 in estrogen-depend-
ent breast cancer [96].
These lncRNAs are likely to function through interaction
with general epigenetic and chromatin remodeling factor
(PCAT-1, SChLAP1) to impact the transcription for cancer pro-
gress. Modulation of miRNAs (H19) or snoRNAs (GAS5) would be
important regulatory mechanism of lncRNAs in cancer.
Regulation of stability or splicing by lncRNAs (PCA3 and
MALAT1) broadens the insight of transcriptional regulation in
cell biology.
Future plans for clinical applications of ncRNA
Long ncRNAs or miRNAs can be used as diagnostic and prognos-
tic markers, and as novel specific therapeutic targets for CRPC.
Current prostate cancer genomic data can be fully exploited if
noncoding regions are studied in detail. In addition, ncRNAs are
interesting targets in cancer therapy because their cancer- and
tissue-specific expression can be a major advantage over other
therapeutic options. Several other studies have also demon-
strated the clinical significance of ncRNAs. However, before we
can make use of these new therapeutic options [97], many more
functional and structural studies will be required. The exponen-
tially growing number of studies reporting new ncRNAs or andro-
gen signaling molecules will contribute to progress in this field.
Key Points
• Oncogenic miRNAs (miR-21, 29a/b, 148a and 125b) are
androgen-regulated and regulate tumor growth by tar-
geting epigenetic status, apoptosis and cell cycle
regulators.
• High-throughput analysis using RNA-seq, GRO-seq
and CAGE has identified abundant androgen-regulated
and prostate cancer-associated lncRNAs.
• Androgen-regulated AS RNA, CTBP1-AS, regulates glo-
bal androgen signals and cell cycle regulators by asso-
ciating with RNA-biding epigenetic complexes.
• RNA-seq analyses of tumor samples have identified
lncRNAs such as PCAT-1 and SChLAP1 that could be
biomarkers for prostate cancer.
• AR interacts with some lncRNAs such as HOTAIR,
eRNAs, SRA, PCGEM and PNCR1 to regulate AR tran-
scriptional activity and promote prostate cancer.
Funding
This work was supported by Grants of the P-Direct (S.I.)
from the MEXT, Japan; by Grants (S.I. and K.T.) from the
JSPS, Japan; by Grants from Takeda Science Foundation (S.I.
and K.T.), Japan; by a Grant from Mochida Memorial
Research Foundation (K.T), Japan.
References
1. Gutschner T, Diederichs S. The hallmarks of cancer: a long
non-coding RNA point of view. RNA Biol 2012;9:703–19.
2. Djebali S, Davis CA, Merkel A, et al. Landscape of transcription
in human cells. Nature 2012;489:101–8.
3. Mercer TR, Mattick JS, et al. Structure and function of long
noncoding RNAs in epigenetic regulation. Nat Struct Mol Biol
2013;20:300–7.
4. Derrien T, Johnson R, Bussotti G, et al. The GENCODE v7 cata-
log of human long noncoding RNAs: analysis of their gene
structure, evolution, and expression. Genome Res
2012;22:1775–89.
5. Du Z, Fei T, Verhaak RG, et al. Integrative genomic analyses
reveal clinically relevant long noncoding RNAs in human
cancer. Nat Struct Mol Biol 2013;20:908–13.
6. Kan Z, Jaiswal BS, Stinson J, et al. Diverse somatic mutation
patterns and pathway alterations in human cancers. Nature
2010;466:869–73.
7. Moran VA, Perera RJ, Khalil AM, et al. Emerging functional and
mechanistic paradigms of mammalian long non-coding
RNAs. Nucleic Acids Res 2012;40:6391–400.
8. Debes JD, Tindall DJ. Mechanism of androgen-refractory pros-
tate cancer. N Eng J Med 2004;351:1488–90.
9. Balk SP, Knudsen KE. AR, the cell cycle, and prostate cancer.
Nucl Recept Signal 2008;6:e001.
10.Schmidt LJ, Tindall DJ. Steroid 5 alpha-reductase inhibitors
targeting BPH and prostate cancer. J Steroid Biochem Mol Biol
2011;125:32–8.
11.Zhou ZX, Lane MV, Kemppainen JA, et al. Specificity of ligand-
dependent androgen receptor stabilization: receptor domain
interactions influence ligand dissociation and receptor stabil-
ity. Mol Endocrinol 1995;9:208–18.
12.Takayama K, Inoue S. Transcriptional network of androgen
receptor in prostate cancer progression. Int J Urol
2013;20(8):756–68.
13.Shang Y Myers M, Brown M. Formation of the androgen re-
ceptor transcription complex. Mol Cell 2002;9:601–10.
14.Chen CD, Welsbie DS, Tran C, et al. Molecular determinants of
resistance to antiandrogen therapy. Nat Med 2004;10:33–9.
15.Waltering KK, Helenius MA, Sahu B, et al. Increased expres-
sion of androgen receptor sensitizes prostate cancer cells to
low levels of androgens. Cancer Res 2009;69:8141–9.
16.Locke JA, Guns ES, Lubik AA, et al. Androgen levels increase
by intratumoral de novo steroidogenesis during progression
262 | Takayama and Inoue
Downloadedfromhttps://academic.oup.com/bfg/article-abstract/15/3/257/1742432bygueston10March2020
of castration-resistant prostate cancer. Cancer Res
2008;68:6407–15.
17.Sun S, Sprenger CC, Vessella RL, et al. Castration resistance in
human prostate cancer is conferred by a frequently occurring
androgen receptor splice variant. J Clin Invest 2010;120:2715–30.
18.Cai C, He HH, Chen S, et al. Androgen receptor gene expres-
sion in prostate cancer is directly suppressed by the androgen
receptor through recruitment of lysine-specific demethylase
1. Cancer Cell 2011;20:457–71.
19.Kichevsky AM, Gabriely G. miR-21: small multi-faceted RNA.
J Cell Mol Med 2009;13:39–53.
20.Ribas J, Ni X, Haffner M, et al. miR-21: an androgen receptor-
regulated microRNA that promotes hormone-dependent and
hormone-independent prostate cancer growth. Cancer Res
2009;69:7165–9.
21.Li T, Li D, Sha J, et al. MicroRNA-21 directly targets MARCKS
and promotes apoptosis resistance and invasion in prostate
cancer cells. Biochem Biophys Res Commun 2009;383:280–5.
22.Coppola V, Musumeci M, Patrizii M, et al. BTG2 loss and miR-
21 upregulation contribute to prostate cell transformation
by inducing luminal markers expression and epithelial-
mesenchymal transition. Oncogene 2013;32:1843–53.
23.Liu LZ, Li C, Chen Q, et al. MiR-21 induced angiogenesis
through AKT and ERK activation and HIF-1a expression. PLoS
One 2011;6:e19139.
24.Shi XB, Xue L, Yang J, et al. An androgen-regulated miRNA
suppresses Bak1 expression and induces androgen-
independent growth of prostate cancer cells. Proc Natl Acad Sci
USA 2007;104:19983–8.
25.Murata T, Takayama K, Katayama S, et al. miR-148a is an an-
drogen-responsive microRNA that promotes LNCaP prostate
cell growth by repressing its target CAND1 expression.
Prostate Cancer Prostatic Dis 2010;13:356–61.
26.Walter BA, Valera VA, Pinto PA, et al. Comprehensive
microRNA profiling of prostate cancer. J Cancer
2013;4:350–7.
27.Jalava SE, Urbanucci A, Latonen L, et al. Androgen-regulated
miR-32 targets BTG2 and is overexpressed in castration-
resistant prostate cancer. Oncogene 2012;31:4460–71.
28.Ambs S, Prueitt RL, Yi M, et al. Genomic profiling of microRNA
and messenger RNA reveals deregulated microRNA expres-
sion in prostate cancer. Cancer Res 2008;68:6162–70.
29.Wang Y, Zhang X, Li H, et al. The role of miRNA-29 family in
cancer. Eur J Cell Biol 2013;92(3):123–8.
30.Jiang H, Zhang G, Wu JH, et al. Diverse roles of miR-29 in can-
cer. Oncol Rep 2014;31(4):1509–16.
31.Wang C, Bian Z, Wei D, et al. miR-29b regulates migration
of human breast cancer cells. Mol Cell Biochem 2011;352(1–2):
197–207.
32.Gebeshuber CA, Zatloukal K, Martinez J. miR-29a suppresses
tristetraprolin, which is a regulator of epithelial polarity and
metastasis. EMBO Rep 2009;10(4):400–5.
33.Kong G, Zhang J, Zhang S, et al. Upregulated microRNA-29a by
hepatitis B virus X protein enhances hepatoma cell migration
by targeting PTEN in cell culture model. PLoS One
2011;6(5):e19518.
34.Takayama K, Misawa A, Suzuki T, et al. TET2 repression by
androgen hormone regulates global hydroxymethylation
status and prostate cancer progression. Nat Commun
2015;6:8219.
35.Sun T, Wang Q, Balk S, et al. The role of microRNA-221 and
microRNA-222 in androgen-independent prostate cancer cell
lines. Cancer Res 2009;69:3356–63.
36.Segal CV, Koufaris C, Powell C, et al. Effects of treatment with
androgen receptor ligands on microRNA expression of pros-
tate cancer cells. Toxicology 2015;333:45–52.
37.Galardi S, Mercatelli N, Giorda E, et al. miR-221 and miR-222
expression affects the proliferation potential of human pros-
tate carcinoma cell lines by targeting p27Kip1. J Biol Chem
2007;282:23716–24.
38.Sun T, Wang X, He HH, et al. MiR-221 promotes the develop-
ment of androgen independence in prostate cancer cells via
downregulation of HECTD2 and RAB1A. Oncogene
2014;33(21):2790–800.
39.Goto Y, Kojima S, Nishikawa R, et al. MicroRNA expression
signature of castration-resistant prostate cancer: the
microRNA-221/222 cluster functions as a tumour suppressor
and disease progression marker. Br J Cancer
2015;113(7):1055–65.
40.Varambally S, Cao Q, Mani RS, et al. Genomic loss of
microRNA-101 leads to overexpression of histone methyl-
transferase EZH2 in cancer. Science 2008;322:1695–9.
41.Melamed J, Einhorn JM, Ittmann MM. Allelic loss on chromo-
some 13q in human prostate carcinoma. Clin Cancer Res
1997;3:1867–72.
42.Bonci D, Coppola V, Musumeci M, et al. The miR-15a-miR-16-1
cluster controls prostate cancer by targeting multiple onco-
genic activities. Nat Med 2008;14:1271–7.
43.Wang Q, Li W, Zhang Y, et al. Androgen receptor regulates a
distinct transcription program in androgen-independent
prostate cancer. Cell 2009;138:245–56.
44.Takayama K, Kaneshiro K, Tsutsumi S, et al. Identification of
novel androgen response genes in prostate cancer cells by
coupling chromatin immunoprecipitation and genomic
microarray analysis. Oncogene 2007;26:4453–63.
45.Takayama K, Tsutsumi S, Horie-Inoue K, et al. Amyloid pre-
cursor protein is a primary androgen target gene that pro-
motes prostate cancer growth. Cancer Res 2009;69:137–42.
46.Wang Q, Li W, Liu XS, et al. A hierarchical network of tran-
scription factors governs androgen receptor-dependent pros-
tate cancer growth. Mol Cell 2007;27:380–92.
47.Jia L, Berman BP, Jariwala U, et al. Genomic androgen recep-
tor-occupied regions with different functions, defined by his-
tone acetylation, coregulators and transcriptional capacity.
PLoS One 2008;3:e3645.
48.Massie CE, Adryan B, Barbosa-Morais NL, et al. New androgen
receptor genomic targets show an interaction with the ETS1
transcription factor. EMBO Rep 2007;8:871–8.
49.Yu J, Yu J, Mani RS, et al. An integrated network of androgen
receptor, polycomb, and TMPRSS2-ERG gene fusions in pros-
tate cancer progression. Cancer Cell 2010;17:443–54.
50.Urbanucci A, Sahu B, Seppa¨la¨ J, et al. Overexpression of an-
drogen receptor enhances the binding of the receptor to
the chromatin in prostate cancer. Oncogene
2012;31:2153–63.
51.Tan PY, Chang CW, Chng KR, et al. Integration of regulatory
networks by NKX3-1 promotes androgen-dependent prostate
cancer survival. Mol Cell Biol 2012;32:399–414.
52.Wang D, Garcia-Bassets I, Benner C, et al. Reprogramming
transcription by distinct classes of enhancers functionally
defined by eRNA. Nature 2011;474:390–4.
53.Shiraki T, Kondo S, Katayama S, et al. Cap analysis gene ex-
pression for high-throughput analysis of transcriptional
starting point and identification of promoter usage. Proc Natl
Acad Sci USA 2003;100:15776–81.
54.Takayama K, Katayama S, Horie-Inoue K, et al. Integration of
cap analysis of gene expression and chromatin
Emerging role of noncoding RNA in prostate cancer progression | 263
Downloadedfromhttps://academic.oup.com/bfg/article-abstract/15/3/257/1742432bygueston10March2020
immunoprecipitation analysis on array reveals genome-wide
androgen receptor signaling in prostate cancer cells. Oncogene
2011;30(5):619–30.
55.Carninci P, Kasukawa T, Katayama S, et al. The transcriptional
landscape of the mammalian genome. Science
2005;309:1559–63.
56.Katayama S, Tomaru Y, Kasukawa T, et al. Antisense tran-
scription in the mammalian transcriptome. Science
2005;309:1564–6.
57.Rosok O, Sioud M. Systematic identification of sense-
antisense transcripts in mammalian cells. Nat Biotechnol
2004;22:104–8.
58.Yu W, Gius D, Onyango P, et al. Epigenetic silencing of tumour
suppressor gene p15 by its antisense RNA. Nature
2008;451:202–6.
59.Yap KL, Li S, Mun˜ oz-Cabello AM, et al. Molecular interplay of
the noncoding RNA ANRIL and methylated histone H3 lysine
27 by polycomb CBX7 in transcriptional silencing of INK4a.
Mol Cell 2010;38:662–74.
60.Ogawa Y, Sun BK, Lee JT, et al. Intersect, ion of the RNA inter-
ference and X-inactivation pathways. Science 2008;320:
1336–41.
61.Gupta RA, Shah N, Wang KC, et al. Long non-coding RNA
HOTAIR reprograms chromatin state to promote cancer me-
tastasis. Nature 2010;464:1071–6.
62.Tsai MC, Manor O, Wan Y, et al. Long noncoding RNA as
modular scaffold of histone modification complexes. Science
2010;329(5992):689–93.
63.Bhan A, Hussain I, Ansari KI, et al. Antisense transcript long
noncoding RNA (lncRNA) HOTAIR is transcriptionally
induced by estradiol. J Mol Biol 2013;425(19):3707–22.
64.Xue X, Yang YA, Zhang A, et al. LncRNA HOTAIR enhances ER
signaling and confers tamoxifen resistance in breast cancer.
Oncogene 2015. doi: 10.1038/onc.2015.340.
65.Zhang A, Zhao JC, Kim J, et al. LncRNA HOTAIR enhances the
androgen-receptor-mediated transcriptional program and
drives castration-resistant prostate cancer. Cell Rep
2015;13(1):209–21.
66.Takayama K, Horie-Inoue K, Katayama K, et al. Androgen-
responsive long noncoding RNA CTBP1-AS promotes prostate
cancer. EMBO J 2013;32:1665–80.
67.Shi Y, Sawada J, Sui G, et al. Coordinated histone modifica-
tions mediated by a CtBP co-repressor complex. Nature
2003;422:735–8.
68.Shav-Tal Y, Zipori D. PSF and p54(nrb)/NonO–multi-
functional nuclear proteins. FEBS Lett 2002;531:109–14.
69.Song X, Sui A, Garen A. Binding of mouse VL30 retrotrans-
poson RNA to PSF protein induces genes repressed by PSF: ef-
fects on steroidogenesis and oncogenesis. Proc Natl Acad Sci
USA 2004;101:621–6.
70.Lanz RB, McKenna NJ, Onate SA, et al. steroid receptor coacti-
vator, SRA, functions as an RNA and is present in an SRC-1
complex. Cell 1999;97(1):17–27.
71.Lanz RB, Razani B, Goldberg AD, et al. Distinct RNA motifs are
important for coactivation of steroid hormone receptors by
steroid receptor RNA activator (SRA). Proc Natl Acad Sci USA
2002;99(25):16081–6.
72.Kawashima H, Takano H, Sugita S, et al. A novel steroid recep-
tor co-activator protein (SRAP) as an alternative form of ster-
oid receptor RNA-activator gene: expression in prostate
cancer cells and enhancement of androgen receptor activity.
Biochem J 2003;369(Pt 1):163–71.
73.Agoulink IU, Weigel NL. Coactivator selective regulation of
androgen receptor activity. Steroids 2009;74:669–74.
74.Hsieh CL, Fei T, Chen Y, et al. Enhancer RNAs participate in
androgen receptor-driven looping that selectively enhances
gene activation. Proc Natl Acad Sci USA 2014;111(20):7319–24.
75.Srikantan V Zou Z, Petrovics G, et al. PCGEM1, a prostate-
specific gene, is overexpressed in prostate cancer. Proc Natl
Acad Sci USA 2000;97:12216–21.
76.Petrovics G, Zhang W, Makarem M, et al. Elevated expression
of PCGEM1, a prostate-specific gene with cell growth-
promoting function, is associated with high-risk prostate
cancer patients. Oncogene 2004;23:605–11.
77.Yang L, Lin C, Jin C, et al. lncRNA-dependent mechanisms of
androgen-receptor regulated gene activation programs.
Nature 2013;500:598–602.
78.Prensner JR, Sahu A, Iyer MK, et al. The IncRNAs PCGEM1 and
PRNCR1 are not implicated in castration resistant prostate
cancer. Oncotarget 2014;5(6):1434–8.
79.Pickard MR, Mourtada-Maarabouni M, Williams GT, et al.
Long non-coding RNA GAS5 regulates apoptosis in pros-
tate cancer cell lines. Biochem Biophys Acta 2013;1832:
1613–23.
80.Mourtada-Maarabouni M, Pickard MR, Hedge VL, et al. GAS5, a
non-protein-coding RNA, controls apoptosis and is downre-
gulated in breast cancer. Oncogene 2009;8(2):195–208.
81.Paris PL, Andaya A, Fridlyand J, et al. Whole genome scanning
identifies genotypes associated with recurrence and metasta-
sis in prostate tumor. Hum Mol Genet 2004;13:1303–13.
82.Ren S, Liu Y, Xu W, et al. Long noncoding RNA MALAT-1 is a
new potential therapeutic target for castration resistant pros-
tate cancer. J Urol 2013;190:2278–87.
83.Guffanti A, Iacono M, Pelucchi P, et al. A transcriptional sketch
of a primary human breast cancer by 454 deep sequencing,
BMC Genomics 2009;10:163.
84.Zhao Z, Chen C, Liu Y, et al. 17beta-Estradiol treatment in-
hibits breast cell proliferation, migration and invasion by
decreasing MALAT-1 RNA level. Biochem Biophys Res Commun
2014;445:388–93.
85.Prensner JR, Iyer MK, Balbin OA, et al. Transcriptome sequenc-
ing across a prostate cancer cohort identifies PCAT-1, an
unannotated lincRNA implicated in disease progression. Nat
Biotechnol 2011;29(8):742–9.
86.Prensner JR, Chen W, Iyer MK, et al. PCAT-1, a long noncoding
RNA, regulates BRCA2 and controls homologous recombin-
ation in cancer. Cancer Res 2014;74(6):1651–60.
87.Prensner JR, Iyer MK, Sahu A, et al. The long noncoding
RNA SChLAP1 promotes aggressive prostate cancer and
antagonizes the SWI/SNF complex. Nat Genet 2013;45(11):
1392–8.
88.Prensner JR, Zhao S, Erho N, et al. RNA biomarkers associated
with metastatic progression in prostate cancer: a multi-
institutional high-throughput analysis of SChLAP1. Lancet
Oncol 2014;15(13):1469–80.
89.Bussemakers MJ, et al. DD3: a new prostate-specific gene,
highly overexpressed in prostate cancer. Cancer Res
1999;59:5975–9.
90.Ferreira LB, van Bokhoven A, Verhaegh GW, et al. PCA3 non-
coding RNA is involved in the control of prostate-cancer cell
survival and modulates androgen receptor signaling. BMC
Cancer 2013;12:507.
91.Salameh A, Lee AK, Cardo-Vila M, et al. PRUNE2 is a human
prostate cancer suppressor regulated by the intronic long
noncoding RNA PCA3. Proc Natl Acad Sci USA 2015;112(27):
8403–8.
92.Hessels D, Schalken JA. The use of PCA3 in the diagnosis of
prostate cancer. Nat Rev Urol 2009;6:255–61.
264 | Takayama and Inoue
Downloadedfromhttps://academic.oup.com/bfg/article-abstract/15/3/257/1742432bygueston10March2020
93.Lee GL, Dobi A, Srivastava S. Prostate cancer: diagnostic
performance of the PCA3 urine test. Nat Rev Urol 2011;8:
123–4.
94.Zhu M, Chen Q, Liu X, et al. lncRNA H19/miR-675 axis re-
pressed prostate cancer metastasis by targeting TGFBI. FEBS J
2014;281; 3766–75.
95.Sun H, Wang G, Peng Y, et al. H19 lncRNA mediates 17b-
estradiol-induced cell proliferation in MCF-7 breast cancer
cells. Oncol Rep 2015;33(6):3045–52.
96.Vennin C, Spruyt N, Dahmani F, et al. H19 non coding RNA-
derived miR-675 enhances tumorigenesis and metastasis of
breast cancer cells by downregulating c-Cbl and Cbl-b.
Oncotarget 2015;6:29209–23.
97.Leyten GH, Hessels D, Jannink SA, et al. Prospective multi-
centre evaluation of PCA3 and TMPRSS2-ERG gene fusions as
diagnostic and prognostic urinary biomarkers for prostate
cancer. Eur Urol 2014;65:534–42.
Emerging role of noncoding RNA in prostate cancer progression | 265
Downloadedfromhttps://academic.oup.com/bfg/article-abstract/15/3/257/1742432bygueston10March2020

More Related Content

What's hot

11.06.13 - 2013 NIH Research Festival Poster
11.06.13 - 2013 NIH Research Festival Poster11.06.13 - 2013 NIH Research Festival Poster
11.06.13 - 2013 NIH Research Festival Poster
Farhoud Faraji
 
Rectal cancer
Rectal cancerRectal cancer
Rectal cancer
Springer
 
2020 regulatory mechanism of micro rna expression in cancer
2020   regulatory mechanism of micro rna expression in cancer2020   regulatory mechanism of micro rna expression in cancer
2020 regulatory mechanism of micro rna expression in cancer
Antar
 
Bmc Cancer Mmp3 2010
Bmc Cancer  Mmp3 2010Bmc Cancer  Mmp3 2010
Bmc Cancer Mmp3 2010
rm8509
 
03.29.12 - SLU PhD Admissions Seminar
03.29.12 - SLU PhD Admissions Seminar03.29.12 - SLU PhD Admissions Seminar
03.29.12 - SLU PhD Admissions Seminar
Farhoud Faraji
 
Thesis-Final Draft
Thesis-Final DraftThesis-Final Draft
Thesis-Final Draft
Emer Shelly
 
Inhibition of KPNA4 Attenuates Prostate Cancer Metastasis
Inhibition of KPNA4 Attenuates Prostate Cancer MetastasisInhibition of KPNA4 Attenuates Prostate Cancer Metastasis
Inhibition of KPNA4 Attenuates Prostate Cancer Metastasis
Xin Li
 
Melanoma frontline approach
Melanoma frontline approachMelanoma frontline approach
Melanoma frontline approach
Ereny Samwel
 
Cancer Res-2014-Chakraborty-3489-500
Cancer Res-2014-Chakraborty-3489-500Cancer Res-2014-Chakraborty-3489-500
Cancer Res-2014-Chakraborty-3489-500
Rachel Stupay
 
Estrogen-induced chromatin decondensation and nuclear re-organization linked ...
Estrogen-induced chromatin decondensation and nuclear re-organization linked ...Estrogen-induced chromatin decondensation and nuclear re-organization linked ...
Estrogen-induced chromatin decondensation and nuclear re-organization linked ...
Sehrish Rafique
 
Micro rna signature with indolent non hodgkin lymphomas
Micro rna signature with indolent non hodgkin  lymphomasMicro rna signature with indolent non hodgkin  lymphomas
Micro rna signature with indolent non hodgkin lymphomas
Mohsin Maqbool
 

What's hot (19)

11.06.13 - 2013 NIH Research Festival Poster
11.06.13 - 2013 NIH Research Festival Poster11.06.13 - 2013 NIH Research Festival Poster
11.06.13 - 2013 NIH Research Festival Poster
 
Rectal cancer
Rectal cancerRectal cancer
Rectal cancer
 
publi Toulouse
publi Toulousepubli Toulouse
publi Toulouse
 
2020 regulatory mechanism of micro rna expression in cancer
2020   regulatory mechanism of micro rna expression in cancer2020   regulatory mechanism of micro rna expression in cancer
2020 regulatory mechanism of micro rna expression in cancer
 
Bmc Cancer Mmp3 2010
Bmc Cancer  Mmp3 2010Bmc Cancer  Mmp3 2010
Bmc Cancer Mmp3 2010
 
Metanalisys
MetanalisysMetanalisys
Metanalisys
 
03.29.12 - SLU PhD Admissions Seminar
03.29.12 - SLU PhD Admissions Seminar03.29.12 - SLU PhD Admissions Seminar
03.29.12 - SLU PhD Admissions Seminar
 
Characteristic mTOR activity in Hodgkin-lymphomas offers a potential therapeu...
Characteristic mTOR activity in Hodgkin-lymphomas offers a potential therapeu...Characteristic mTOR activity in Hodgkin-lymphomas offers a potential therapeu...
Characteristic mTOR activity in Hodgkin-lymphomas offers a potential therapeu...
 
mi RNA en route to the clinic
mi RNA en route to the   clinic mi RNA en route to the   clinic
mi RNA en route to the clinic
 
Thesis-Final Draft
Thesis-Final DraftThesis-Final Draft
Thesis-Final Draft
 
Inhibition of KPNA4 Attenuates Prostate Cancer Metastasis
Inhibition of KPNA4 Attenuates Prostate Cancer MetastasisInhibition of KPNA4 Attenuates Prostate Cancer Metastasis
Inhibition of KPNA4 Attenuates Prostate Cancer Metastasis
 
Melanoma frontline approach
Melanoma frontline approachMelanoma frontline approach
Melanoma frontline approach
 
Cancer Res-2014-Chakraborty-3489-500
Cancer Res-2014-Chakraborty-3489-500Cancer Res-2014-Chakraborty-3489-500
Cancer Res-2014-Chakraborty-3489-500
 
microRNA discovery and biomarker development in clinical samples
microRNA discovery and biomarker development in clinical samplesmicroRNA discovery and biomarker development in clinical samples
microRNA discovery and biomarker development in clinical samples
 
Ovariancancer
Ovariancancer Ovariancancer
Ovariancancer
 
The role of selected microRNAs (miRNAs) in the negative regulation of pediatr...
The role of selected microRNAs (miRNAs) in the negative regulation of pediatr...The role of selected microRNAs (miRNAs) in the negative regulation of pediatr...
The role of selected microRNAs (miRNAs) in the negative regulation of pediatr...
 
Mechanism of cancer development pdf
Mechanism of cancer development pdfMechanism of cancer development pdf
Mechanism of cancer development pdf
 
Estrogen-induced chromatin decondensation and nuclear re-organization linked ...
Estrogen-induced chromatin decondensation and nuclear re-organization linked ...Estrogen-induced chromatin decondensation and nuclear re-organization linked ...
Estrogen-induced chromatin decondensation and nuclear re-organization linked ...
 
Micro rna signature with indolent non hodgkin lymphomas
Micro rna signature with indolent non hodgkin  lymphomasMicro rna signature with indolent non hodgkin  lymphomas
Micro rna signature with indolent non hodgkin lymphomas
 

Similar to Elv057

BiPday 2014 --Creanza Teresa
BiPday 2014 --Creanza TeresaBiPday 2014 --Creanza Teresa
BiPday 2014 --Creanza Teresa
eventi-ITBbari
 
paraphrase the review in your own wordsThe tumor suppressor PTEN .pdf
paraphrase the review in your own wordsThe tumor suppressor PTEN .pdfparaphrase the review in your own wordsThe tumor suppressor PTEN .pdf
paraphrase the review in your own wordsThe tumor suppressor PTEN .pdf
arihantgiftgallery
 
The Role of MicroRNAs in the Progression, Prognostication, and Treatment of B...
The Role of MicroRNAs in the Progression, Prognostication, and Treatment of B...The Role of MicroRNAs in the Progression, Prognostication, and Treatment of B...
The Role of MicroRNAs in the Progression, Prognostication, and Treatment of B...
CrimsonpublishersCancer
 
Michael's IUCRL Poster 2014 Close to Final with CDW edits
Michael's IUCRL Poster 2014 Close to Final with CDW editsMichael's IUCRL Poster 2014 Close to Final with CDW edits
Michael's IUCRL Poster 2014 Close to Final with CDW edits
Michael Araya
 

Similar to Elv057 (20)

The role of micro rn as in the occurrence and development of esophageal squam...
The role of micro rn as in the occurrence and development of esophageal squam...The role of micro rn as in the occurrence and development of esophageal squam...
The role of micro rn as in the occurrence and development of esophageal squam...
 
Micro RNA biogenesis pathways in cancer - Article.
Micro RNA biogenesis pathways in cancer - Article.Micro RNA biogenesis pathways in cancer - Article.
Micro RNA biogenesis pathways in cancer - Article.
 
BiPday 2014 --Creanza Teresa
BiPday 2014 --Creanza TeresaBiPday 2014 --Creanza Teresa
BiPday 2014 --Creanza Teresa
 
Roles of circular rn as and their interactions with micro rnas in human disor...
Roles of circular rn as and their interactions with micro rnas in human disor...Roles of circular rn as and their interactions with micro rnas in human disor...
Roles of circular rn as and their interactions with micro rnas in human disor...
 
Role of micro rna in oncogenesis
Role of micro rna in oncogenesisRole of micro rna in oncogenesis
Role of micro rna in oncogenesis
 
paraphrase the review in your own wordsThe tumor suppressor PTEN .pdf
paraphrase the review in your own wordsThe tumor suppressor PTEN .pdfparaphrase the review in your own wordsThe tumor suppressor PTEN .pdf
paraphrase the review in your own wordsThe tumor suppressor PTEN .pdf
 
Crimson Publishers-SiRNA Delivery for Cancer Therapy: Challenges and Future P...
Crimson Publishers-SiRNA Delivery for Cancer Therapy: Challenges and Future P...Crimson Publishers-SiRNA Delivery for Cancer Therapy: Challenges and Future P...
Crimson Publishers-SiRNA Delivery for Cancer Therapy: Challenges and Future P...
 
A review of micro rn as related to the occurrence, diagnosis, and prognosis o...
A review of micro rn as related to the occurrence, diagnosis, and prognosis o...A review of micro rn as related to the occurrence, diagnosis, and prognosis o...
A review of micro rn as related to the occurrence, diagnosis, and prognosis o...
 
Molecular target therapy_in_colorectal_cancer[1]
Molecular target therapy_in_colorectal_cancer[1]Molecular target therapy_in_colorectal_cancer[1]
Molecular target therapy_in_colorectal_cancer[1]
 
The Role of MicroRNAs in the Progression, Prognostication, and Treatment of B...
The Role of MicroRNAs in the Progression, Prognostication, and Treatment of B...The Role of MicroRNAs in the Progression, Prognostication, and Treatment of B...
The Role of MicroRNAs in the Progression, Prognostication, and Treatment of B...
 
Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...
Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...
Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...
 
Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...
Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...
Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...
 
Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...
Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...
Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...
 
Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...
Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...
Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...
 
Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...
Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...
Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...
 
Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...
Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...
Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...
 
Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...
Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...
Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...
 
Michael's IUCRL Poster 2014 Close to Final with CDW edits
Michael's IUCRL Poster 2014 Close to Final with CDW editsMichael's IUCRL Poster 2014 Close to Final with CDW edits
Michael's IUCRL Poster 2014 Close to Final with CDW edits
 
Oncotarget-2016
Oncotarget-2016Oncotarget-2016
Oncotarget-2016
 
2009-Hepatology
2009-Hepatology2009-Hepatology
2009-Hepatology
 

More from University of Zambia, School of Pharmacy, Lusaka, Zambia

More from University of Zambia, School of Pharmacy, Lusaka, Zambia (20)

7 biotechnology and human disease
7 biotechnology and human disease7 biotechnology and human disease
7 biotechnology and human disease
 
6 radiopharmaceutical systems
6 radiopharmaceutical systems6 radiopharmaceutical systems
6 radiopharmaceutical systems
 
4 preformulation
4 preformulation4 preformulation
4 preformulation
 
2 colloidal system
2 colloidal system2 colloidal system
2 colloidal system
 
1 general polymer science
1 general polymer science1 general polymer science
1 general polymer science
 
15 sedimentation
15 sedimentation15 sedimentation
15 sedimentation
 
15 lyophilization
15 lyophilization15 lyophilization
15 lyophilization
 
15 heat transfer
15 heat transfer15 heat transfer
15 heat transfer
 
15 extraction
15 extraction15 extraction
15 extraction
 
15 evaporation transpiration sublimation
15 evaporation transpiration sublimation15 evaporation transpiration sublimation
15 evaporation transpiration sublimation
 
15 distillation
15 distillation15 distillation
15 distillation
 
15 crystallization
15 crystallization15 crystallization
15 crystallization
 
15 coagulation and flocculation
15 coagulation and flocculation15 coagulation and flocculation
15 coagulation and flocculation
 
15 mixing
15 mixing15 mixing
15 mixing
 
15 filtration
15 filtration15 filtration
15 filtration
 
15 drying
15 drying15 drying
15 drying
 
15 communition
15 communition15 communition
15 communition
 
15 adsorption
15 adsorption15 adsorption
15 adsorption
 
14 rheology
14 rheology14 rheology
14 rheology
 
13 polymer science
13 polymer science13 polymer science
13 polymer science
 

Recently uploaded

📱Dehradun Call Girls Service 📱☎️ +91'905,3900,678 ☎️📱 Call Girls In Dehradun 📱
📱Dehradun Call Girls Service 📱☎️ +91'905,3900,678 ☎️📱 Call Girls In Dehradun 📱📱Dehradun Call Girls Service 📱☎️ +91'905,3900,678 ☎️📱 Call Girls In Dehradun 📱
📱Dehradun Call Girls Service 📱☎️ +91'905,3900,678 ☎️📱 Call Girls In Dehradun 📱
@Chandigarh #call #Girls 9053900678 @Call #Girls in @Punjab 9053900678
 
VIP Call Girls Pollachi 7001035870 Whatsapp Number, 24/07 Booking
VIP Call Girls Pollachi 7001035870 Whatsapp Number, 24/07 BookingVIP Call Girls Pollachi 7001035870 Whatsapp Number, 24/07 Booking
VIP Call Girls Pollachi 7001035870 Whatsapp Number, 24/07 Booking
dharasingh5698
 
( Pune ) VIP Baner Call Girls 🎗️ 9352988975 Sizzling | Escorts | Girls Are Re...
( Pune ) VIP Baner Call Girls 🎗️ 9352988975 Sizzling | Escorts | Girls Are Re...( Pune ) VIP Baner Call Girls 🎗️ 9352988975 Sizzling | Escorts | Girls Are Re...
( Pune ) VIP Baner Call Girls 🎗️ 9352988975 Sizzling | Escorts | Girls Are Re...
nilamkumrai
 
在线制作约克大学毕业证(yu毕业证)在读证明认证可查
在线制作约克大学毕业证(yu毕业证)在读证明认证可查在线制作约克大学毕业证(yu毕业证)在读证明认证可查
在线制作约克大学毕业证(yu毕业证)在读证明认证可查
ydyuyu
 

Recently uploaded (20)

"Boost Your Digital Presence: Partner with a Leading SEO Agency"
"Boost Your Digital Presence: Partner with a Leading SEO Agency""Boost Your Digital Presence: Partner with a Leading SEO Agency"
"Boost Your Digital Presence: Partner with a Leading SEO Agency"
 
(INDIRA) Call Girl Pune Call Now 8250077686 Pune Escorts 24x7
(INDIRA) Call Girl Pune Call Now 8250077686 Pune Escorts 24x7(INDIRA) Call Girl Pune Call Now 8250077686 Pune Escorts 24x7
(INDIRA) Call Girl Pune Call Now 8250077686 Pune Escorts 24x7
 
2nd Solid Symposium: Solid Pods vs Personal Knowledge Graphs
2nd Solid Symposium: Solid Pods vs Personal Knowledge Graphs2nd Solid Symposium: Solid Pods vs Personal Knowledge Graphs
2nd Solid Symposium: Solid Pods vs Personal Knowledge Graphs
 
𓀤Call On 7877925207 𓀤 Ahmedguda Call Girls Hot Model With Sexy Bhabi Ready Fo...
𓀤Call On 7877925207 𓀤 Ahmedguda Call Girls Hot Model With Sexy Bhabi Ready Fo...𓀤Call On 7877925207 𓀤 Ahmedguda Call Girls Hot Model With Sexy Bhabi Ready Fo...
𓀤Call On 7877925207 𓀤 Ahmedguda Call Girls Hot Model With Sexy Bhabi Ready Fo...
 
Hire↠Young Call Girls in Tilak nagar (Delhi) ☎️ 9205541914 ☎️ Independent Esc...
Hire↠Young Call Girls in Tilak nagar (Delhi) ☎️ 9205541914 ☎️ Independent Esc...Hire↠Young Call Girls in Tilak nagar (Delhi) ☎️ 9205541914 ☎️ Independent Esc...
Hire↠Young Call Girls in Tilak nagar (Delhi) ☎️ 9205541914 ☎️ Independent Esc...
 
Pune Airport ( Call Girls ) Pune 6297143586 Hot Model With Sexy Bhabi Ready...
Pune Airport ( Call Girls ) Pune  6297143586  Hot Model With Sexy Bhabi Ready...Pune Airport ( Call Girls ) Pune  6297143586  Hot Model With Sexy Bhabi Ready...
Pune Airport ( Call Girls ) Pune 6297143586 Hot Model With Sexy Bhabi Ready...
 
20240510 QFM016 Irresponsible AI Reading List April 2024.pdf
20240510 QFM016 Irresponsible AI Reading List April 2024.pdf20240510 QFM016 Irresponsible AI Reading List April 2024.pdf
20240510 QFM016 Irresponsible AI Reading List April 2024.pdf
 
📱Dehradun Call Girls Service 📱☎️ +91'905,3900,678 ☎️📱 Call Girls In Dehradun 📱
📱Dehradun Call Girls Service 📱☎️ +91'905,3900,678 ☎️📱 Call Girls In Dehradun 📱📱Dehradun Call Girls Service 📱☎️ +91'905,3900,678 ☎️📱 Call Girls In Dehradun 📱
📱Dehradun Call Girls Service 📱☎️ +91'905,3900,678 ☎️📱 Call Girls In Dehradun 📱
 
VIP Call Girls Pollachi 7001035870 Whatsapp Number, 24/07 Booking
VIP Call Girls Pollachi 7001035870 Whatsapp Number, 24/07 BookingVIP Call Girls Pollachi 7001035870 Whatsapp Number, 24/07 Booking
VIP Call Girls Pollachi 7001035870 Whatsapp Number, 24/07 Booking
 
Busty Desi⚡Call Girls in Vasundhara Ghaziabad >༒8448380779 Escort Service
Busty Desi⚡Call Girls in Vasundhara Ghaziabad >༒8448380779 Escort ServiceBusty Desi⚡Call Girls in Vasundhara Ghaziabad >༒8448380779 Escort Service
Busty Desi⚡Call Girls in Vasundhara Ghaziabad >༒8448380779 Escort Service
 
Wagholi & High Class Call Girls Pune Neha 8005736733 | 100% Gennuine High Cla...
Wagholi & High Class Call Girls Pune Neha 8005736733 | 100% Gennuine High Cla...Wagholi & High Class Call Girls Pune Neha 8005736733 | 100% Gennuine High Cla...
Wagholi & High Class Call Girls Pune Neha 8005736733 | 100% Gennuine High Cla...
 
Pirangut | Call Girls Pune Phone No 8005736733 Elite Escort Service Available...
Pirangut | Call Girls Pune Phone No 8005736733 Elite Escort Service Available...Pirangut | Call Girls Pune Phone No 8005736733 Elite Escort Service Available...
Pirangut | Call Girls Pune Phone No 8005736733 Elite Escort Service Available...
 
best call girls in Hyderabad Finest Escorts Service 📞 9352988975 📞 Available ...
best call girls in Hyderabad Finest Escorts Service 📞 9352988975 📞 Available ...best call girls in Hyderabad Finest Escorts Service 📞 9352988975 📞 Available ...
best call girls in Hyderabad Finest Escorts Service 📞 9352988975 📞 Available ...
 
( Pune ) VIP Baner Call Girls 🎗️ 9352988975 Sizzling | Escorts | Girls Are Re...
( Pune ) VIP Baner Call Girls 🎗️ 9352988975 Sizzling | Escorts | Girls Are Re...( Pune ) VIP Baner Call Girls 🎗️ 9352988975 Sizzling | Escorts | Girls Are Re...
( Pune ) VIP Baner Call Girls 🎗️ 9352988975 Sizzling | Escorts | Girls Are Re...
 
APNIC Policy Roundup, presented by Sunny Chendi at the 5th ICANN APAC-TWNIC E...
APNIC Policy Roundup, presented by Sunny Chendi at the 5th ICANN APAC-TWNIC E...APNIC Policy Roundup, presented by Sunny Chendi at the 5th ICANN APAC-TWNIC E...
APNIC Policy Roundup, presented by Sunny Chendi at the 5th ICANN APAC-TWNIC E...
 
VVIP Pune Call Girls Sinhagad WhatSapp Number 8005736733 With Elite Staff And...
VVIP Pune Call Girls Sinhagad WhatSapp Number 8005736733 With Elite Staff And...VVIP Pune Call Girls Sinhagad WhatSapp Number 8005736733 With Elite Staff And...
VVIP Pune Call Girls Sinhagad WhatSapp Number 8005736733 With Elite Staff And...
 
在线制作约克大学毕业证(yu毕业证)在读证明认证可查
在线制作约克大学毕业证(yu毕业证)在读证明认证可查在线制作约克大学毕业证(yu毕业证)在读证明认证可查
在线制作约克大学毕业证(yu毕业证)在读证明认证可查
 
Ganeshkhind ! Call Girls Pune - 450+ Call Girl Cash Payment 8005736733 Neha T...
Ganeshkhind ! Call Girls Pune - 450+ Call Girl Cash Payment 8005736733 Neha T...Ganeshkhind ! Call Girls Pune - 450+ Call Girl Cash Payment 8005736733 Neha T...
Ganeshkhind ! Call Girls Pune - 450+ Call Girl Cash Payment 8005736733 Neha T...
 
20240509 QFM015 Engineering Leadership Reading List April 2024.pdf
20240509 QFM015 Engineering Leadership Reading List April 2024.pdf20240509 QFM015 Engineering Leadership Reading List April 2024.pdf
20240509 QFM015 Engineering Leadership Reading List April 2024.pdf
 
Sarola * Female Escorts Service in Pune | 8005736733 Independent Escorts & Da...
Sarola * Female Escorts Service in Pune | 8005736733 Independent Escorts & Da...Sarola * Female Escorts Service in Pune | 8005736733 Independent Escorts & Da...
Sarola * Female Escorts Service in Pune | 8005736733 Independent Escorts & Da...
 

Elv057

  • 1. The emerging role of noncoding RNA in prostate cancer progression and its implication on diagnosis and treatment Ken-ichi Takay , MuungoLungwani-Tysonama and Satoshi Inoue Corresponding author: Satoshi Inoue, Bunkyo-ku, Tokyo 113-8655, Japan. E-mail: INOUE-GER@h.u-tokyo.ac.jp Abstract Recent transcriptome studies using next-generation sequencing have detected aberrant changes in the expression of non- coding RNAs (ncRNAs) associated with cancer. For prostate cancer, the expression levels of ncRNAs including microRNAs and long noncoding RNAs are strongly associated with diagnosis, carcinogenesis and tumor growth. Moreover, androgen and its cognate receptor, androgen receptor (AR), regulate various signaling pathways for prostate tumor growth. In add- ition, progression to lethal castration-resistant prostate cancer (CRPC) is also owing to AR function. Systematic analysis of AR-binding sites and their regulated transcripts revealed that many ncRNAs are widely regulated at the transcriptional level. Thus, recent studies provide new insight into the complicated molecular mechanism of prostate cancer progression. This review focused on the role of various ncRNAs in prostate cancer and the association between their expression and CRPC. Key words: CRPC; AR; cap analysis of gene expression (CAGE); miRNA; lncRNA; epigenome The role of noncoding RNAs in prostate cancer biology Recent advances in transcriptome technology have revealed that >90% of the human genome is actively transcribed [1]. The encyclopedia of DNA elements (ENCODE) project has shown that only 2% of these transcripts are translated into proteins [2]. The noncoding RNAs (ncRNAs), which occupy the majority of transcripts in the nucleus, were initially thought as the ‘dark matter’. Generally, ncRNAs are broadly divided into short (<200 nt) and long (>200 nt) transcripts [3]. In this review, we summarize the function and clinical significance of long non- coding RNAs (lncRNAs) and microRNAs (miRNAs) in prostate cancer and their regulation by the androgen receptor (AR). miRNAs play important roles in cancer by posttranscriptional modification of target mRNA or protein expression. LncRNAs represent most of the transcribed ncRNA in the human genome. GENCODE v19 includes 13 870 human lncRNA-related genes, which produce 23 898 lncRNAs [4]. These lncRNAs exhibit simi- lar structure and biogenesis as mRNAs. They are polyadeny- lated and may function in either nuclear or cytoplasmic compartments. Evidence showing that lncRNAs are aberrantly expressed in numerous human diseases including cancer sup- ports their importance [5–7]. The function of AR in castration resistance AR is a member of the nuclear receptor superfamily [8, 9] and is a key molecule for androgen signaling in its target organ, the prostate. Testosterone that is produced in the testes is the most abundant circulating androgen (90%). Testosterone diffuses into prostate cells and is converted to dihydrotestosterone (DHT) by the enzyme 5a-reductase [10]. DHT directly binds to and activates AR even more tightly than testosterone [11]. Before ligand binding, AR is found in the cytoplasm in a com- plex that includes molecular chaperones and co-chaperones from the heat shock protein (Hsp) family. After binding to an- drogen, a conformational change in the complex leads to nu- clear translocation of AR. In the nucleus, AR binds as a dimer to specific genomic sequences called androgen-responsive elem- ents (AREs), which are found in the promoter and enhancer Ken-ichi Takayama is a researcher of Geriatric Medicine, Graduate School of Medicine, The University of Tokyo. Satoshi Inoue is a professor of Anti-Aging Medicine, Graduate School of Medicine, The University of Tokyo. VC The Author 2015. Published by Oxford University Press. All rights reserved. For permissions, please email: journals.permissions@oup.com 257 Briefings in Functional Genomics, 15(3), 2016, 257–265 doi: 10.1093/bfgp/elv057 Advance Access Publication Date: 17 December 2015 Review paper Downloadedfromhttps://academic.oup.com/bfg/article-abstract/15/3/257/1742432bygueston10March2020
  • 2. regions of target genes (Figure 1) [12]. AR regulates its enhancer action by modifying the epigenetic condition of AR-binding sites (ARBSs) [13]. AR also induces prostate cancer development and progres- sion. Despite favorable responses to initial hormone therapy, most patients progress to lethal castration-resistant prostate cancer (CRPC) with elevated AR expression [14], hypersensitivity to androgens [15], intra-tumoral steroidogenesis [16] and devel- opment of AR variants [17]. Thus, identification of signaling events downstream of AR is critical for understanding the pro- gression of hormone-responsive prostate cancer to CRPC. Although AR overexpression is commonly observed in CRPC, the mechanism underlying altered AR expression is not com- pletely understood. However, recent studies showed that both transcriptional and epigenetic changes are important for AR upregulation in prostate cancer. A recent study analyzing an an- drogen-dependent prostate cancer cell line identified ARBSs in the introns of the AR gene [18]. Androgen treatment causes AR to bind the enhancer and recruit lysine-specific demethylase 1 (LSD1), which represses transcription by inhibiting histone H3K4 methylation, thus demonstrating a negative feedback loop that limits endogenous AR expression. However, if cells are incubated in castration levels of androgens, AR expression in- creases. Furthermore, low levels of androgens in CRPC are suffi- cient to activate AR target genes, but are insufficient to suppress AR itself. Identification of androgen-regulated mIRNAs miRNAs are a class of naturally occurring, short noncoding RNAs that negatively regulate gene expression by binding to the 30 untranslated region (UTR) of mRNAs and inhibiting their translation. Recent research demonstrates that dysregulation of miRNA expression profiles contributes to pathogenesis of human malignancies. In prostate cancer, miRNAs regulated by androgen and AR have been identified using short RNA se- quence studies or miRNA microarray, and their functions for prostate cancer progression have been investigated. miR-21 miR-21 is an oncogene encoded at a fragile genomic location, 17q23.2, which is amplified in several types of tumors [19]. miR- 21 is an androgen-regulated miRNA and is induced by AR re- cruitment to its promoter in the presence of androgens (Figure 1). Expression analysis studies have shown that miR-21 expres- sion is associated with prostate cancer aggressiveness. Elevated miR-21 expression promotes androgen-dependent cell growth and castration-resistant tumor growth [20]. The target gene of miR-21 is MRCKS (myristoylated alanine rich protein kinase C substrate), which regulates cell motility, mitogenesis and plasma membrane trafficking [21]. Other factors repressed by miR-21 are B-cell translocation gene 2 (BTG2) and phosphatase and tensin homolog (PTEN) [22, 23]. miR-125b This miRNA is induced by androgen and stimulates the growth of prostate cancer cells (Figure 1). Expression of miR-125b is increased in prostate cancer samples compared with normal controls and correlates with Gleason scores, thus demonstrat- ing that miR-125b is an oncogenic miRNA that participates in the development of CRPC [24]. miR-125b targets the 30 UTR of apoptosis regulators such as p53, p53 downstream modulator of apoptosis (PUMA) and BCL2-antagonist/killer 1 (BAK1). Thus, dysregulation of miR-125b results in an imbalance between pro- and anti-apoptotic processes by targeting these pathways. Other androgen-regulated miRNAs (miR-141, -148a and -32) We have previously performed short RNA sequence analysis in LNCaP cells and reported several androgen-regulated miRNAs that promote cell proliferation including miR-141, miR-200a and miR-148a [25]. miR-148a is highly expressed in prostate cancer cell line derived from the lymph node (LNCaP) cells and targets cell cycle regulator cullin-associated and neddylation- dissociated 1 to promote cell proliferation (Figure 1). Expression of miR148a is upregulated in prostate cancer samples compared with normal tissues [26]. miR-32 is an androgen-regulated miRNA, which confers a growth advantage as demonstrated by reduction of apoptosis, and this effect is mediated by targeting BCL2 interacting media- tor (BIM), a pro-apoptotic member of the BCL2-antagonist/killer 1 (BCL2) family [27, 28]. In addition, miR-32 directly targets the 30 UTR of BTG2 to induce cell proliferation. Interestingly, both miR-32 and miR-148a are reported to be upregulated in CRPC samples, suggesting roles for both miRNAs in the emergence of castration resistance [27]. miR-29a/b Because the expression of the miR-29 family is reduced in sev- eral cancer tissues than in normal tissues, their role in cancer is controversial [29, 30]. While the precise role of the miR-29 fam- ily members in cancer is still unclear, they are known to pro- mote metastasis in breast cancer [31, 32]. Upregulated miR-29a enhances hepatoma migration by targeting PTEN [33], suggest- ing that miR-29 family may exhibit a dual function as a tumor Testosterone ARE AR Activation of transcriptionNuclear translocation Pri-miRNA Androgen responsive element Mature miRNAs Processing miR-21, miR-141, miR-148a miR-32, miR-125b, miR-29a/b Cell membrane DHT Anti- apoptosis Anti- apoptosis Epigenetic regulation Epigenetic regulation Cell cycleCell cycle Cell invasion Cell invasion Figure 1. AR functions as a nuclear receptor to regulate its target genes including miRNAs. AR translocates to the nucleus and binds AREs. AR epigenetically acti- vates transcription by recruiting coactivators for histone modifications. Primary miRNAs are induced by AR binding to its promoters. These transcripts are cleaved and processed to form mature miRNAs. Some miRNAs have been docu- mented to be regulated by AR and promote tumor growth. (A colour version of this figure is available online at: http://bfg.oxfordjournals.org) 258 | Takayama and Inoue Downloadedfromhttps://academic.oup.com/bfg/article-abstract/15/3/257/1742432bygueston10March2020
  • 3. suppressing and oncogenic miRNAs depending on cellular con- text and disease progression. We found that miR-29a/b is androgen-inducible miRNA tar- geting ten-eleven translocation 2 (TET2) and is highly expressed in cancers with poor prognoses. In vitro, miR-29 family members promote cell proliferation and migration of hormone refractory prostate cancer cells by activating cell motility and cell cycle- associated gene expression. In vivo, we showed that miR-29 family members enhance tumor growth using several AR antag- onists- and castration-resistant prostate cancer models. TET2 repression is associated with decreased 5-hydroxymethylation (5-hmC) in prostate cancer progression. Decreased 5-hmC pro- motes AR collaborating factor, forkhead box A1 (FOXA1) tran- scriptional activity and prostate cancer-associated expression of genes such as mTOR (Mammalian target of rapamycin). These results indicate a significant oncogenic role of miR-29 in pros- tate cancer progression by regulating epigenetic status [34]. AR-regulated miRNAs such as miR-21-125b-141 and -148a regulates apoptosis, cell cycle and cell proliferation by repress- ing various factors for tumor progression as AR downstream. In addition, our research highlights the epigenetic regulation by androgen-mediated miRNAs for activating FOXA1 and AR tran- scriptional activity and prostate cancer progression. These find- ings showed the important role of androgen-regulated miRNAs as tumor-promoting factors in prostate cancer. CRPC-associated mIRNAs In addition to androgen-induced miRNAs, oncogenic miRNAs for CRPC development have been reported. miR-221 and miR- 222 are transcribed from a cluster on chromosome X and are upregulated in CRPC tissues and cell lines [35]. Although andro- gen regulation of these miRNAs is unclear, miR-221 is reported to be repressed by androgen treatment [36]. Ectopic expression of miR-221 and -222 resulted in decreased sensitivity to andro- gen and induced androgen-independent growth. Both miRNAs target the expression of the important tumor suppressor gene p27kip1 [37]. HECT domain containing E3 ubiquitin ligase 2 (HECTD2) and Ras-related protein (RAB1A) are other miR-221 target genes. HECTD2 affected AR signaling, and its downregula- tion enhances androgen-independent cell growth [38]. These findings suggest the important roles of miR-221/222 in cell cycle regulation and AR activation for CRPC development. However, the functions of miR-221/22 remain to be determined because another study has shown the tumor-suppressive function of miR-221 and -222 by targeting oncogene Ecm29 [39]. Tumor-suppressive miRNAs in prostate cancer Several studies have revealed the importance of miRNAs as tumor-suppressive factors targeting oncogenic signaling. Recent advances of genomic biology have identified important roles of miRNAs repressed in prostate cancer development by genomic loss. miR-101 miR-101 is significantly reduced in prostate cancer, and its ex- pression is further decreased during cancer progression because the genomic locus encoding miR-101 is found to be significantly lost in metastatic prostate cancers. Interestingly, loss of miR- 101 is associated with overexpression of enhancer of zeste homolog 2 (EZH2), a polycomb group protein, whose primary function is to trimethylate histone H3 lysine 27 (H3K27) at gene promoters [40]. miR-15/16 miR-15 and -16 are tumor suppressors likely involved in pros- tate cancer development at multiple levels. Both miRNAs are located at a fragile chromosomal locus, 13q14, in which gen- omic loss is reportedly associated with clinically advanced pros- tate cancer [41]. Delivery of miR-15 and -16 suppressed tumor formation from nontumorigenic cells in severe combined immunodeficiency (SCID) mice [42]. These miRNAs control cell survival, proliferation and invasion via deregulation of onco- genes such as BCL2, cyclinD1 and WNT3A (wingless type MMTV integration site family). Thus, in addition to AR-mediated pathways, these tumor- suppressive miRNAs possesses other critical roles for progres- sion to metastatic or advanced prostate cancer. Integrative analysis to discover androgen- regulated lncRNAs Next-generation sequencing has been used to analyze the tran- scriptome of prostate cancer cells. Combined analyses of gen- ome-wide ARBSs obtained by chromatin immunoprecipitation (ChIP)-based analyses such as ChIP-chip [43–48] or ChIP-seq [49–51] and the androgen-regulated transcriptome have re- cently been reported. A new technique, global nuclear run-on sequencing (GRO-seq), has been used to analyze sequential gene expression on androgen treatment [52]. For nuclear run-on reactions, cell nuclei are isolated after treatment with andro- gens for a specific amount of time. In the run-on step, RNA poly- merases are allowed to run on about 100 bases in the presence of a ribonucleotide analog [5-bromouridine 50 -triphosphate (BrUTP)]. BrU-containing RNA is selected by purification with an antibody specific for the nucleotide analog. A cDNA library is then prepared for next-generation sequencing. Using this method, the authors discovered the production of enhancer- templated noncoding RNAs (enhancer RNA, eRNAs). Cap analysis of gene expression (CAGE) [53] is a high- throughput method for analysis of gene expression and to pro- file transcription start sites (TSS), including those for promoter usage. CAGE is based on sequencing of concatamers of DNA tags derived from the initial 20 nucleotides at the 50 ends of mRNAs. The frequency of CAGE tags is consistent with results from other analyses, such as microarrays. This analysis is high- throughput, and enables understanding of gene networks via correlation between promoter usage and expression of gene transcription factors. We performed CAGE to determine andro- gen-regulated TSSs and ChIP-chip analysis to identify genome- wide ARBSs and histone H3 acetylated sites in the complete human genome [54]. Using CAGE, we identified 13,110 distinct, androgen-regulated TSSs. Cross-referencing with the gene ex- pression database for prostate cancer (Oncomine), the majority of androgen-upregulated genes containing adjacent ARBSs and CAGE tag clusters in our study were previously confirmed as upregulated genes in prostate cancer. The integrated high- throughput genome analyses of CAGE and ChIP-chip provide useful information for elucidating the AR-mediated transcrip- tional network that contributes to the development and pro- gression of prostate cancer. ncRNAs, including miRNAs, were also identified as androgen target transcripts in this study. We found many androgen-dependent TSSs widely distributed throughout the genome, including in the antisense (AS) Emerging role of noncoding RNA in prostate cancer progression | 259 Downloadedfromhttps://academic.oup.com/bfg/article-abstract/15/3/257/1742432bygueston10March2020
  • 4. direction of RefSeq genes. Several pairs of sense/AS promoters were newly identified within single RefSeq gene regions, sug- gesting the involvement of AS ncRNA in transcriptional regulation. Functional analysis of as ncRNA in prostate cancer Global transcriptome analysis revealed that the majority of the mammalian genome can generate transcripts from both strands of the DNA double helix, and identified paired sense/AS tran- scripts, indicating that AS transcription is important for gene regulation [55–57]. One example of AS transcripts is the cyclin-dependent kinase (CDKN) 2B antisense RNA1 (CDKN2B-AS1) [58], which is a natural AS transcript located within the CDKN2A/CDKN2B tumor suppressor locus on 9q21.3. Cycline dependent kinase (CDK) inhibitors, p14, p15(CDKN2B) and p16(CDKN2A) are produced from this locus and regulate cell cycle progression at G1/S. CDKN2B is specifically silenced by CDKN2B-AS1 through heterochromatin formation, by increasing dimethylation of H3K9 and demethylation of H3K4 at the gene promoter. Another study showed the coordinated upregulation of ANRIL (antisense noncoding RNA in the INK4 locus) and chromo- box7 (CBX7), a member of Polycomb repressive complex 1 in prostate cancer. CBX7 interacts with ANRIL at the promoters of CDKN2A/2B for histone H3K27 methylation [59]. There are two potential mechanisms of sense-transcript re- pression: (1) post-transcriptional repression [X-inactive specific transcript (XIST) repression by XIST antisense RNA (TSIX)] by degradation [60], and (2) epigenetic regulation by recruitment of transcription factors. The interaction of lncRNAs with chroma- tin remodeling complexes induces heterochromatin formation in specific loci, leading to reduced target gene expression (Figure 2). For example, HOX Antisense Intergenic RNA (HOTAIR) [61], a 2.2 kb ncRNA, is transcribed in the AS direction from the HOXC gene (homeobox gene) cluster. HOTAIR functions in trans by interacting with and by recruiting the polycomb repressive complex 2 to the HOXD locus, resulting in transcriptional silenc- ing across a 40 kb region. In addition, HOTAIR interacts with a second histone modification complex, the LSD1/corepressor for Rest (CoREST)/RE1 silencing transcription factor (REST) complex, which coordinates targeting of polycomb repressive complex2 (PRC2) and LSD1 to chromatin for coupled histone H3K27 methylation and K4 demethylation [62]. HOTAIR expression is associated with hormone-dependent cancer such as breast and prostate cancer. HOTAIR expression level was higher in breast tumors than in normal breast epithelia, and high HOTAIR ex- pression is associated with poor prognosis of patients. This clin- ical finding also reflects the HOTAIR-mediated regulation of hormone action. Although the regulation of HOTAIR by estrogen is controversial [63, 64], HOTAIR enhanced estrogen receptor (ER) protein level and enhanced downstream signaling [64]. Moreover, HOTAIR is repressed by androgen and upregulated by hormone deprivation. In addition, HOTAIR binds to AR and blocked E3-ubiquitine ligase murine double minute 2 (MDM2) to stabilize AR and enhance the AR-mediated transcriptional ac- tivity and drives CRPC development [65]. We investigated the functional role of the novel androgen- responsive AS RNA, CTBP1-AS [66]. We focused on the mechan- ism of androgen-mediated cancer proliferation by this lncRNA and the clinical significance in prostate cancer progression. We showed that C-terminal binding protein 1 (CTBP1) [67] is a tran- scriptional corepressor of AR and negatively regulates androgen signaling. CTBP1-AS is directly upregulated by AR and interacts with RNA binding protein, PSF [68, 69], for transcriptional re- pression using HDACs. Downstream signals of CTBP1-AS/PSF (polypyrimidine tract binding protein associated splicing factor) complex include cell cycle regulators such as p53 or SMAD3 in addition to CTBP1. Castration-resistant tumor growth was pro- moted by androgen-mediated repression of these cell cycle regulators and activation of AR signaling. Activation of AR transcription activity by lncRNAs In addition to CTBP1-AS and HOTAIR, several lncRNAs have been reported to regulate AR activity for prostate cancer progression. Steroid receptor RNA activator Steroid receptor RNA activator (SRA) interacts with nuclear re- ceptors, including AR, ER, progesterone receptor, glucocorticoid receptor (GR) and thyroid hormone receptor, and regulates their transcriptional activities. As a mechanism for activation, SRA interacts with a nuclear receptor coactivator SRC-1 (steroid re- ceptor coactivator). Mutation analysis revealed that six stem- loop motifs are important for coactivation by SRA [70, 71]. Aberrant SRA transcription was observed in various tumors. It is upregulated in tumors such as breast cancer, ovarian cancer and prostate cancer, compared with normal tissues. In prostate cancer, SRA and its alternative splicing form, steroid receptor RNA activator protein (SRAP), were shown to be required for ex- pression of AR target genes in the presence of androgens [72, 73]. Epigenetic and transcriptional regulation SWI/ SNF Epigenetic control PSF Deacethylation Chromatin remodeling Enhancer action eRNA Promoter enhancer Loop formation CTBP1-AS Post-transcriptional regulation miRNA synthesis Regulation of mRNA AAAAA SChLAP1 PCGEM1 PNCNR1 SRA MALAT1 miR-675 H19 TGFBI AR Inhibition of AR binding GAS5 AR HOTAIR MDM2 Enhancement of AR binding Figure 2. Representative mechanisms of lncRNAs in promoting prostate cancer. (1) Transcriptional regulation by epigenetic mechanisms (interaction with epi- genetic factors or accelerating AR transcriptional activity by direct interaction) and (2) posttranscriptional regulation (modulating the target mRNA stability or AR protein) have been reported. (A colour version of this figure is available online at: http://bfg.oxfordjournals.org) 260 | Takayama and Inoue Downloadedfromhttps://academic.oup.com/bfg/article-abstract/15/3/257/1742432bygueston10March2020
  • 5. Enhancer RNA In AR signals, AR-activated enhancers marked by increased eRNA are required for activation of genes in their vicinity. eRNA knockdown disrupts ligand-induced promoter–enhancer inter- action. As a mechanism of enhancer–promoter interaction, eRNA interacts with a cohesion complex and this interaction is required for its recruitment to enhancers [74]. Prostate cancer gene expression marker 1 and Prostate cancer noncoding RNA 1 Prostate cancer gene expression marker 1 (PCGEM1) was originally identified as a prostate tissue-specific long ncRNA [75]. It is involved in inhibition of apoptosis by delaying p53 and p21 in- duction in an androgen-dependent manner [76]. PCGEM1 is overexpressed in at least half of all prostate tumors. LncRNA prostate cancer noncoding RNA 1 (PRNCR1) was found by a single nucleotide polymorphism (SNP) associated with prostate cancer susceptibility. Two SNPs, rs1456315 and rs7463708 in the chromosome 8q24, which is a gene desert region, were shown to be most significantly associated with prostate cancer suscep- tibility. PCGEM1 and PRNCR1 are both involved in AR-mediated gene transcription [77]. Knockdown of these lncRNAs inhibits AR target gene regulation in a DHT-dependent manner and is associated with tumor growth in CRPC xenograft model. Interactions of PCGEM1 and PRNCR1 with AR were confirmed by RNA pull down and RNA immunoprecipitation assay assays. For the mechanism of AR activation, PCGEM1 was shown to interact with pygopus homolog2 (Pygo2), and PRNCR1 interacts with DOT1-like histone H3 methyltransferase for AR methylation. By 3C-ChIP experiments, these two lncRNAs were found to be required for AR-bound enhancer promoter loop formation. However, this mechanism is still controversial because another study failed to reproduce the interaction of AR with these two lncRNAs [78]. Growth arrest-specific 5 Growth arrest-specific 5 (GAS5) interacts with GR and sup- presses GR-mediated transcriptional activity. The interaction of GAS5 and GR reduces GR binding to GR-responsive elements (GREs). The binding element of GAS5 with GR contains GRE-like sequences, suggesting a competing role of GAS5 with GRE DNA for GR binding. GAS5 also binds to other nuclear receptors such as AR. GAS5 was found to be downregulated in breast cancer and prostate cancer and then inhibit cell proliferation [79]. However, GAS5 inhibition of ERa transcriptional activity has not been found. Noncoding multiple small nuclear RNAs (snoRNAs) encoded in introns of GAS5, particularly U50, showed tumor suppressor characteristics in breast cancer [80]. Functional lncRNA extends the insight of regulatory mech- anism of AR. The major mechanism is the direct interaction with AR and subsequent modulation of cofactors (SRA), stability of AR protein itself (HOTAIR), inhibition of genomic binding (GAS5) and enhancer promoter loop forming (eRNA, PCGEM1 and PRNCR1). Regulation of co-repressor or co-activator by epi- genetic action of androgen-regulated lncRNA (CTBP1-AS) would be another mechanism to enhance AR activity in prostate can- cer. However, the number of lncRNAs described in detail is small. We have to investigate the tissue-, spatial-specific func- tion of lncRNAs or the secondary structure or high-level struc- ture to determine the interaction and recruitment to specific genomic regions. Molecular mechanism of lncRNAs in prostate cancer Some lncRNAs are not related to the function of AR, but play an important role in prostate cancer progression. These lncRNAs could serve as cancer markers for clinical diagnosis. Metastasis-associated lung adenocarcinoma transcript 1 Metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) is involved in pre-mRNA processing and regulates the alternative splicing of pre-mRNA by modulating levels of serine/arginine splicing factors. Although the mechanism of MALAT1 upregula- tion in cancer cells is still unclear, multiple copy-number gains at the locus and chromosomal translocations have been re- ported [81]. In prostate cancer, MALAT1 overexpression is corre- lated with poor prognosis and cancer progression [82]. The regulation of MALAT1 by androgen has not been reported, and MALAT1 is upregulated in primary breast cancer and inhibited by estrogen treatment. MALAT1 knockdown inhibits breast can- cer cell proliferation, suggesting the tumor-promoting effect of MALAT1 in breast cancer [83, 84]. Thus, MALAT1 may play a crit- ical role in hormone-dependent cancer development. Prostate cancer-associated ncRNA transcript-1 Prostate cancer-associated ncRNA transcript-1 (PCAT-1) is a rela- tively small intergenic ncRNA located in the 8q24 gene desert, and it was identified through global transcriptomic sequencing of prostate tumors [85]. PCAT-1 expression is inversely corre- lated with expression of the EZH2, a histone methyltransferase that encodes components of PRC2 and a marker for prostate cancer progression; thus, PRC2 represses PCAT-1 expression. PCAT-1 induces cell proliferation in vitro and has a predomin- antly repressive effect on gene expression, most notably on the expression of the tumor suppressor gene breast cancer 2 (BRCA2) [86]. PCAT-1 also interacts with the SUZ12 component of PRC2. Second chromosome locus associated with prostate 1 Second chromosome locus associated with prostate 1 (SChLAP1) is an lncRNA transcribed from within an intergenic gene desert on 2q31.1. It was originally identified in an analysis of intergenic lncRNAs that are selectively upregulated in aggressive prostate cancer samples. SChLAP1 is highly expressed in 25% of prostate tumors and shows increased expression in metastatic cancer cells. SChLAP1 is involved in the regulation of the switch/su- crose nonfermenting (SWI/SNF) complex (Figure 2). This com- plex canonically controls transcription by using ATP hydrolysis to remodel chromatin and physically mobilize nucleosomes, particularly at the gene promoters. SChLAP1 co-immunoprecipi- tates with SNF5 and prevents its genomic binding, thus antago- nizing tumor-suppressive SWI/SNF-mediated gene regulation [87]. Using another cohort of 1008 patients, SChLAP1 was identi- fied and validated as the highest-ranked overexpressed gene in cancer with metastatic progression [88]. These findings demon- strated the usefulness of SChLAP1 as a biomarker of prostate cancer progression. Thus, both PCAT1-1 and SChLAP1 can be useful biomarkers for clinically detecting advanced prostate cancer. Emerging role of noncoding RNA in prostate cancer progression | 261 Downloadedfromhttps://academic.oup.com/bfg/article-abstract/15/3/257/1742432bygueston10March2020
  • 6. Prostate cancer gene 3 Prostate cancer gene 3 (PCA3) is an lncRNA associated with pros- tate cancer and is a potentially useful biomarker [89, 90]. It was originally discovered in 1999 by a differential display analysis of prostate tissues and cell lines. Its expression in 95% of the pros- tate tumors is up to 100-fold higher than that in adjacent non-neoplastic tissues. PCA3 regulates the expression of its overlapping gene PRUNE2, a tumor suppressor gene in prostate cancer, by posttranscriptionally regulating the stability of mRNA [91]. Urinary measurement of PCA3 RNA levels can be helpful to detect prostate cancer with superior tumor specificity to prostate-specific antigen [92, 93]. H19 This lncRNA is transcribed from the H19/Igf2 gene cluster on chromosome 11p11.5. Some evidence suggests H19 plays a crit- ical role in tumor progression by exhibiting oncogenic activity in some types of tumors, such as bladder cancer. However, H19 was shown to have a tumor-suppressive role in metastatic pros- tate cancer by repression of TGFb1 signals, which is involved in metastasis. H19 and miR-675, derived from H19, were downre- gulated in metastatic cancers. miR-675 inhibits tumor growth factor beta 1 (TGFb1) signals by directly binding to the 30 UTR of TGFb1. Overexpression of miR-675 and H19 repressed prostate cancer cell migration [94]. In contrast, miR-675 enhances tumorigenesis and metastasis of breast cancer cells [95]. H19 mediates estrogen-dependent cell proliferation in breast cancer cell, suggesting the oncogenic role of H19 in estrogen-depend- ent breast cancer [96]. These lncRNAs are likely to function through interaction with general epigenetic and chromatin remodeling factor (PCAT-1, SChLAP1) to impact the transcription for cancer pro- gress. Modulation of miRNAs (H19) or snoRNAs (GAS5) would be important regulatory mechanism of lncRNAs in cancer. Regulation of stability or splicing by lncRNAs (PCA3 and MALAT1) broadens the insight of transcriptional regulation in cell biology. Future plans for clinical applications of ncRNA Long ncRNAs or miRNAs can be used as diagnostic and prognos- tic markers, and as novel specific therapeutic targets for CRPC. Current prostate cancer genomic data can be fully exploited if noncoding regions are studied in detail. In addition, ncRNAs are interesting targets in cancer therapy because their cancer- and tissue-specific expression can be a major advantage over other therapeutic options. Several other studies have also demon- strated the clinical significance of ncRNAs. However, before we can make use of these new therapeutic options [97], many more functional and structural studies will be required. The exponen- tially growing number of studies reporting new ncRNAs or andro- gen signaling molecules will contribute to progress in this field. Key Points • Oncogenic miRNAs (miR-21, 29a/b, 148a and 125b) are androgen-regulated and regulate tumor growth by tar- geting epigenetic status, apoptosis and cell cycle regulators. • High-throughput analysis using RNA-seq, GRO-seq and CAGE has identified abundant androgen-regulated and prostate cancer-associated lncRNAs. • Androgen-regulated AS RNA, CTBP1-AS, regulates glo- bal androgen signals and cell cycle regulators by asso- ciating with RNA-biding epigenetic complexes. • RNA-seq analyses of tumor samples have identified lncRNAs such as PCAT-1 and SChLAP1 that could be biomarkers for prostate cancer. • AR interacts with some lncRNAs such as HOTAIR, eRNAs, SRA, PCGEM and PNCR1 to regulate AR tran- scriptional activity and promote prostate cancer. Funding This work was supported by Grants of the P-Direct (S.I.) from the MEXT, Japan; by Grants (S.I. and K.T.) from the JSPS, Japan; by Grants from Takeda Science Foundation (S.I. and K.T.), Japan; by a Grant from Mochida Memorial Research Foundation (K.T), Japan. References 1. Gutschner T, Diederichs S. The hallmarks of cancer: a long non-coding RNA point of view. RNA Biol 2012;9:703–19. 2. Djebali S, Davis CA, Merkel A, et al. Landscape of transcription in human cells. Nature 2012;489:101–8. 3. Mercer TR, Mattick JS, et al. Structure and function of long noncoding RNAs in epigenetic regulation. Nat Struct Mol Biol 2013;20:300–7. 4. Derrien T, Johnson R, Bussotti G, et al. The GENCODE v7 cata- log of human long noncoding RNAs: analysis of their gene structure, evolution, and expression. Genome Res 2012;22:1775–89. 5. Du Z, Fei T, Verhaak RG, et al. Integrative genomic analyses reveal clinically relevant long noncoding RNAs in human cancer. Nat Struct Mol Biol 2013;20:908–13. 6. Kan Z, Jaiswal BS, Stinson J, et al. Diverse somatic mutation patterns and pathway alterations in human cancers. Nature 2010;466:869–73. 7. Moran VA, Perera RJ, Khalil AM, et al. Emerging functional and mechanistic paradigms of mammalian long non-coding RNAs. Nucleic Acids Res 2012;40:6391–400. 8. Debes JD, Tindall DJ. Mechanism of androgen-refractory pros- tate cancer. N Eng J Med 2004;351:1488–90. 9. Balk SP, Knudsen KE. AR, the cell cycle, and prostate cancer. Nucl Recept Signal 2008;6:e001. 10.Schmidt LJ, Tindall DJ. Steroid 5 alpha-reductase inhibitors targeting BPH and prostate cancer. J Steroid Biochem Mol Biol 2011;125:32–8. 11.Zhou ZX, Lane MV, Kemppainen JA, et al. Specificity of ligand- dependent androgen receptor stabilization: receptor domain interactions influence ligand dissociation and receptor stabil- ity. Mol Endocrinol 1995;9:208–18. 12.Takayama K, Inoue S. Transcriptional network of androgen receptor in prostate cancer progression. Int J Urol 2013;20(8):756–68. 13.Shang Y Myers M, Brown M. Formation of the androgen re- ceptor transcription complex. Mol Cell 2002;9:601–10. 14.Chen CD, Welsbie DS, Tran C, et al. Molecular determinants of resistance to antiandrogen therapy. Nat Med 2004;10:33–9. 15.Waltering KK, Helenius MA, Sahu B, et al. Increased expres- sion of androgen receptor sensitizes prostate cancer cells to low levels of androgens. Cancer Res 2009;69:8141–9. 16.Locke JA, Guns ES, Lubik AA, et al. Androgen levels increase by intratumoral de novo steroidogenesis during progression 262 | Takayama and Inoue Downloadedfromhttps://academic.oup.com/bfg/article-abstract/15/3/257/1742432bygueston10March2020
  • 7. of castration-resistant prostate cancer. Cancer Res 2008;68:6407–15. 17.Sun S, Sprenger CC, Vessella RL, et al. Castration resistance in human prostate cancer is conferred by a frequently occurring androgen receptor splice variant. J Clin Invest 2010;120:2715–30. 18.Cai C, He HH, Chen S, et al. Androgen receptor gene expres- sion in prostate cancer is directly suppressed by the androgen receptor through recruitment of lysine-specific demethylase 1. Cancer Cell 2011;20:457–71. 19.Kichevsky AM, Gabriely G. miR-21: small multi-faceted RNA. J Cell Mol Med 2009;13:39–53. 20.Ribas J, Ni X, Haffner M, et al. miR-21: an androgen receptor- regulated microRNA that promotes hormone-dependent and hormone-independent prostate cancer growth. Cancer Res 2009;69:7165–9. 21.Li T, Li D, Sha J, et al. MicroRNA-21 directly targets MARCKS and promotes apoptosis resistance and invasion in prostate cancer cells. Biochem Biophys Res Commun 2009;383:280–5. 22.Coppola V, Musumeci M, Patrizii M, et al. BTG2 loss and miR- 21 upregulation contribute to prostate cell transformation by inducing luminal markers expression and epithelial- mesenchymal transition. Oncogene 2013;32:1843–53. 23.Liu LZ, Li C, Chen Q, et al. MiR-21 induced angiogenesis through AKT and ERK activation and HIF-1a expression. PLoS One 2011;6:e19139. 24.Shi XB, Xue L, Yang J, et al. An androgen-regulated miRNA suppresses Bak1 expression and induces androgen- independent growth of prostate cancer cells. Proc Natl Acad Sci USA 2007;104:19983–8. 25.Murata T, Takayama K, Katayama S, et al. miR-148a is an an- drogen-responsive microRNA that promotes LNCaP prostate cell growth by repressing its target CAND1 expression. Prostate Cancer Prostatic Dis 2010;13:356–61. 26.Walter BA, Valera VA, Pinto PA, et al. Comprehensive microRNA profiling of prostate cancer. J Cancer 2013;4:350–7. 27.Jalava SE, Urbanucci A, Latonen L, et al. Androgen-regulated miR-32 targets BTG2 and is overexpressed in castration- resistant prostate cancer. Oncogene 2012;31:4460–71. 28.Ambs S, Prueitt RL, Yi M, et al. Genomic profiling of microRNA and messenger RNA reveals deregulated microRNA expres- sion in prostate cancer. Cancer Res 2008;68:6162–70. 29.Wang Y, Zhang X, Li H, et al. The role of miRNA-29 family in cancer. Eur J Cell Biol 2013;92(3):123–8. 30.Jiang H, Zhang G, Wu JH, et al. Diverse roles of miR-29 in can- cer. Oncol Rep 2014;31(4):1509–16. 31.Wang C, Bian Z, Wei D, et al. miR-29b regulates migration of human breast cancer cells. Mol Cell Biochem 2011;352(1–2): 197–207. 32.Gebeshuber CA, Zatloukal K, Martinez J. miR-29a suppresses tristetraprolin, which is a regulator of epithelial polarity and metastasis. EMBO Rep 2009;10(4):400–5. 33.Kong G, Zhang J, Zhang S, et al. Upregulated microRNA-29a by hepatitis B virus X protein enhances hepatoma cell migration by targeting PTEN in cell culture model. PLoS One 2011;6(5):e19518. 34.Takayama K, Misawa A, Suzuki T, et al. TET2 repression by androgen hormone regulates global hydroxymethylation status and prostate cancer progression. Nat Commun 2015;6:8219. 35.Sun T, Wang Q, Balk S, et al. The role of microRNA-221 and microRNA-222 in androgen-independent prostate cancer cell lines. Cancer Res 2009;69:3356–63. 36.Segal CV, Koufaris C, Powell C, et al. Effects of treatment with androgen receptor ligands on microRNA expression of pros- tate cancer cells. Toxicology 2015;333:45–52. 37.Galardi S, Mercatelli N, Giorda E, et al. miR-221 and miR-222 expression affects the proliferation potential of human pros- tate carcinoma cell lines by targeting p27Kip1. J Biol Chem 2007;282:23716–24. 38.Sun T, Wang X, He HH, et al. MiR-221 promotes the develop- ment of androgen independence in prostate cancer cells via downregulation of HECTD2 and RAB1A. Oncogene 2014;33(21):2790–800. 39.Goto Y, Kojima S, Nishikawa R, et al. MicroRNA expression signature of castration-resistant prostate cancer: the microRNA-221/222 cluster functions as a tumour suppressor and disease progression marker. Br J Cancer 2015;113(7):1055–65. 40.Varambally S, Cao Q, Mani RS, et al. Genomic loss of microRNA-101 leads to overexpression of histone methyl- transferase EZH2 in cancer. Science 2008;322:1695–9. 41.Melamed J, Einhorn JM, Ittmann MM. Allelic loss on chromo- some 13q in human prostate carcinoma. Clin Cancer Res 1997;3:1867–72. 42.Bonci D, Coppola V, Musumeci M, et al. The miR-15a-miR-16-1 cluster controls prostate cancer by targeting multiple onco- genic activities. Nat Med 2008;14:1271–7. 43.Wang Q, Li W, Zhang Y, et al. Androgen receptor regulates a distinct transcription program in androgen-independent prostate cancer. Cell 2009;138:245–56. 44.Takayama K, Kaneshiro K, Tsutsumi S, et al. Identification of novel androgen response genes in prostate cancer cells by coupling chromatin immunoprecipitation and genomic microarray analysis. Oncogene 2007;26:4453–63. 45.Takayama K, Tsutsumi S, Horie-Inoue K, et al. Amyloid pre- cursor protein is a primary androgen target gene that pro- motes prostate cancer growth. Cancer Res 2009;69:137–42. 46.Wang Q, Li W, Liu XS, et al. A hierarchical network of tran- scription factors governs androgen receptor-dependent pros- tate cancer growth. Mol Cell 2007;27:380–92. 47.Jia L, Berman BP, Jariwala U, et al. Genomic androgen recep- tor-occupied regions with different functions, defined by his- tone acetylation, coregulators and transcriptional capacity. PLoS One 2008;3:e3645. 48.Massie CE, Adryan B, Barbosa-Morais NL, et al. New androgen receptor genomic targets show an interaction with the ETS1 transcription factor. EMBO Rep 2007;8:871–8. 49.Yu J, Yu J, Mani RS, et al. An integrated network of androgen receptor, polycomb, and TMPRSS2-ERG gene fusions in pros- tate cancer progression. Cancer Cell 2010;17:443–54. 50.Urbanucci A, Sahu B, Seppa¨la¨ J, et al. Overexpression of an- drogen receptor enhances the binding of the receptor to the chromatin in prostate cancer. Oncogene 2012;31:2153–63. 51.Tan PY, Chang CW, Chng KR, et al. Integration of regulatory networks by NKX3-1 promotes androgen-dependent prostate cancer survival. Mol Cell Biol 2012;32:399–414. 52.Wang D, Garcia-Bassets I, Benner C, et al. Reprogramming transcription by distinct classes of enhancers functionally defined by eRNA. Nature 2011;474:390–4. 53.Shiraki T, Kondo S, Katayama S, et al. Cap analysis gene ex- pression for high-throughput analysis of transcriptional starting point and identification of promoter usage. Proc Natl Acad Sci USA 2003;100:15776–81. 54.Takayama K, Katayama S, Horie-Inoue K, et al. Integration of cap analysis of gene expression and chromatin Emerging role of noncoding RNA in prostate cancer progression | 263 Downloadedfromhttps://academic.oup.com/bfg/article-abstract/15/3/257/1742432bygueston10March2020
  • 8. immunoprecipitation analysis on array reveals genome-wide androgen receptor signaling in prostate cancer cells. Oncogene 2011;30(5):619–30. 55.Carninci P, Kasukawa T, Katayama S, et al. The transcriptional landscape of the mammalian genome. Science 2005;309:1559–63. 56.Katayama S, Tomaru Y, Kasukawa T, et al. Antisense tran- scription in the mammalian transcriptome. Science 2005;309:1564–6. 57.Rosok O, Sioud M. Systematic identification of sense- antisense transcripts in mammalian cells. Nat Biotechnol 2004;22:104–8. 58.Yu W, Gius D, Onyango P, et al. Epigenetic silencing of tumour suppressor gene p15 by its antisense RNA. Nature 2008;451:202–6. 59.Yap KL, Li S, Mun˜ oz-Cabello AM, et al. Molecular interplay of the noncoding RNA ANRIL and methylated histone H3 lysine 27 by polycomb CBX7 in transcriptional silencing of INK4a. Mol Cell 2010;38:662–74. 60.Ogawa Y, Sun BK, Lee JT, et al. Intersect, ion of the RNA inter- ference and X-inactivation pathways. Science 2008;320: 1336–41. 61.Gupta RA, Shah N, Wang KC, et al. Long non-coding RNA HOTAIR reprograms chromatin state to promote cancer me- tastasis. Nature 2010;464:1071–6. 62.Tsai MC, Manor O, Wan Y, et al. Long noncoding RNA as modular scaffold of histone modification complexes. Science 2010;329(5992):689–93. 63.Bhan A, Hussain I, Ansari KI, et al. Antisense transcript long noncoding RNA (lncRNA) HOTAIR is transcriptionally induced by estradiol. J Mol Biol 2013;425(19):3707–22. 64.Xue X, Yang YA, Zhang A, et al. LncRNA HOTAIR enhances ER signaling and confers tamoxifen resistance in breast cancer. Oncogene 2015. doi: 10.1038/onc.2015.340. 65.Zhang A, Zhao JC, Kim J, et al. LncRNA HOTAIR enhances the androgen-receptor-mediated transcriptional program and drives castration-resistant prostate cancer. Cell Rep 2015;13(1):209–21. 66.Takayama K, Horie-Inoue K, Katayama K, et al. Androgen- responsive long noncoding RNA CTBP1-AS promotes prostate cancer. EMBO J 2013;32:1665–80. 67.Shi Y, Sawada J, Sui G, et al. Coordinated histone modifica- tions mediated by a CtBP co-repressor complex. Nature 2003;422:735–8. 68.Shav-Tal Y, Zipori D. PSF and p54(nrb)/NonO–multi- functional nuclear proteins. FEBS Lett 2002;531:109–14. 69.Song X, Sui A, Garen A. Binding of mouse VL30 retrotrans- poson RNA to PSF protein induces genes repressed by PSF: ef- fects on steroidogenesis and oncogenesis. Proc Natl Acad Sci USA 2004;101:621–6. 70.Lanz RB, McKenna NJ, Onate SA, et al. steroid receptor coacti- vator, SRA, functions as an RNA and is present in an SRC-1 complex. Cell 1999;97(1):17–27. 71.Lanz RB, Razani B, Goldberg AD, et al. Distinct RNA motifs are important for coactivation of steroid hormone receptors by steroid receptor RNA activator (SRA). Proc Natl Acad Sci USA 2002;99(25):16081–6. 72.Kawashima H, Takano H, Sugita S, et al. A novel steroid recep- tor co-activator protein (SRAP) as an alternative form of ster- oid receptor RNA-activator gene: expression in prostate cancer cells and enhancement of androgen receptor activity. Biochem J 2003;369(Pt 1):163–71. 73.Agoulink IU, Weigel NL. Coactivator selective regulation of androgen receptor activity. Steroids 2009;74:669–74. 74.Hsieh CL, Fei T, Chen Y, et al. Enhancer RNAs participate in androgen receptor-driven looping that selectively enhances gene activation. Proc Natl Acad Sci USA 2014;111(20):7319–24. 75.Srikantan V Zou Z, Petrovics G, et al. PCGEM1, a prostate- specific gene, is overexpressed in prostate cancer. Proc Natl Acad Sci USA 2000;97:12216–21. 76.Petrovics G, Zhang W, Makarem M, et al. Elevated expression of PCGEM1, a prostate-specific gene with cell growth- promoting function, is associated with high-risk prostate cancer patients. Oncogene 2004;23:605–11. 77.Yang L, Lin C, Jin C, et al. lncRNA-dependent mechanisms of androgen-receptor regulated gene activation programs. Nature 2013;500:598–602. 78.Prensner JR, Sahu A, Iyer MK, et al. The IncRNAs PCGEM1 and PRNCR1 are not implicated in castration resistant prostate cancer. Oncotarget 2014;5(6):1434–8. 79.Pickard MR, Mourtada-Maarabouni M, Williams GT, et al. Long non-coding RNA GAS5 regulates apoptosis in pros- tate cancer cell lines. Biochem Biophys Acta 2013;1832: 1613–23. 80.Mourtada-Maarabouni M, Pickard MR, Hedge VL, et al. GAS5, a non-protein-coding RNA, controls apoptosis and is downre- gulated in breast cancer. Oncogene 2009;8(2):195–208. 81.Paris PL, Andaya A, Fridlyand J, et al. Whole genome scanning identifies genotypes associated with recurrence and metasta- sis in prostate tumor. Hum Mol Genet 2004;13:1303–13. 82.Ren S, Liu Y, Xu W, et al. Long noncoding RNA MALAT-1 is a new potential therapeutic target for castration resistant pros- tate cancer. J Urol 2013;190:2278–87. 83.Guffanti A, Iacono M, Pelucchi P, et al. A transcriptional sketch of a primary human breast cancer by 454 deep sequencing, BMC Genomics 2009;10:163. 84.Zhao Z, Chen C, Liu Y, et al. 17beta-Estradiol treatment in- hibits breast cell proliferation, migration and invasion by decreasing MALAT-1 RNA level. Biochem Biophys Res Commun 2014;445:388–93. 85.Prensner JR, Iyer MK, Balbin OA, et al. Transcriptome sequenc- ing across a prostate cancer cohort identifies PCAT-1, an unannotated lincRNA implicated in disease progression. Nat Biotechnol 2011;29(8):742–9. 86.Prensner JR, Chen W, Iyer MK, et al. PCAT-1, a long noncoding RNA, regulates BRCA2 and controls homologous recombin- ation in cancer. Cancer Res 2014;74(6):1651–60. 87.Prensner JR, Iyer MK, Sahu A, et al. The long noncoding RNA SChLAP1 promotes aggressive prostate cancer and antagonizes the SWI/SNF complex. Nat Genet 2013;45(11): 1392–8. 88.Prensner JR, Zhao S, Erho N, et al. RNA biomarkers associated with metastatic progression in prostate cancer: a multi- institutional high-throughput analysis of SChLAP1. Lancet Oncol 2014;15(13):1469–80. 89.Bussemakers MJ, et al. DD3: a new prostate-specific gene, highly overexpressed in prostate cancer. Cancer Res 1999;59:5975–9. 90.Ferreira LB, van Bokhoven A, Verhaegh GW, et al. PCA3 non- coding RNA is involved in the control of prostate-cancer cell survival and modulates androgen receptor signaling. BMC Cancer 2013;12:507. 91.Salameh A, Lee AK, Cardo-Vila M, et al. PRUNE2 is a human prostate cancer suppressor regulated by the intronic long noncoding RNA PCA3. Proc Natl Acad Sci USA 2015;112(27): 8403–8. 92.Hessels D, Schalken JA. The use of PCA3 in the diagnosis of prostate cancer. Nat Rev Urol 2009;6:255–61. 264 | Takayama and Inoue Downloadedfromhttps://academic.oup.com/bfg/article-abstract/15/3/257/1742432bygueston10March2020
  • 9. 93.Lee GL, Dobi A, Srivastava S. Prostate cancer: diagnostic performance of the PCA3 urine test. Nat Rev Urol 2011;8: 123–4. 94.Zhu M, Chen Q, Liu X, et al. lncRNA H19/miR-675 axis re- pressed prostate cancer metastasis by targeting TGFBI. FEBS J 2014;281; 3766–75. 95.Sun H, Wang G, Peng Y, et al. H19 lncRNA mediates 17b- estradiol-induced cell proliferation in MCF-7 breast cancer cells. Oncol Rep 2015;33(6):3045–52. 96.Vennin C, Spruyt N, Dahmani F, et al. H19 non coding RNA- derived miR-675 enhances tumorigenesis and metastasis of breast cancer cells by downregulating c-Cbl and Cbl-b. Oncotarget 2015;6:29209–23. 97.Leyten GH, Hessels D, Jannink SA, et al. Prospective multi- centre evaluation of PCA3 and TMPRSS2-ERG gene fusions as diagnostic and prognostic urinary biomarkers for prostate cancer. Eur Urol 2014;65:534–42. Emerging role of noncoding RNA in prostate cancer progression | 265 Downloadedfromhttps://academic.oup.com/bfg/article-abstract/15/3/257/1742432bygueston10March2020