31
1 The lncRNA DRAIC/PCAT29 locus constitutes a tumor suppressive nexus Kouhei Sakurai 1 , Brian J. Reon 1 , Jordan Anaya 1 , Anindya Dutta 1 1 Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA Running title: A novel lncRNA, DRAIC represses migration Keywords prostate cancer, castration resistance, lncRNA, androgen receptor Financial support This work was supported by P01CA104106 from NIH to AD Corresponding author: Anindya Dutta 1340 Jefferson Park Ave, Jordan Hall, Room 1232, Charlottesville, VA, 22908-0733, USA [email protected], (434) 924-2466 Disclosure of Potential Conflicts of Interest No potential conflicts of interest were disclosed. Word count (excluding references) : 5042 Total number of figures : 7 on October 17, 2020. © 2015 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 20, 2015; DOI: 10.1158/1541-7786.MCR-15-0016-T

Molecular Cancer Research - The lncRNA …...2015/02/20  · 4 repressed by AR (14,15). We also showed that multiple oncogenes, mTOR, SMARCD1, SNF2H and IGF1R targeted by miR-99a contribute

  • Upload
    others

  • View
    3

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Molecular Cancer Research - The lncRNA …...2015/02/20  · 4 repressed by AR (14,15). We also showed that multiple oncogenes, mTOR, SMARCD1, SNF2H and IGF1R targeted by miR-99a contribute

1

The lncRNA DRAIC/PCAT29 locus constitutes a tumor suppressive nexus

Kouhei Sakurai1, Brian J. Reon1, Jordan Anaya1, Anindya Dutta1

1Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine,

Charlottesville, VA, 22908, USA

Running title: A novel lncRNA, DRAIC represses migration

Keywords

prostate cancer, castration resistance, lncRNA, androgen receptor

Financial support

This work was supported by P01CA104106 from NIH to AD

Corresponding author: Anindya Dutta

1340 Jefferson Park Ave, Jordan Hall, Room 1232, Charlottesville, VA, 22908-0733, USA

[email protected], (434) 924-2466

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

Word count (excluding references) : 5042

Total number of figures : 7

on October 17, 2020. © 2015 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 20, 2015; DOI: 10.1158/1541-7786.MCR-15-0016-T

Page 2: Molecular Cancer Research - The lncRNA …...2015/02/20  · 4 repressed by AR (14,15). We also showed that multiple oncogenes, mTOR, SMARCD1, SNF2H and IGF1R targeted by miR-99a contribute

2

ABSTRACT

Long non-coding RNAs (lncRNAs) are emerging as major regulators of cellular

phenotypes and implicated as oncogenes or tumor suppressors. Here we report a novel tumor

suppressive locus on human chromosome 15q23 that contains two multi-exonic lncRNA genes of

100 kb each: DRAIC (LOC145837) and the recently reported PCAT29. The DRAIC lncRNA was

identified from RNA-seq data and is downregulated as prostate cancer cells progress from an

androgen dependent (AD) to castration resistant (CR) state. Prostate cancers persisting in patients

after androgen deprivation therapy (ADT) select for decreased DRAIC expression, and higher levels

of DRAIC in prostate cancer is associated with longer disease-free survival (DFS). Androgen

induced androgen receptor (AR) binding to the DRAIC locus and repressed DRAIC expression. In

contrast, FOXA1 and NKX3-1 are recruited to the DRAIC locus to induce DRAIC, and FOXA1

specifically counters the repression of DRAIC by AR. The decrease of FOXA1 and NKX3-1, and

aberrant activation of AR, thus accounts for the decrease of DRAIC during prostate cancer

progression to the CR state. Consistent with DRAIC being a good prognostic marker, DRAIC

prevents the transformation of cuboidal epithelial cells to fibroblast-like morphology and prevents

cellular migration and invasion. A second tumor suppressive lncRNA PCAT29, located 20 kb

downstream of DRAIC, is regulated identically by AR and FOXA1 and also suppresses cellular

migration and metastasis. Finally, based on TCGA analysis, DRAIC expression predicts good

prognosis in a wide range of malignancies: bladder cancer, low grade gliomas, lung adenocarcinoma,

stomach adenocarcinoma, renal clear cell carcinoma, hepatocellular carcinoma, skin melanoma and

stomach adenocarcinoma. Implications: This study reveals a novel tumor suppressive locus

encoding two hormone-regulated lncRNAs, DRAIC and PCAT29, that are prognostic for a wide

variety of cancer types.

on October 17, 2020. © 2015 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 20, 2015; DOI: 10.1158/1541-7786.MCR-15-0016-T

Page 3: Molecular Cancer Research - The lncRNA …...2015/02/20  · 4 repressed by AR (14,15). We also showed that multiple oncogenes, mTOR, SMARCD1, SNF2H and IGF1R targeted by miR-99a contribute

3

INTRODUCTION

The growth of prostate cancer cells initially depends on androgen. Therefore, Androgen

Deprivation Therapy (ADT) is useful for primary prostate cancer. However, prostate cancer cells

progress after ADT to grow in low androgen, a condition called castration resistant (CR) (formerly

androgen-independent) state, leading to a tumor recurrence and metastasis (1). Several lines of

evidence have shown that the Androgen Receptor (AR) or androgen-responsive pathways are

differently activated in the CR cells so that pathways are active in low or absent androgen (1,2) . In

addition, alternative pathways such as mTOR and IGF1R signaling are activated to mimic the action

of androgens and promote prostate cancer cell growth (3). However, the detailed mechanisms by

which androgen dependent (AD) cells become CR remain unclear.

Recent transcriptome analyses have identified a variety of non-coding RNAs as important

gene regulators (4–9). Long non-coding RNAs (lncRNAs) are defined as RNAs >200 nt in length

with no functional open reading frame (10). Our lab has identified two novel lncRNAs, APTR

(Alu-mediated p21 transcriptional regulator), which recruits PRC2 (Polycomb repressive complex 2)

to p21 promoter region to repress the transcription of p21 (4) and MUNC (MyoD Upstream

Non-Coding), which can promote myogenesis (6). Some lncRNAs are known to be aberrantly

expressed and act as oncogenes or tumor suppressors in cancers including prostate cancer. Nuclear

lncRNAs, PCGEM1 and PRNCR1 bind to AR to stimulate AR-mediated gene programs (11).

Cytoplasmic lncRNA, PCAT-1 suppresses BRCA2 through its 3’UTR (untranslated region) to control

homologous recombination (12). But how these prostate cancer related lncRNAs are regulated or

whether they contribute to prostate cancer progression is largely unknown (11–13).

In our previous work, we performed microRNA (miRNA) screening using AD and CR

cells and identified a tumor suppressive miRNA, miR-99a, that is downregulated in CR cells and

on October 17, 2020. © 2015 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 20, 2015; DOI: 10.1158/1541-7786.MCR-15-0016-T

Page 4: Molecular Cancer Research - The lncRNA …...2015/02/20  · 4 repressed by AR (14,15). We also showed that multiple oncogenes, mTOR, SMARCD1, SNF2H and IGF1R targeted by miR-99a contribute

4

repressed by AR (14,15). We also showed that multiple oncogenes, mTOR, SMARCD1, SNF2H and

IGF1R targeted by miR-99a contribute to prostate cancer progression (14–16). In this study, we

report a novel lncRNA designated as DRAIC (Downregulated RNA in Androgen Independent Cells)

that is similarly regulated. AR is recruited to DRAIC locus to repress DRAIC. Conversely, DRAIC is

induced by FOXA1 and NKX3-1, which are recruited to the same region as AR at the DRAIC locus

and FOXA1 counters the repression of DRAIC by AR. Interestingly, a tumor suppressive lncRNA,

PCAT29, which was recently reported by Malik et.al. (13), is located 20kb downstream of DRAIC

locus and we report that it is also regulated by AR, FOXA1 and NKX3-1 just like DRAIC.

Functional analyses show that DRAIC inhibits cancer cell migration and invasion. This study

indicates that progression of prostate cancer is accompanied by a decrease of FOXA1 and NKX3-1,

which leads to the decrease both the novel tumor suppressive lncRNAs, DRAIC and PCAT29,

thereby increasing prostate cancer migration and invasion and decreasing disease free survival. This

is the first report of a novel lncRNA cluster, DRAIC/PCAT29 regulated by the same mechanism and

suppressing prostate cancer progression. Analysis of publicly available data from TCGA (The Cancer

Genome Atlas) revealed that DRAIC is a predictor of good prognosis in at least seven other

malignancies.

on October 17, 2020. © 2015 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 20, 2015; DOI: 10.1158/1541-7786.MCR-15-0016-T

Page 5: Molecular Cancer Research - The lncRNA …...2015/02/20  · 4 repressed by AR (14,15). We also showed that multiple oncogenes, mTOR, SMARCD1, SNF2H and IGF1R targeted by miR-99a contribute

5

MATERIALS AND METHODS

Cell culture

VCap cells were maintained in DMEM. PC3M-luc cells were maintained in MEM-L

glutamine containing MEM Non-Essential Amino Acids, MEM Vitamin Solution, Sodium Pyruvate

(All are Life technology.). Other cells were maintained in RPMI1640 medium. All medium contain

10% fetal calf serum, except when measuring the effect of androgen. For the experiments on

androgen responsiveness, LNCaP cells were cultured in phenol red–free RPMI 1640 medium

supplemented with charcoal:dextran-stripped FBS (Hyclone) for 48 hours before the addition of

R1881 (Perkin-Elmer).

Transfection

Transfections of siRNA (50nM) and plasmid vector were performed with Lipofectamine RNAiMax

and Lipofectamine 2000 (Invitrogen), respectively. siRNA sequences are shown in Supplementary

Table1.

Scratch wound healing assay

Scratches were performed by pipet tip in 6 well plate. After incubation for 24h or 48h, the migration

of cells into the scratch was imaged. Gap areas were calculated by Image J.

Matrigel invasion assay

Cells were seeded into 24 well Matrigel Invasion Chamber (BD Biosciences) at 1x105 cells in serum

free medium. 10% FCS as chemoattractant was added only to the lower compartment. After

on October 17, 2020. © 2015 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 20, 2015; DOI: 10.1158/1541-7786.MCR-15-0016-T

Page 6: Molecular Cancer Research - The lncRNA …...2015/02/20  · 4 repressed by AR (14,15). We also showed that multiple oncogenes, mTOR, SMARCD1, SNF2H and IGF1R targeted by miR-99a contribute

6

incubation for 48h, the non-invaded cells were removed from the upper surface of the membrane by

a cotton swab. The invaded cells were fixed using methanol, stained by Crystal Violet and counted

per membrane.

RNA isolation, RT-PCR, Western Blotting and ChIP assay

Total RNA and nuclear/cytoplasmic RNAs were extracted using Trizol total RNA isolation reagent

(Invitrogen), PARIS kit (Ambion), respectively. RT-PCR and Western Blotting were performed

according to standard protocols. ChIP assay was performed with cells crosslinked with 1%

formaldehyde and using 5 ug of antibody on Dynabeads according to published protocol (4). All

details of the protocols are in Supplementary Information.

ChIP-seq analysis and RNA-seq analysis

Publicly available ChIP-seq and RNA-seq data were downloaded and analyzed by standard

bioinformatics protocols. Details are described in Supplementary Information.

Kaplan-Meier plot analysis

Publicly available TCGA data at cBioPortal (17) was used to plot Kaplan-Meier plots on tumors

divided into two groups based on level of DRAIC expressed as a Z-score (18–20) Only those plots

are included that showed a statistically significant (p<0.05) survival difference between the two

groups of patients. Similar trends were seen in other plots of these malignancies but are not included

because the p-value did not reach significance.

on October 17, 2020. © 2015 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 20, 2015; DOI: 10.1158/1541-7786.MCR-15-0016-T

Page 7: Molecular Cancer Research - The lncRNA …...2015/02/20  · 4 repressed by AR (14,15). We also showed that multiple oncogenes, mTOR, SMARCD1, SNF2H and IGF1R targeted by miR-99a contribute

7

RESULTS

DRAIC is a novel lncRNA decreased in CR cells and repressed by R1881

In order to identify novel lncRNAs involved in prostate cancer progression, we compared

two published RNA-sequencing (RNA-seq) datasets (21,22), [A] LNCap vs C4-2B cells and [B]

vehicle vs R1881 (androgen analog)-treated LNCap cells (Fig.1A). C4-2B cells are bone metastatic

CR cells derived from parental AD, LNCap cells (23). We tried to identify the lncRNAs that are (a)

increased in C4-2B compared to LNCaP cells and induced by R1881 in LNCap cells or (b) decreased

in C4-2B compared to LNCaP cells and repressed by R1881 in LNCap cells.

903 and 751 genes were differentially expressed (p<0.05) in [A] and [B] comparisons,

respectively (Fig.1A). Intersection of these genes identified 72 genes that meet (a) or (b) criteria as

mentioned above. Among them, there were two lncRNAs, LOC728431 (also known as LINC01137)

and LOC145837. Both were lower in C4-2B than LNCap cells and repressed by R1881 in LNCap

cells (Fig.1A). LOC728431 and LOC145837 are composed of 3 exons at Chr.1p34.3 and 5 exons at

Chr.15q23, respectively (Fig.1B).

Q-RT-PCR showed that LOC728431 is almost at the same level in LNCap and C4-2B cells

and is not drastically decreased by R1881, contrary to the RNA-seq comparisons (Fig.1C, D).

Therefore we excluded LOC728431 from further analysis.

In contrast, Q-RT-PCR confirmed that C4-2B cells express lower level of LOC145837

(renamed by us as DRAIC) than LNCap cells and the expression is also decreased in other CR cells

(Fig. 1E). In addition, DRAIC was repressed by R1881 in dose and time-dependent manners (Fig.

1F).

DRAIC is a cytoplasmic and poly-adenylated RNA.

on October 17, 2020. © 2015 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 20, 2015; DOI: 10.1158/1541-7786.MCR-15-0016-T

Page 8: Molecular Cancer Research - The lncRNA …...2015/02/20  · 4 repressed by AR (14,15). We also showed that multiple oncogenes, mTOR, SMARCD1, SNF2H and IGF1R targeted by miR-99a contribute

8

DRAIC is a spliced transcript of 1.7 kb that is expressed mainly in the cytoplasm

(Supplementary Fig.1A). The coding potential (calculated by CPAT, http://rna-cpat.sourceforge.net/)

of DRAIC is 0.342, which is comparable with those of other cytoplasmic lncRNAs: PCAT1 (12)

(0.693) (2.1 kb RNA) and TINCR (24) (0.204) (3.8 kb RNA). For comparison, the coding potential

of protein coding genes like GAPDH and Orc1 are 0.99. We confirmed the 3’end of DRAIC by

3’RACE using LNCap polyA+ RNA (Supplementary Fig. 1B). There are at least 3 additional

transcript variants of DRAIC (Supplementary Fig. 2A) although RNA-seq data in LNCap cells

(vehicle) (21) shows that the read counts of these 3 additional variants are much less than the ones of

DRAIC (data not shown). Q-RT-PCR with variant-specific primers revealed that their expression

patterns are similar to DRAIC (Supplementary Fig. 2B). There is no evidence in the 3'RACE-PCR

products, the EST database or the RNAseq data of DRAIC being spliced to the PCAT29 gene (13)

that is located 20 kb downstream.

DRAIC is a clinically relevant lncRNA in a variety of cancers.

In order to test whether Androgen Deprivation Therapy (ADT) selects for changes in

expression of DRAIC as the cancer progresses to CR state, we analyzed published RNA-seq of seven

prostate cancer rich tumor biopsies before and after 22 weeks of ADT (25). Prostate cancer that

persisted after ADT shows a 10X decrease of DRAIC (Fig. 2A), suggesting that androgen deprivation

in patients selects for cancer cells with low expression of DRAIC. Note that the original publication

(25) shows that only about 1600 genes are increased or decreased >2x by ADT with the vast majority

of genes remaining unchanged, suggesting that the decrease of DRAIC was not due to a change in the

lineage of cells surviving ADT.

If decreased DRAIC is a marker for progression of prostate cancer to CR state, one would

on October 17, 2020. © 2015 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 20, 2015; DOI: 10.1158/1541-7786.MCR-15-0016-T

Page 9: Molecular Cancer Research - The lncRNA …...2015/02/20  · 4 repressed by AR (14,15). We also showed that multiple oncogenes, mTOR, SMARCD1, SNF2H and IGF1R targeted by miR-99a contribute

9

predict that high levels of DRAIC may predict a good prognosis. Kaplan-Meier plot based on

RNAseq and disease progression data from “Prostate Adenocarcinoma (MSKCC, 2010)” available at

cBioPortal (18) revealed that lower expression of DRAIC predicts a lower probability of

disease-free-survival of patients (Fig. 2B). Thus, DRAIC is a clinically relevant lncRNA that favors a

good response to therapy of prostate cancer.

We wondered whether the good prognostic function of DRAIC could be extended to an

unrelated malignancy. Kaplan-Meier plots were calculated using RNAseq and overall survival or

disease-free survival data for the tumors indicated in Fig. 2C, D. Seven malignancies showed

statistically significant survival benefit of DRAIC overeexpression in either overall survival (bladder

cancer, lower grade glioma, lung adnocarcinoma) or disease free survival (renal clear cell carcinoma,

hepatocellular carcinoma, skin melanoma) or both (stomach adenocarcinoma).

AR is recruited to DRAIC promoter and required for the repression of DRAIC

We next sought to identify how DRAIC is repressed by Androgen. The downregulation of

DRAIC by the androgen analog, R1881, was reversed by androgen antagonist, Bicalutamide and by

AR knockdown (Fig. 3A, B). We analyzed published AR Chromatin

Immunoprecipitation-sequencing (ChIP-seq) data (26) and identified several sites upstream and

within DRAIC that are bound by AR in the presence of R1881 (Fig. 3C). AR ChIP-PCR confirmed

that AR is recruited to Regions 1, 2 and 4 by R1881 (second grey bar in each set) and that the

recruitment is diminished by Bicalutamide (third grey bar in each set) (Fig. 3D). Therefore,

androgen-driven AR recruitment to the DRAIC locus is associated with the repression.

FOXA1 and NKX3-1 occupy the same regions where AR is recruited at DRAIC promoter

on October 17, 2020. © 2015 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 20, 2015; DOI: 10.1158/1541-7786.MCR-15-0016-T

Page 10: Molecular Cancer Research - The lncRNA …...2015/02/20  · 4 repressed by AR (14,15). We also showed that multiple oncogenes, mTOR, SMARCD1, SNF2H and IGF1R targeted by miR-99a contribute

10

AR often co-localizes with other transcriptional factors across the prostate genome (27).

Tan PY et.al. showed that the binding motifs of FOXA1 and NKX3-1 are highly enriched in AR

ChIP-seq samples (27). In addition, FOXA1 has been reported to act as a pioneer factor that opens

local chromatin structure to allow AR to be recruited (28–31). We therefore analyzed published

FOXA1 and NKX3-1 ChIP-seq datasets (26,27) to examine the binding of these transcription factors

to the DRAIC locus (Fig. 4A, B). Interestingly, ChIP-seq peaks of these two transcriptional factors

overlapped with those of AR at the DRAIC locus (Fig. 4A, B, Fig. 3C). We confirmed by ChIP-PCR

that Regions 1, 2 and 4 bind to FOXA1 and to NKX3-1 (Fig. 4A, B). Regions 1, 2 and 4 contain

several ARE (androgen-responsive element) half-sites and several FOXA1 and NKX3-1 binding

sites close to the AREs (Supplementary Fig. 3).

The expression of DRAIC is positively regulated by FOXA1 and NKX3-1

Contrary to our expectation that FOXA1 is a pioneer factor for AR and should repress

DRAIC, the expression pattern of FOXA1 and NKX3-1 was similar to that of DRAIC: lower in most

CR cells (except C4-2) compared to the AD cells (Fig. 4C and Fig. 1E). RNA-seq data from prostate

adenocarcinomas from the cBioPortal (n=487) show weak but statistically significant positive

correlation between the expression of FOXA1 and DRAIC or NKX3-1 and DRAIC (Fig. 4D, E).

In addition, knockdown of FOXA1 or NKX3-1 decreased DRAIC levels (Fig. 4F). The

siRNA-resistant forms of FOXA1 or NKX3-1 partially rescued the downregulation of DRAIC

induced by the cognate siRNAs, ruling out the possibility of off-target effects of the siRNAs (Fig.

4G). FOXA1 or NKX3-1 therefore have an opposite effect on DRAIC expression compared to AR,

suggesting that FOXA1 is not acting as a pioneer factor for AR at the DRAIC promoter.

Knockdown of FOXA1 also decreased NKX3-1 protein and mRNA (Fig. 4F). Indeed, the

on October 17, 2020. © 2015 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 20, 2015; DOI: 10.1158/1541-7786.MCR-15-0016-T

Page 11: Molecular Cancer Research - The lncRNA …...2015/02/20  · 4 repressed by AR (14,15). We also showed that multiple oncogenes, mTOR, SMARCD1, SNF2H and IGF1R targeted by miR-99a contribute

11

FOXA1 and NKX3-1 mRNA levels were positively correlated in clinical samples (Fig. 4H). We did

not see any significant FOXA1 ChIP-seq peaks (26) at the NKX3-1 locus (data not shown),

suggesting that FOXA1 stimulates NKX3-1 expression by an unknown indirect mechanism.

A lncRNA, PCAT29 is regulated by AR, FOXA1 and NKX3-1.

Malik et.al. recently reported a tumor suppressive lncRNA, PCAT29, whose expression is

repressed by AR (13). Interestingly, PCAT29 gene is located 20 kb downstream of DRAIC. We

therefore analyzed published ChIP-seq dataset for AR, FOXA1 and NKX3-1 and identified that

these transcriptional factors are also recruited to PCAT29 locus (Fig. 5A, B, C). The expression

pattern of PCAT29 in a panel of prostate cancer cells is similar to that of DRAIC except for C4-2B

cells (Fig. 5D and Fig. 1E). Since PCAT29 is not annotated in the level 3 data from TCGA, we

used RNA from de-identified prostate cancer samples collected at UVA and used in a previous paper

to analyze the correlation between DRAIC and PCAT29 (14). Q-RT-PCR of these lncRNAs showed

a positive correlation between the expression of the two lncRNAs (Fig. 5E). From these results, we

hypothesized that PCAT29 is regulated by FOXA1 and NKX3-1 in a manner similar to DRAIC.

Indeed, siRNA against FOXA1 or NKX3-1 decreased PCAT29 expression (Fig. 5F, Fig. 4F).

Jin et.al. recently reported that FOXA1 knockdown can shift or increase AR binding to

selected sites (30). We analyzed their AR ChIP-seq data and found that shFOXA1 increases AR

recruitment at DRAIC/PCAT29 cluster (Fig. 5G). Thus, FOXA1 actually decreases the recruitment of

AR to the DRAIC/PCAT29 locus. Consistent with this, R1881 treatment or FOXA1 knockdown

independently repress DRAIC and PCAT29, but together they repress both genes even further (Fig.

5H). This result suggests that instead of being a pioneer factor of AR, FOXA1 counters the action of

AR at the DRAIC/PCAT29 cluster.

on October 17, 2020. © 2015 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 20, 2015; DOI: 10.1158/1541-7786.MCR-15-0016-T

Page 12: Molecular Cancer Research - The lncRNA …...2015/02/20  · 4 repressed by AR (14,15). We also showed that multiple oncogenes, mTOR, SMARCD1, SNF2H and IGF1R targeted by miR-99a contribute

12

DRAIC represses cellular migration and invasion.

Like PCAT29 (13), DRAIC is a marker for good prognosis in prostate cancer (Fig. 2B), and

so is expected to repress oncogenic phenotypes. PCAT29 has been reported to repress invasion and

metastasis (13). The ability of a panel of prostate cancer cells to invade through Matrigel in a

Boyden Chamber assay was anti-correlated with the level of expression of DRAIC in the same cells

(Fig. 6A; Fig. 1E), suggesting that DRAIC, like PCAT29, represses invasion. Transient knockdown

of DRAIC by siRNA in LNCap cells unexpectedly decreased cell numbers by about 30-50% (Fig.

6B), suggesting that DRAIC has a pro-proliferative function. When DRAIC was stably knocked down

by shRNA in LNCap cells, the cell proliferation was similarly decreased (data not shown) but

interestingly, the cell morphology was changed from cuboidal to fibroblast-like shape (Fig. 6C).

Stable DRAIC overexpression in PC3M-luc cells, in contrast, showed the opposite phenotype, with a

change in morphology from fibroblast shape to cuboidal shape (Fig. 6D). In a scratch assay to

measure cell migration and in a Matrigel invasion assay, the migration and invasion of LNCaP cells

is increased by DRAIC knockdown (Fig. 6E, F). In similar assays, the migration and invasion of

PC3M-luc cells is decreased by DRAIC overexpression (Fig. 6G, H). Taken together, these results

suggest that DRAIC promotes cell proliferation but inhibits cell migration and invasion. We

summarized the similarities and differences between DRAIC and PCAT29 in Figure 7A.

DISCUSSION

The regulation of DRAIC and PCAT29 genes is remarkably similar to that we reported for

the miR-99 family (14,15). AR is recruited to the pri-miR-99a promoter and represses transcription

in concert with EZH2 (14,15). Considering that AR is recruited to broad regions around DRAIC (and

on October 17, 2020. © 2015 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 20, 2015; DOI: 10.1158/1541-7786.MCR-15-0016-T

Page 13: Molecular Cancer Research - The lncRNA …...2015/02/20  · 4 repressed by AR (14,15). We also showed that multiple oncogenes, mTOR, SMARCD1, SNF2H and IGF1R targeted by miR-99a contribute

13

the transcript variants) and PCAT29 gene, it is conceivable that a chromatin looping mechanism

following AR recruitment is involved to produce a large domain with gene suppression.

FOXA1 and NKX3-1 have been variably thought to be tumor suppressive (32,33) and

oncogenic (34,35). In the regulation described here, the two factors appear to be tumor suppressive

in that their levels are decreased in CR cells and they are positive transcription factors for DRAIC

and PCAT29, both of which decrease migration and invasion and predict good prognosis. We

propose a model that FOXA1 and NKX3-1 induce the expression of DRAIC/PCAT29 in AD prostate

epithelial cells but are downregulated in CR cells, leading to the decrease of DRAIC (Fig. 7B).

Moreover, DRAIC is further repressed in the CR cells by differently activated androgen-responsive

pathways (Fig. 7B).

FOXA1 is well known to be a pioneer factor and stimulates AR-mediated gene regulation

(28–31). But our study clearly shows that FOXA1 counters the repression of DRAIC/PCAT29 by AR

(Fig. 5H). Jin et. al. showed that excess of FOXA1 opens up an excess of chromatin regions and

ends up diluting AR across the genome thereby indirectly inhibiting specific AR binding events (30).

Therefore we need to study whether FOXA1 directly or indirectly represses AR recruitment to

DRAIC/PCAT29 cluster. Similarly, NKX3-1 is known to be positively regulated by AR and

facilitates regulation of the AR downstream genes by associating with AR (27). However, at the

DRAIC/PCAT29 locus NKX3-1 has the opposite effect of AR on gene expression, suggesting a

different mode of action.

Our functional analysis showed that DRAIC represses migration and invasion (Fig. 6) just

like PCAT29 (13). However, knockdown of DRAIC represses cell proliferation (Fig. 6B) while

knockdown of PCAT29 induces proliferation (13), suggesting that all functions of these lncRNA are

not identical. This is borne out by the different cellular localization of DRAIC (cytoplasmic) and

on October 17, 2020. © 2015 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 20, 2015; DOI: 10.1158/1541-7786.MCR-15-0016-T

Page 14: Molecular Cancer Research - The lncRNA …...2015/02/20  · 4 repressed by AR (14,15). We also showed that multiple oncogenes, mTOR, SMARCD1, SNF2H and IGF1R targeted by miR-99a contribute

14

PCAT29 (nuclear) (13). Future studies will analyze whether DRAIC and PCAT29 synergize with

each other in repressing cell migration and invasion in vitro and in vivo.

Although it is tempting to propose that DRAIC represses epithelial-mesenchymal transition

(EMT), preliminary results suggest that levels of mRNA involved in EMT are unchanged by DRAIC

knockdown or overexpression (data not shown). Diverse mechanisms have been proposed by which

lncRNAs could regulate many phenotypes at transcriptional and post-transcriptional levels (36).

Thus a detailed analysis is needed to determine the downstream targets of this cytoplasmic lncRNA

and the molecular mechanism by which DRAIC regulates cellular migration and invasion.

It will be interesting to investigate in the future whether DRAIC/PCAT29 expression levels

are related to the Gleason grade and whether they are useful as an independent prognostic

biomarkers of prostate cancer. The results reported here highlight that a thorough study of lncRNAs

altered during prostate cancer genesis and progression will be very important for improving our

understanding and the therapy of this cancer.

Finally, DRAIC expression predicts good prognosis in a wide range of malignancies from

many other tissues, suggesting that it is an important and ubiquitous tumor suppressor. Whether the

mechanism by which clinical progression is suppressed is the same in all these tumors, and whether

PCAT29 has a similar anti-progression effect in these tumors as in prostate cancer, will be important

questions for the future.

on October 17, 2020. © 2015 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 20, 2015; DOI: 10.1158/1541-7786.MCR-15-0016-T

Page 15: Molecular Cancer Research - The lncRNA …...2015/02/20  · 4 repressed by AR (14,15). We also showed that multiple oncogenes, mTOR, SMARCD1, SNF2H and IGF1R targeted by miR-99a contribute

15

ACKNOWLEDGEMENT

We thank members of the Prostate Cancer Research Working group at UVA and the Dutta laboratory

for advice and helpful discussions.

REFERENCES

1. Karantanos T, Corn PG, Thompson TC. Prostate cancer progression after androgen deprivation therapy: mechanisms of castrate resistance and novel therapeutic approaches. Oncogene 2013;32:5501–11.

2. Chen CD, Welsbie DS, Tran C, Baek SH, Chen R, Vessella R, et al. Molecular determinants of resistance to antiandrogen therapy. Nat Med 2004;10:33–9.

3. Majumder PK, Sellers WR. Akt-regulated pathways in prostate cancer. Oncogene 2005;24:7465–74.

4. Negishi M, Wongpalee SP, Sarkar S, Park J, Lee KY, Shibata Y, et al. A new lncRNA, APTR, associates with and represses the CDKN1A/p21 promoter by recruiting polycomb proteins. PLoS One 2014;9:e95216.

5. Dey BK, Pfeifer K, Dutta A. The H19 long noncoding RNA gives rise to microRNAs miR-675-3p and miR-675-5p to promote skeletal muscle differentiation and regeneration. Genes Dev 2014;28:491–501.

6. Mueller AC, Cichewicz M a, Dey BK, Layer R, Reon BJ, Gagan JR, et al. MUNC: A lncRNA that induces the expression of pro-myogenic genes in skeletal myogenesis. Mol Cell Biol 2014 (in press)

7. Kumar P, Anaya J, Mudunuri SB, Dutta A. Meta-analysis of tRNA derived RNA fragments reveals that they are evolutionarily conserved and associate with AGO proteins to recognize specific RNA targets. BMC Biol 2014;12:78.

on October 17, 2020. © 2015 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 20, 2015; DOI: 10.1158/1541-7786.MCR-15-0016-T

Page 16: Molecular Cancer Research - The lncRNA …...2015/02/20  · 4 repressed by AR (14,15). We also showed that multiple oncogenes, mTOR, SMARCD1, SNF2H and IGF1R targeted by miR-99a contribute

16

8. Cesana M, Cacchiarelli D, Legnini I, Santini T, Sthandier O, Chinappi M, et al. A long noncoding RNA controls muscle differentiation by functioning as a competing endogenous RNA. Cell 2011;147:358–69.

9. Yuan J, Yang F, Wang F, Ma J, Guo Y, Tao Q, et al. A long noncoding RNA activated by TGF-β promotes the invasion-metastasis cascade in hepatocellular carcinoma. Cancer Cell 2014;25:666–81.

10. Wahlestedt C. Targeting long non-coding RNA to therapeutically upregulate gene expression. Nat Rev Drug Discov 2013;12:433–46.

11. Yang L, Lin C, Jin C, Yang JC, Tanasa B, Li W, et al. lncRNA-dependent mechanisms of androgen-receptor-regulated gene activation programs. Nature 2013;500:598–602.

12. Prensner JR, Chen W, Iyer MK, Cao Q, Ma T, Han S, et al. PCAT-1, a long noncoding RNA, regulates BRCA2 and controls homologous recombination in cancer. Cancer Res 2014;74:1651–60.

13. Malik R, Patel L, Prensner JR, Shi Y, Iyer MK, Subramaniyan S, et al. The lncRNA PCAT29 Inhibits Oncogenic Phenotypes in Prostate Cancer. Mol Cancer Res 2014;12:1081–7.

14. Sun D, Lee YS, Malhotra A, Kim HK, Matecic M, Evans C, et al. miR-99 family of MicroRNAs suppresses the expression of prostate-specific antigen and prostate cancer cell proliferation. Cancer Res 2011;71:1313–24.

15. Sun D, Layer R, Mueller a C, Cichewicz M a, Negishi M, Paschal BM, et al. Regulation of several androgen-induced genes through the repression of the miR-99a/let-7c/miR-125b-2 miRNA cluster in prostate cancer cells. Oncogene 2014;33:1448–57.

16. Mueller a C, Sun D, Dutta a. The miR-99 family regulates the DNA damage response through its target SNF2H. Oncogene 2013;32:1164–72.

17. Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov 2012;2:401–4.

18. Taylor BS, Schultz N, Hieronymus H, Gopalan A, Xiao Y, Carver BS, et al. Integrative genomic profiling of human prostate cancer. Cancer Cell 2011;18:11–22.

on October 17, 2020. © 2015 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 20, 2015; DOI: 10.1158/1541-7786.MCR-15-0016-T

Page 17: Molecular Cancer Research - The lncRNA …...2015/02/20  · 4 repressed by AR (14,15). We also showed that multiple oncogenes, mTOR, SMARCD1, SNF2H and IGF1R targeted by miR-99a contribute

17

19. Weinstein JN, Akbani R, Broom BM, Wang W, Verhaak RG, McConkey D, et al. Comprehensive molecular characterization of urothelial bladder carcinoma. Nature 2014;507:315–22.

20. Collisson E a., Campbell JD, Brooks AN, Berger AH, Lee W, Chmielecki J, et al. Comprehensive molecular profiling of lung adenocarcinoma. Nature 2014;511:543-50.

21. Decker KF, Zheng D, He Y, Bowman T, Edwards JR, Jia L. Persistent androgen receptor-mediated transcription in castration-resistant prostate cancer under androgen-deprived conditions. Nucleic Acids Res 2012;40:10765–79.

22. Tewari AK, Yardimci GG, Shibata Y, Sheffield NC, Song L, Taylor BS, et al. Chromatin accessibility reveals insights into androgen receptor activation and transcriptional specificity. Genome Biol 2012;13:R88.

23. Thalmann GN, Anezinis PE, Chang S, Thaimann N, Hopwood VL, Pathak S, et al. Androgen-independent Cancer Progression and Bone Metastasis in the LNCaP Model of Human Prostate Cancer Model of Human Prostate Cancer. Cancer Res;1994;54:2577–81.

24. Kretz M, Siprashvili Z, Chu C, Webster DE, Zehnder A, Qu K, et al. Control of somatic tissue differentiation by the long non-coding RNA TINCR. Nature 2013;493:231–5.

25. Rajan P, Sudbery IM, Villasevil MEM, Mui E, Fleming J, Davis M, et al. Next-generation sequencing of advanced prostate cancer treated with androgen-deprivation therapy. Eur Urol 2014;66:32–9.

26. Yu J, Yu J, Mani R-S, Cao Q, Brenner CJ, Cao X, et al. An integrated network of androgen receptor, polycomb, and TMPRSS2-ERG gene fusions in prostate cancer progression. Cancer Cell 2010;17:443–54.

27. Tan PY, Chang CW, Chng KR, Wansa KDSA, Sung W-K, Cheung E. Integration of regulatory networks by NKX3-1 promotes androgen-dependent prostate cancer survival. Mol Cell Biol 2012;32:399–414.

28. Cai C, He HH, Gao S, Chen S, Yu Z, Gao Y, et al. Lysine-Specific Demethylase 1 Has Dual Functions as a Major Regulator of Androgen Receptor Transcriptional Activity. Cell Rep 2014;9:1618–27.

on October 17, 2020. © 2015 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 20, 2015; DOI: 10.1158/1541-7786.MCR-15-0016-T

Page 18: Molecular Cancer Research - The lncRNA …...2015/02/20  · 4 repressed by AR (14,15). We also showed that multiple oncogenes, mTOR, SMARCD1, SNF2H and IGF1R targeted by miR-99a contribute

18

29. Robinson JLL, Hickey TE, Warren a Y, Vowler SL, Carroll T, Lamb a D, et al. Elevated levels of FOXA1 facilitate androgen receptor chromatin binding resulting in a CRPC-like phenotype. Oncogene 2014;33:5666–74.

30. Jin H-J, Zhao JC, Wu L, Kim J, Yu J. Cooperativity and equilibrium with FOXA1 define the androgen receptor transcriptional program. Nat Commun 2014;5:3972.

31. Mills IG. Maintaining and reprogramming genomic androgen receptor activity in prostate cancer. Nat Rev Cancer 2014;14:187–98.

32. Jin H-J, Zhao JC, Ogden I, Bergan RC, Yu J. Androgen receptor-independent function of FoxA1 in prostate cancer metastasis. Cancer Res 2013;73:3725–36.

33. Bowen C, Bubendorf L, Voeller HJ, Progression T, Slack R, Willi N, et al. Loss of NKX3.1 Expression in Human Prostate Cancers Correlates with Tumor Progression Cancer Res 2000;60:6111–5.

34. Gerhardt J, Montani M, Wild P, Beer M, Huber F, Hermanns T, et al. FOXA1 promotes tumor progression in prostate cancer and represents a novel hallmark of castration-resistant prostate cancer. Am J Pathol 2012;180:848–61.

35. Xu LL, Srikantan V, Sesterhenn IA, Augustus M, Dean R, Moul JW, et al. Expression profile of an androgen regulated prostate specific homeobox gene NKX3.1 in primary prostate cancer. J Urol 2000;163:972–9.

36. B.K. Dey, AC. Mueller, A. Dutta. Long non-coding RNA as emerging regulators of differentiation, development, and disease. Transcription 2014 (in press)

on October 17, 2020. © 2015 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 20, 2015; DOI: 10.1158/1541-7786.MCR-15-0016-T

Page 19: Molecular Cancer Research - The lncRNA …...2015/02/20  · 4 repressed by AR (14,15). We also showed that multiple oncogenes, mTOR, SMARCD1, SNF2H and IGF1R targeted by miR-99a contribute

19

FIGURE LEGENDS

Figure 1

LncRNA, DRAIC is downregulated in CR cells and decreased by R1881

(A) Left: analysis of published RNA-seq datasets. Right: Relative expression (FPKM: Fragments Per

Kilobase of exon per Million fragments mapped) of LOC728431 and LOC145837 (DRAIC) in

LNCap cells treated with vehicle or R1881 and C4-2B cells treated with vehicle (B) Top and bottom

show LOC728431 and LOC145837 (DRAIC) gene structures and qPCR primer locations,

respectively. (C) The expression of LOC728431 in a panel of prostate cancer cells cultured in the

growth medium was measured by RT-qPCR and normalized to GAPDH. Mean±S.D. n=3. The

expression in LNCap is set as 1. (D) LNCap cells were treated with R1881 at different doses (left)

and times (right) and the expression of LOC728431 was measured by RT-qPCR. The expression in 0

pM or 0 h is set as 1. Rest as in Fig. 1C. (E) The expression of DRAIC in a panel of prostate cancer

cells cultured in growth medium was measured by RT-qPCR. Rest as in Fig. 1C. (F) LNCap cells

were treated with R1881 at different doses (left) and times (right) and the expression of DRAIC was

measured by RT-qPCR. Rest as in Fig. 1C, D.

Figure 2

DRAIC is a clinically relevant lncRNA.

(A) Relative expression (FPKM) of DRAIC in prostate cancer pre- and post-ADT (Androgen

Deprivation Therapy) (post-ADT: prostate cancer harvested approximately 22 weeks after ADT

initiation) using published RNA-seq dataset from 7 patients (25). The expression of DRAIC was

determined using the Tuxedo suite and plotted using R. The statistical significance of the changes in

DRAIC expression was evaluated using a paired t-test. p=0.0158. (B). Kaplan–Meier plot of

on October 17, 2020. © 2015 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 20, 2015; DOI: 10.1158/1541-7786.MCR-15-0016-T

Page 20: Molecular Cancer Research - The lncRNA …...2015/02/20  · 4 repressed by AR (14,15). We also showed that multiple oncogenes, mTOR, SMARCD1, SNF2H and IGF1R targeted by miR-99a contribute

20

disease-free survival (DFS) of patient with prostate adenocarcinoma from the MSKCC dataset (18)

stratified by level of DRAIC expression. In Log rank test p=0.018857. High: DRAIC level >

+0.4z and Low: DRAIC level < +0.4z. (C and D) Kaplan–Meier plot of Overall survival (OS) (C) or

Disease Free Survival (D) for indicated malignancies from TCGA (19,20) stratified by level of

DRAIC expression. EXP: the DRAIC expression level z-score cut-off used for dividing high

expressers from low expressers. n = number of patients in that group, M = median survival in

months of that group. NA: not available.

Figure 3

AR is recruited to DRAIC locus and required for DRAIC downregulation

(A) LNCaP cells were treated with no androgen, 10 nM R1881, 10 nM R1881 plus 10 uM

Bicalutamide for 24h. The expression of DRAIC in LNCaP cells was measured by RT-qPCR and

normalized to GAPDH. Rest as in Fig. 1C. (B) The expression of DRAIC in LNCaP cells after

knocking down AR by siRNA for 72 h in the absence or presence of 10 nM R1881 for 24h. **

indicates p-value of difference from siGL2 < 0.01. Rest as in Figure 1C. The expression of AR and

Actin (loading control) were detected by western blotting. (C) Published AR ChIP-seq (26) peaks in

LNCap cells at DRAIC locus in the absence (upper) or presence (lower) of R1881. Regions 1-4 in the

DRAIC locus are marked. (D) LNCap cells were treated with or without 10 nM R1881 and 10uM

Bicalutamide for 24h and AR ChIP-PCR performed. PSA promoter was used as a positive control

(15) and region 3 was used as a negative control. The value was expressed as percentage of input

DNA. ** and * indicate p-value <0.01, 0.05, respectively.

Figure 4

on October 17, 2020. © 2015 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 20, 2015; DOI: 10.1158/1541-7786.MCR-15-0016-T

Page 21: Molecular Cancer Research - The lncRNA …...2015/02/20  · 4 repressed by AR (14,15). We also showed that multiple oncogenes, mTOR, SMARCD1, SNF2H and IGF1R targeted by miR-99a contribute

21

FOXA1 and NKX3-1 positively regulate DRAIC

(A) Left: Published FOXA1 ChIP-seq (26) peaks in LNCap cells cultured in the growth medium.

Right: FOXA1 ChIP-PCR was performed with cells in 10% FCS. SLUG promoter was used as a

positive control (32). Rest as in Fig. 3D. (B) Left: Published NKX3-1 ChIP-seq (27) peaks in LNCap

cells treated with Dihydrotestosterone (DHT). Rgith: NKX3-1 ChIP-PCR was performed with cells

in 10% FCS. Rest as in Fig. 3D. (C) The mRNA and protein levels of FOXA1 and NKX3-1 were

measured by RT-qPCR and western blotting, respectively. Rest as in Fig. 1C. (D and E) The

correlation of levels of DRAIC and FOXA1 or DRAIC and NKX3-1 RNAs in prostate

adenocarcinoma (PRAD) samples (n=487) from tier-3 RNA-seq data of TCGA. Spearman

correlation coefficients and p-values are shown. (F) LNCap cells cultured in growth medium were

transfected with siRNA against FOXA1, NKX3-1 or siGL2 for 72h. The RNA levels of DRAIC,

FOXA1 and NKX3-1 and the protein levels of FOXA1, NKX3-1 and Actin were measured by

RT-qPCR and western blotting, respectively. In RT-qPCR, the expression in siGL2 is set as 1. Rest as

in Fig. 1C. (G) LNCap cells cultured in the growth medium were transfected with siRNA against

FOXA1 no.1, NKX3-1 no.1 or siGL2 and 3µg expression vector of pcDNA3-FOXA1, -NKX3-1 or

-Empty for 72 h. The expression of DRAIC was measured by RT-qPCR. The expression of DRAIC in

siGL2 plus Empty vector cells is set as 1. ** indicates p-value<0.01. Rest as in Fig. 1C. (H) The

correlation curve between FOXA1 and NKX3-1 RNAs in prostate adenocarcinoma (PRAD) samples

(n=487). Rest as in Fig.4D, E.

Figure 5

A neighboring lncRNA, PCAT29 is also repressed by AR and activated by FOXA1 and

NKX3-1.

on October 17, 2020. © 2015 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 20, 2015; DOI: 10.1158/1541-7786.MCR-15-0016-T

Page 22: Molecular Cancer Research - The lncRNA …...2015/02/20  · 4 repressed by AR (14,15). We also showed that multiple oncogenes, mTOR, SMARCD1, SNF2H and IGF1R targeted by miR-99a contribute

22

(A) Published AR ChIP-seq (26) peaks in LNCap cells at DRAIC and PCAT29 loci in the absence

(upper) or presence (lower) of R1881. (B) Published FOXA1 ChIP-seq (26) peaks in LNCap cells

cultured in the growth medium at DRAIC and PCAT29 loci. (C) Published NKX3-1 ChIP-seq (27)

peaks in LNCap cells treated with Dihydrotestosterone (DHT) at DRAIC and PCAT29 loci. (D) The

expression of PCAT29 in a panel of prostate cancer cells was measured by RT-qPCR. Rest as in Fig.

1C. (E) The delta Ct values of DRAIC and PCAT29 (normalized to GAPDH) in 12 prostate cancer

patients (University of Virginia) were subjected to Pearson correlation analysis. (F) The expression

of PCAT29 after transfection of siRNA against FOXA1, NKX3-1 or siGL2 was measured by

RT-qPCR. Rest as in Fig. 4F. (G) Published AR ChIP-seq results (30) show induction of AR binding

(arrows) at DRAIC and PCAT29 loci in LNCap cells treated with shFOXA1 or shCtrl (negative

control). R1881 is present in both cultures. (H) LNCap cells were treated with siGL2 or siFOXA1 for

72h in the absence or presence of R1881 (10 nM) for 24h. The expression of DRAIC (Left) and

PCAT29 (Right) are measured by RT-qPCR. The expression in siGL2/R1881 (-) is set as 1. Rest as

in Fig. 1C.

Figure 6

DRAIC represses cancer cell migration and invasion

(A) The relative number of cells invaded through matrigel is normalized to number in DU145 cells.

(B) Left; Proliferation of LNCap cells after transfection of siRNAs. Right; DRAIC RNA measured by

RT-qPCR and normalized to GAPDH. Rest as in Fig. 1C. (C) LNCap transduced with lentivirus

expressing shGL2, -shDRAIC no1 or no2. Left: DRAIC mRNA normalized to GAPDH. Rest as in Fig.

1C. Right: Cells stained by Crystal Violet. (D) PC3M-luc cells stably transfected with

pcDNA3-DRAIC or pcDNA3-Empty. Left: DRAIC RNA normalized to GAPDH. The level in

on October 17, 2020. © 2015 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 20, 2015; DOI: 10.1158/1541-7786.MCR-15-0016-T

Page 23: Molecular Cancer Research - The lncRNA …...2015/02/20  · 4 repressed by AR (14,15). We also showed that multiple oncogenes, mTOR, SMARCD1, SNF2H and IGF1R targeted by miR-99a contribute

23

DRAIC overexpressing cells is set as 1. Rest as in Fig. 1C. (E) Scratch wound healing assay with

LNCap cells stably expressing shGL2 or shDRAIC. Left: Representative images of scratch shown.

Scale bar: 20µm. Right: gap area quantitated by Image J. Mean±S.D. n=5. ** : difference from

shGL2 p<0.01. (F) Matrigel Invasion assay with LNCap expressing shGL2 or shDRAIC. Left:

Invaded cells fixed in methanol, stained by Crystal Violet. Right: Number of invaded cells. Rest as

in (E). (G) Scratch wound healing assay with Empty or DRAIC overexpressing PC3M-luc cells.

Image and bar graph as in (E) *: difference from Empty p<0.05. (H) Matrigel invasion assay was

performed using Empty or DRAIC overexpressing PC3M-luc cells. Rest as in (F).

Figure 7

Schematic representation of the proposed regulation of lncRNA cluster, DRAIC/PCAT29.

(A) Comparison of DRAIC and PCAT29. (B) In AD cells, even though AR activated by androgen is

recruited to DRAIC/PCAT29 cluster to repress these two lncRNA, the high level of FOXA1 counters

the repression of DRAIC/PCAT29 by AR and induces the transcription of these lncRNAs. NKX3-1,

which is indirectly up-regulated by FOXA1, contributes to the induction of DRAIC/PCAT29. During

tumor progression, the expression of FOXA1 and NKX3-1 is downregulated and AR pathways are

differentially activated despite low androgen in CR cells. The decrease of FOXA1 enhances AR

recruitment to the cluster and represses DRAIC/PCAT29. ADT selects for cells with decreased

DRAIC expression. The decrease of tumor suppressive lncRNAs, DRAIC and PCAT29, leads to

higher invasion ability and lower disease free survival in prostate cancer patients.

on October 17, 2020. © 2015 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 20, 2015; DOI: 10.1158/1541-7786.MCR-15-0016-T

Page 24: Molecular Cancer Research - The lncRNA …...2015/02/20  · 4 repressed by AR (14,15). We also showed that multiple oncogenes, mTOR, SMARCD1, SNF2H and IGF1R targeted by miR-99a contribute

751 genes

LNCapvehicle vs R1881LNCap vs C4-2B

903 genes

72 genes

LOC145837

A

LNCapLNCap R1881

C4-2B

18

16

14

12

10

8

6

4

2

0LOC728431 LOC1458372 lncRNAs

LOC728431,

[A] RNA-seq [B] RNA-seq

Re

lative

Exp

ressio

n (

FP

KM

)

(DRAIC)

0p

M

10

pM

10

0p

M

1n

M

10

nM

00.2

0.40.60.81.0

1.21.4

Re

lative

Exp

ressio

n

00.20.40.60.8

1.01.2

1.4

Re

lative

Exp

ressio

n

0h

3h 6h

12

h

24

h

R1881, 24h R1881,10nM

LNCap LNCap

C

E F

01234567

VC

ap

LN

Ca

p

C4

-2

PC

3

DU

14

5

CR cellsAD cells

C4

-2B

x 4

.19

N.D. N.D.

PC

3M

-lu

c

x 1

.0

x 0

.48

x 0

.19

N.D.

DRAIC

2.5

2.0

1.5

1.0

0.5

0

LOC728431

VC

ap

LN

Ca

p

C4

-2

PC

3

DU

14

5

CR cellsAD cells

C4

-2B

PC

3M

-lu

c

0p

M

10

pM

10

0p

M

1n

M

10

nM

R1881, 24h

0h

3h 6h

12

h

24

h

R1881,10nM

00.2

0.40.60.81.0

1.21.4

Re

lative

Exp

ressio

n0

0.2

0.40.60.81.0

1.21.4

Re

lative

Exp

ressio

n

D

DRAIC DRAIC

LNCap LNCap

LOC728431 LOC728431

B

LOC728431 (Chr.1)

5’ 3’

LOC145837 (DRAIC) (Chr.15)

5’ 3’

Re

lative

Exp

ressio

nR

ela

tive

Exp

ressio

n

Figure 1. Sakurai et.al.on October 17, 2020. © 2015 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 20, 2015; DOI: 10.1158/1541-7786.MCR-15-0016-T

Page 25: Molecular Cancer Research - The lncRNA …...2015/02/20  · 4 repressed by AR (14,15). We also showed that multiple oncogenes, mTOR, SMARCD1, SNF2H and IGF1R targeted by miR-99a contribute

Figure 2. Sakurai et.al.

A

0 20 40 60 80 100 120

DRAIC high, EXP>0.4z

DRAIC low, EXP<0.4z

p=0.018857

Months Disease Free

80

100

40

60

0

20Dis

ea

se

fre

e (

%)

n=69 (M=NA)

n=11 (M=64.66)

pre-ADT

post-ADT

100

80

60

40

20

0

Re

lative

Exp

ressio

n (

FP

KM

)DRAIC

p=0.0158

B

C

Prostate Adenocarcinoma (MSKCC, Cancer Cell 2010)

80

100

40

60

0

20

Su

rviv

ing

(%

)

80

100

40

60

0

20

Su

rviv

ing

(%

)

80

100

40

60

0

20

Su

rviv

ing

(%

)

80

100

40

60

0

20

Su

rviv

ing

(%

)

0 20 40 60 80 100120140 0 40 80 120 160 200 0 40 80 120 160 200 240 0 10 20 30 40 50

0 20 40 60 80 100 120 0 20 40 60 80 100 0 50 100150 200 250300 350 0 10 20 30 40 50

D

80

100

40

60

0

20Dis

ea

se

fre

e (

%) 80

100

40

60

0

20Dis

ea

se

fre

e (

%) 80

100

40

60

0

20Dis

ea

se

fre

e (

%) 80

100

40

60

0

20Dis

ea

se

fre

e (

%)

Bladder Urothelial carcinoma

(TCGA, Nature 2014)

Brain Lower Grade Glioma

(TCGA, Provisional)

Lung Adenocarcinoma

(TCGA, Nature 2014)

Stomach Adenocarcinoma

(TCGA, Provisional)

Kidney Renal Clear Cell

Carcinoma (TCGA, Provisional)

Liver Hepatocellular carcinoma

(TCGA, Provisional)

Skin cutaneous melanoma

(TCGA, Provisional)

Stomach Adenocarcinoma

(TCGA, Provisional)

Months Survival Months Survival Months Survival Months Survival

Months Disease Free Months Disease Free Months Disease Free Months Disease Free

EXP>0z EXP>0z EXP>0z

EXP>0z

EXP>0.05z

EXP>0.3z EXP>0.8z EXP>0.1z

n=27 (M=NA)

n=70 (M=134.17)

n=47 (M=53.29)

n=13 (M=NA)

n=75 (M=89.82)

n=23 (M=42.21)

n=83 (M=72.8)

n=12 (M=NA)

n=104 (M=19.49)

n=198 (M=63.5)

n=156 (M=37.68)

n=20 (M=18.33)

n=258 (M=88.21)

n=93 (M=18.43)

n=147 (M=48.59)

n=13 (M=11.99)

p=0.021718 p=0.004557 p=0.039773 p=0.024828

p=0.030799 p=0.049417 p=0.041919 p=0.001827

on October 17, 2020. © 2015 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 20, 2015; DOI: 10.1158/1541-7786.MCR-15-0016-T

Page 26: Molecular Cancer Research - The lncRNA …...2015/02/20  · 4 repressed by AR (14,15). We also showed that multiple oncogenes, mTOR, SMARCD1, SNF2H and IGF1R targeted by miR-99a contribute

Figure 3. Sakurai et.al.

A B

DRAICRPLP1

KIF23

67,590,000 67,630,00067,550,000

1 2 43

R1881 (-)

R1881 (+)

60

60

0

0

AR

Ch

IP-s

eq

DRAIC locus

1.0

0.8

0.6

0.4

0.2

0

% In

pu

t

AR ChIP-PCR

LNCap

R1881Bicalutamide

+++

+++

-- -

-- -

DRAIC locus

PSA

promoter

1 2 3 4

IgGAR

5’ 3’

DRAICRPLP1

KIF235’ 3’

**

**

**

*

+++

+++

-- -

-- -

+++

+++

-- -

-- -

+++

+++

-- -

-- -

+++

+++

-- -

-- -

0

0.2

0.4

0.6

0.8

1.0

1.2

Re

lative

Exp

ressio

n 1.4

R1881Bicalutamide

- + +- - +

DRAIC

00.20.40.60.81.01.2

Re

lative

Exp

ressio

n 1.4

R1881 - + - +

siGL2 siAR

LNCapDRAIC

LNCap

AR

Actin

WB

**

C DLNCap

Chr.15 (hg18)

on October 17, 2020. © 2015 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 20, 2015; DOI: 10.1158/1541-7786.MCR-15-0016-T

Page 27: Molecular Cancer Research - The lncRNA …...2015/02/20  · 4 repressed by AR (14,15). We also showed that multiple oncogenes, mTOR, SMARCD1, SNF2H and IGF1R targeted by miR-99a contribute

Figure 4. Sakurai et.al.

F

0

5000

10000

15000

20000

25000

30000

35000

40000

0 10000 20000 30000 40000 50000 60000 70000 80000FO

XA

1 e

xp

ressio

n (

RS

EM

)

NKX3-1 expression (RSEM)

ρ=0.463

p = 2.806e-27

PRAD (n=487) (TCGA)

H

FOXA1

Actin

WB

LNCap

siGL2no1 no2

siFOXA1

LNCap

Actin

WB

NKX3-1

no1 no2

siNKX3-1

1.41.21.00.80.60.40.2

0Re

lative

Exp

ressio

n

1.61.8

DRAICNKX3-1 mRNA

FOXA1NKX3-1

siGL2

FOXA1 mRNADRAICNKX3-1 mRNA

1.41.21.00.80.60.40.2

0Re

lative

Exp

ressio

n

1.61.8

1.4

1.2

1.0

0.8

0.6

0.4

0.20

Re

lative

Exp

ressio

n

DRAIC

LNCap

siR

NA

cD

NA

NKX3-1

siGL2siFOXA1siNKX3-1

Empty

FOXA1

**

**

G

C

0

5000

10000

15000

20000

25000

30000

35000

40000

0 1000 2000 3000 4000 5000

FO

XA

1 e

xp

ressio

n (

RS

EM

)

DRAIC expression (RSEM)

PRAD (n=487) (TCGA)

0

10000

20000

30000

40000

50000

60000

70000

80000

0 1000 2000 3000 4000 5000

NK

X3

-1 e

xp

ressio

n (

RS

EM

)

DRAIC expression (RSEM)

ρ = 0.198

p = 1.033e-05

ρ = 0.257

p = 9.063e-09

PRAD (n=487) (TCGA)

D E

A

B

LNCap

% In

pu

t

IgGFOXA1

1.41.21.00.80.60.40.2

0SLUG

promoter

FO

XA

1 C

hIP

-se

q

67,590,000 67,630,000

DRAIC RPLP1

KIF23

1 2 43 1 2 3 4

60

0

67,550,000

FOXA1 ChIP-PCR5’ 3’

DRAIC locus DRAIC locus

LNCapChr.15 (hg18)

NK

X3

-1 C

hIP

-se

q

60

0

67,590,000 67,630,000

RPLP1

KIF23

1 2 43

% In

pu

t

0.140.120.100.080.060.040.02

0

NKX3-1 ChIP-PCR

DRAIC locus

1 2 3 4

IgGNKX3-1

LNCap

DRAIC

67,550,000

5’ 3’

DRAIC locus

LNCap

Chr.15 (hg18)

VC

ap

LN

Ca

p

C4

-2

PC

3

DU

14

5

C4

-2B

PC

3M

-lu

c

AD cells CR cells

FOXA1

WB

FOXA1 mRNANKX3-1 mRNA

Re

lative

Exp

ressio

n

NKX3-1

Actin

1.41.21.00.80.60.40.2

0

1.61.82.0

on October 17, 2020. © 2015 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 20, 2015; DOI: 10.1158/1541-7786.MCR-15-0016-T

Page 28: Molecular Cancer Research - The lncRNA …...2015/02/20  · 4 repressed by AR (14,15). We also showed that multiple oncogenes, mTOR, SMARCD1, SNF2H and IGF1R targeted by miR-99a contribute

Figure 5. Sakurai et.al.

B

FO

XA

1 C

hIP

-se

q

0

6067,600,000 67,650,000 67,700,000 67,750,000

12 4

DRAIC loucs

DRAIC PCAT29

PCAT29 locus

LNCap

F

H

D

PCAT29

Re

lative

Exp

ressio

n

VC

ap

LN

Ca

p

C4

-2

PC

3

DU

14

5

CR cellsAD cells

C4

-2B

PC

3M

-lu

c

543210

67

x 4

.31

x 1

.0

x 0

.70

x 0

.95

x 0

.15

x 0

.02

x 0

.05

E

3

4

5

6

7

8

9

2 4 6 8

DRAIC ΔCt

PC

AT

29

ΔC

t 12 prostate cancer patients

(U. of Virginia)

R=0.62

p=0.01

NK

X3

-1 C

hIP

-se

q

0

6067,600,000 67,650,000 67,700,000 67,750,000

12 4

DRAIC locus

DRAIC PCAT29

PCAT29 locus

LNCapC

AA

R C

hIP

-se

q

0

600

6067,600,000 67,650,000 67,700,000 67,750,000

12 4

DRAIC locus

DRAIC PCAT29

DRAIC PCAT29

R1881 (-)

R1881 (+)

PCAT29 locus

LNCapChr.15 (hg18)

1.41.21.00.80.60.40.2

0

LNCap LNCap

PCAT29 PCAT29

Re

lative

Exp

ressio

n siGL2 no1 no2

siFOXA1

no1 no2

siNKX3-1

siGL2

1.41.21.00.80.60.40.2

0

Re

lative

Exp

ressio

n

siFOXA1siGL2R1881

1.41.21.00.80.60.40.2

0

Re

lative

Exp

ressio

n

siR

NA

PCAT29

LNCap

x 1.0

x 0.28 x 0.25x 0.05

0 0

69,550,000 69,650,00069,550,000 69,650,000LNCap

shCtrl, R1881(+) shFOXA1, R1881(+)

DRAIC PCAT29DRAIC PCAT29

LNCap

20

40

60

AR

Ch

IP-s

eq

Chr.15 (hg38) Chr.15 (hg38)

20

40

60

G

siFOXA1siGL2

R1881

1.41.21.00.80.60.40.2

0

Re

lative

Exp

ressio

n

siR

NA

DRAIC

LNCapx 1.0

x 0.42

x 0.25x 0.07

on October 17, 2020. © 2015 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 20, 2015; DOI: 10.1158/1541-7786.MCR-15-0016-T

Page 29: Molecular Cancer Research - The lncRNA …...2015/02/20  · 4 repressed by AR (14,15). We also showed that multiple oncogenes, mTOR, SMARCD1, SNF2H and IGF1R targeted by miR-99a contribute

Figure 6. Sakurai et.al.

E

050

100150200250

LNCap1.41.21.00.80.60.40.2

0

LNCap

Re

lative

Exp

ressio

n DRAIC shGL2 shDRAIC no1 shDRAIC no2 Empty DRAIC

PC3M-luc

Empty DRAIC

1.41.21.00.80.60.40.2

0

DRAIC

Empty DRAIC

PC3M-luc

no1 no2shGL2

shGL2 shDRAIC no1

LNCap

shDRAIC no2

0h

48

h

F

shGL2 shDRAIC no1

LNCap

shDRAIC no2 Empty DRAIC

PC3M-luc250200150100500no1 no2

shDRAIC

shGL2

100

120

8060

4020

0

Gap area (%)140

shGL2 sh no1 sh no2

0h 48h

****

*

*

**

D

G

H

A

VC

ap

LN

Ca

p

C4

-2B

C4

-2

PC

3

PC

3M

-lu

c

DU

14

50

0.40.60.81.01.21.41.61.8

Re

lative

Inva

de

d c

ell

nu

mb

ers

0.2

AD cells CR cells

B

Ce

ll n

um

be

rs (

X1

0 )

siDRAIC

0

1

2

3

4

5

6

siGL2 no1 no2 siGL2 no1 no2

siDRAIC

24h 72h

LNCap

6 1.41.21.00.8

0.6

0.40.2

0siGL2 no1 no2

siDRAIC

72h

LNCap

Re

lative

Exp

ressio

n

DRAIC

Re

lative

Exp

ressio

n

C

0h

24

h

Empty DRAIC

PC3M-luc

Emp

DRAIC

100120

80604020

0

140

*

0h 24h

Gap area (%)

Invaded cell numbers

Invaded cell numbers

Matrigel Invasion assay Cell growth

on October 17, 2020. © 2015 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 20, 2015; DOI: 10.1158/1541-7786.MCR-15-0016-T

Page 30: Molecular Cancer Research - The lncRNA …...2015/02/20  · 4 repressed by AR (14,15). We also showed that multiple oncogenes, mTOR, SMARCD1, SNF2H and IGF1R targeted by miR-99a contribute

Figure 7. Sakurai et.al.

androgen

ARAR FOXA1NKX3-1

Invasion ability / low

Androgen Dependent (AD)

prostate cancer cells

Castration Resistant (CR)

prostate cancer cells

Low androgen

ARARFOXA1

NKX3-1

Invasion ability / high

Disease free survival / low

Turmor Progression

Androgen Deprivation Therapy (ADT)

tumor suppressive lncRNA cluster

DRAIC high PCAT29 lowDRAIC low

AR pathways activated

by androgen

High experssions of

FOXA1 and NKX3-1

AR pathways activated

despite low androgen

Low experssions of

FOXA1 and NKX3-1

tumor suppressive lncRNA cluster

PCAT29 high

Chr.15 Chr.15

A

B

DRAIC PCAT29

transcript size

cellular localization

Kaplan Meier plot

negative regulator

positive regulator

cell growth

migration, invasion

694bp1702bp

poly adenylation Yes Yes

cytoplasm nuclear

University of MichiganMSKCC, Cancer Cell, 2010good prognostic marker good prognostic marker

AR AR

FOXA1, NKX3-1 FOXA1, NKX3-1

induce repress

repress repress

on October 17, 2020. © 2015 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 20, 2015; DOI: 10.1158/1541-7786.MCR-15-0016-T

Page 31: Molecular Cancer Research - The lncRNA …...2015/02/20  · 4 repressed by AR (14,15). We also showed that multiple oncogenes, mTOR, SMARCD1, SNF2H and IGF1R targeted by miR-99a contribute

Published OnlineFirst February 20, 2015.Mol Cancer Res   Kouhei Sakurai, Brian J. Reon, Jordan Anaya, et al.   suppressive nexus

locus constitutes a tumorDRAIC/PCAT29The lncRNA

  Updated version

  10.1158/1541-7786.MCR-15-0016-Tdoi:

Access the most recent version of this article at:

  Material

Supplementary

  http://mcr.aacrjournals.org/content/suppl/2015/02/21/1541-7786.MCR-15-0016-T.DC1

Access the most recent supplemental material at:

  Manuscript

Authoredited. Author manuscripts have been peer reviewed and accepted for publication but have not yet been

   

   

   

  E-mail alerts related to this article or journal.Sign up to receive free email-alerts

  Subscriptions

Reprints and

  [email protected] at

To order reprints of this article or to subscribe to the journal, contact the AACR Publications

  Permissions

  Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

.http://mcr.aacrjournals.org/content/early/2015/02/20/1541-7786.MCR-15-0016-TTo request permission to re-use all or part of this article, use this link

on October 17, 2020. © 2015 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 20, 2015; DOI: 10.1158/1541-7786.MCR-15-0016-T