ArticlePDF Available

ESR1 Is Co-Expressed with Closely Adjacent Uncharacterised Genes Spanning a Breast Cancer Susceptibility Locus at 6q25.1

PLOS
PLOS Genetics
Authors:

Abstract and Figures

Approximately 80% of human breast carcinomas present as oestrogen receptor α-positive (ER+ve) disease, and ER status is a critical factor in treatment decision-making. Recently, single nucleotide polymorphisms (SNPs) in the region immediately upstream of the ER gene (ESR1) on 6q25.1 have been associated with breast cancer risk. Our investigation of factors associated with the level of expression of ESR1 in ER+ve tumours has revealed unexpected associations between genes in this region and ESR1 expression that are important to consider in studies of the genetic causes of breast cancer risk. RNA from tumour biopsies taken from 104 postmenopausal women before and after 2 weeks treatment with an aromatase (oestrogen synthase) inhibitor was analyzed on Illumina 48K microarrays. Multiple-testing corrected Spearman correlation revealed that three previously uncharacterized open reading frames (ORFs) located immediately upstream of ESR1, C6ORF96, C6ORF97, and C6ORF211 were highly correlated with ESR1 (Rs = 0.67, 0.64, and 0.55 respectively, FDR
Content may be subject to copyright.
ESR1
Is Co-Expressed with Closely Adjacent
Uncharacterised Genes Spanning a Breast Cancer
Susceptibility Locus at 6q25.1
Anita K. Dunbier
1,2
*, Helen Anderson
1,2
, Zara Ghazoui
1,2
, Elena Lopez-Knowles
1,2
, Sunil Pancholi
2
,
Ricardo Ribas
2
, Suzanne Drury
1,2
, Kally Sidhu
1
, Alexandra Leary
1
, Lesley-Ann Martin
2
, Mitch Dowsett
1,2
1Royal Marsden Hospital, London, United Kingdom, 2Breakthrough Breast Cancer Research Centre, Institute of Cancer Research, London, United Kingdom
Abstract
Approximately 80% of human breast carcinomas present as oestrogen receptor a-positive (ER+ve) disease, and ER status is a
critical factor in treatment decision-making. Recently, single nucleotide polymorphisms (SNPs) in the region immediately
upstream of the ER gene (ESR1) on 6q25.1 have been associated with breast cancer risk. Our investigation of factors
associated with the level of expression of ESR1 in ER+ve tumours has revealed unexpected associations between genes in
this region and ESR1 expression that are important to consider in studies of the genetic causes of breast cancer risk. RNA
from tumour biopsies taken from 104 postmenopausal women before and after 2 weeks treatment with an aromatase
(oestrogen synthase) inhibitor was analyzed on Illumina 48K microarrays. Multiple-testing corrected Spearman correlation
revealed that three previously uncharacterized open reading frames (ORFs) located immediately upstream of ESR1,C6ORF96,
C6ORF97, and C6ORF211 were highly correlated with ESR1 (Rs = 0.67, 0.64, and 0.55 respectively, FDR,1610
27
). Publicly
available datasets confirmed this relationship in other groups of ER+ve tumours. DNA copy number changes did not
account for the correlations. The correlations were maintained in cultured cells. An ERaantagonist did not affect the ORFs’
expression or their correlation with ESR1, suggesting their transcriptional co-activation is not directly mediated by ERa.
siRNA inhibition of C6ORF211 suppressed proliferation in MCF7 cells, and C6ORF211 positively correlated with a proliferation
metagene in tumours. In contrast, C6ORF97 expression correlated negatively with the metagene and predicted for improved
disease-free survival in a tamoxifen-treated published dataset, independently of ESR1. Our observations suggest that some
of the biological effects previously attributed to ER could be mediated and/or modified by these co-expressed genes. The
co-expression and function of these genes may be important influences on the recently identified relationship between
SNPs in this region and breast cancer risk.
Citation: Dunbier AK, Anderson H, Ghazoui Z, Lopez-Knowles E, Pancholi S, et al. (2011) ESR1 Is Co-Expressed with Closely Adjacent Uncharacterised Genes
Spanning a Breast Cancer Susceptibility Locus at 6q25.1. PLoS Genet 7(4): e1001382. doi:10.1371/journal.pgen.1001382
Editor: Marshall S. Horwitz, University of Washington, United States of America
Received September 15, 2010; Accepted March 25, 2011; Published April 28, 2011
Copyright: ß2011 Dunbier et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits
unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Funding: This study was supported by the Mary-Jean Mitchell Green Foundation, Breakthrough Breast Cancer, and NHS funding to the Royal Marsden NIHR
Biomedical Research Centre. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
Competing Interests: MD has received honoraria for advice and lectures for Astra Zeneca and has acted as an expert witness for them.
* E-mail: anita.dunbier@icr.ac.uk
Introduction
Breast cancer is the most common malignancy in women,
accounting for more than 400,000 deaths per year worldwide [1].
Approximately 80% of human breast carcinomas present as
oestrogen receptor a-positive (ER+ve) disease and ER status is
arguably the most clinically important biological factor in all
oncology [2]. The major molecular features of breast cancer
segregate differentially between ER+ve and ER2ve tumours [3,4].
Tumours which express ERahave been termed luminal type [3,5]
and are associated with response to antioestrogen therapy and
improved survival, although the mechanisms by which oestrogen
receptor dictates tumour status are poorly understood.
Recent genome wide studies have identified SNPs around
C6ORF97, an open reading frame (ORF) immediately upstream of
the gene encoding ER (ESR1) to be associated with increased risk
of breast cancer. Zheng et al. found that heterozygosity at
rs2046210, a SNP in the region between C6ORF97 and ESR1,
increased breast cancer risk by an odds ratio of 1.59 in a Chinese
population and that this risk was also present in a European
population, albeit to a weaker extent [6]. Easton and colleagues
confirmed the risk associated with this SNP and reported an at
least partly independent risk associated with a second adjacent
SNP (rs3757318) in intron 7 of C6ORF97 [7]. Using ancestry-shift
refinement mapping, Stacey et al. closed in on the identification of
the pathogenic variant and found that the risk allele of a novel
SNP in this region (rs77275268), disrupts a partially methylated
CpG sequence within a known CTCF binding site [8]. More
recently, two further studies have confirmed an association with
the region [9,10]. Our studies have revealed unexpected
relationships in the expression patterns in breast carcinomas
between ESR1,C6ORF97 and the two genes immediately
upstream (C6ORF211 and C6ORF96 [RMND1]).
Oestrogenic ligands, predominantly oestradiol, are the key
mitogens for ER+ve breast cancer. In recent years, high
throughput genomic technologies have revealed significant
numbers of genes that are expressed in response to oestradiol
stimulation in vitro [11–13] and downregulated in response to
oestrogen deprivation in tumours [14–16]. Similarly, the tran-
scriptional targets of ERahave been characterised in detail using
PLoS Genetics | www.plosgenetics.org 1 April 2011 | Volume 7 | Issue 4 | e1001382
genome wide chromatin interaction mapping in MCF7 cells
[17,18]. Key oestrogen responsive genes such as TFF1 and GREB1
have been shown to be highly responsive to oestradiol stimulation
in cell culture models through the binding of ERato their
promoters [19,20]. Additional genes have been found in
hierarchical clustering analyses of ER+ve and ER2ve tumours
as part of the so-called ‘‘luminal epithelial’’ gene set characterized
by the expression of genes typically expressed in the cells that line
the ducts of normal mammary glands including GATA3 and
FOXA1 [12]. However, the correlates of ESR1 within an
exclusively ER+ve group and the inherent heterogeneity within
an exclusively ER+subgroup remain poorly defined.
Modern, non-steroidal aromatase inhibitors (AIs) are widely
used, effective treatments for ER+ve breast cancer [21,22] and are
also excellent pharmacological probes for oestrogen-dependent
processes in vivo because of their specificity and highly effective
suppression of oestrogen synthesis. In this study, we found that the
expression of genes in the region immediately upstream of ESR1
associate strongly with ESR1 expression in ER+ve primary breast
cancers before and after AI treatment and uncover evidence that
these associations might impact upon the biological and clinical
importance of ERa.
Results
ESR1 expression is correlated with three open reading
frames on chromosome 6 in tumours
To investigate correlates of ESR1, expression profiles were
derived from pairs of 14-guage core cut biopsies before and after 2
weeks’ treatment with 1 mg/d anastrozole, an AI, from 104
patients with ER+ve primary breast cancer [23]. Genes whose
expression correlated with expression of ESR1 levels pre-treatment
were identified (Spearman corrected for multiple testing at false
discovery rate ,1610
27
, Table 1 pre-treatment). The mRNA
species most highly correlated with ESR1 were chromosome 6
ORF 97 (C6ORF97, Rs = 0.67) (Figure 1a), followed by
C6ORF211. Other notable inclusions amongst the top 20 most
correlated genes included well-established ER-associated genes
such as FOXA1,MYB and GATA3, plus C6ORF96, also known as
RMND1 (Required for Meiotic Nuclear Division 1 homolog). The
mean pre-treatment expression of the three ORFs was highly
correlated with ESR1 (Rs = 0.70, Figure 1b). After 2 weeks’ AI
treatment, the top three genes correlating with ESR1 were
C6ORF96, C6ORF97 and C6ORF211 (Rs.0.7 for all, Table 1
two weeks post-treatment). These three ORFs are all located less
than 0.5 MB upstream of the ESR1 start site on the q arm of
chromosome 6 (Figure 1e). The expression of other genes located
within a 50 MB region surrounding ESR1 were not correlated
with ESR1 expression (Rs,0.25) (Table S1).
The correlation was present in all of five published microarray
data sets of ER+ve breast cancer in which the C6orfs were
included on the array (Table 2). The expression of the three ORFs
was lower in ER2ve than ER+ve tumours in the Wang dataset
[24] (p = 0.002). No significant correlation was found in the
ER2ve subgroup of this dataset. This may be a characteristic of
ER2ve tumours or, alternatively, the measurement error
associated with low levels of ESR1 transcript could preclude
detection of a significant correlation in microarray data.
Correlation between ESR1 and the C6orfs is not explained
by amplification
Amplification of the ESR1 locus has been reported inconsis-
tently [25,26]. To determine whether the ESR1/C6ORFs correla-
tion may be the result of underlying genomic co-amplification or
deletion events, copy number (CN) status of ESR1 and the C6orfs
was examined using array CGH analysis (resolution 40–60 kb)
[27] on DNA from the 44 tumour samples from which adequate
further tissue was available. One tumour was shown to be
amplified and eight showed gains at ESR1,C6ORF96,C6ORF97
and C6ORF211, while four showed losses at all four loci. One was
measured as having loss of C6ORF96,C6ORF211 and part of
C6ORF97. While there was some correlation between CN and
transcription of the four genes (Figure S1), CN alterations did not
explain the correlation between ESR1 and the C6orfs. In fact,
when samples with identified CN changes were removed from the
dataset, the correlation between ESR1 and mean C6orf expression
levels strengthened rather than weakened (Rs = 0.83) (Figure 1c),
suggesting that transcriptional co-regulation rather than genomic
changes is more likely to underlie ESR1/C6ORF co-expression.
Change in ESR1 expression upon aromatase inhibitor
treatment is correlated with change in C6orf expression
To assess whether the correlation in ESR1/C6ORF expression
seen in pre-treatment biopsies is reflected in a concordant change in
expression of these genes upon treatment, the relationship between
the magnitude of change of each of these genes was investigated.
Change in expression of ESR1 induced by aromatase inhibitor
treatment over 2 weeks was strongly correlated with change in the
C6orfs (Rs = 0.70) (Figure 1d). Given that this short duration of
treatment, which has no measurable impact on cellularity or tumour
size, is unlikely to facilitate DNA copy number changes throughout
the sample this supports the probability that the co-regulation of
these genes is at a transcriptional level.
Expression of ESR1 and the C6orfs are correlated in MCF7
and BT-474 cells in vitro
To determine whether the ESR1/C6ORF correlations were
maintained in vitro, transcript levels of ERaand the three C6orfs
were measured in oestrogen-deprived MCF7 cells and lapatinib-
treated BT-474 cells over a 48- and 96-hour period, respectively.
These treatments are both known to have significant effects on the
Author Summary
Recent genome-wide analysis has revealed that the way in
which genes are arranged on chromosomes and the
conformation of these chromosomes are crucial for the
regulation of gene expression. Reflecting this arrange-
ment, clusters of genes which are regulated together have
been discovered. We have identified a previously unre-
ported transcriptional activity hub spanning ESR1, the
gene encoding the important breast cancer biomarker
oestrogen receptor. Genetic variants immediately up-
stream of ESR1 have recently been linked to breast cancer
risk. We found that three open reading frames within this
region are tightly co-expressed with ESR1. We investigated
the function of these genes and discovered that one of
these co-expressed genes, C6ORF211, affects proliferation
in cultured cells and is correlated with proliferation in
breast tumours. Another of the genes, C6ORF97,is
negatively correlated with proliferation in breast tumours
and predicts for outcome on the anti-oestrogen drug
tamoxifen. These findings suggest that the genes could
contribute to the phenotype associated with oestrogen-
receptor positivity. In addition, they may be involved in the
mechanism by which genetic variation in this region of the
genome contributes to breast cancer susceptibility.
ESR1 Is Co-Expressed with Closely Adjacent Genes
PLoS Genetics | www.plosgenetics.org 2 April 2011 | Volume 7 | Issue 4 | e1001382
expression of ESR1. Lapatinib has been shown to increase ERain
BT-474 cells [28,29], potentially via loss of Akt and de-repression of
FOXO3a. This provides a useful model for manipulation to test the
correlation between ESR1 and the C6orfs in vitro. Conversely,
absence of oestradiol leads to a short-term reduction in ER
expression [30]. Expression of all four genes followed a similar time-
course of expression and was highly correlated (Figure 2a and 2b).
ICI 182,780 (ICI) is a steroidal pure anti-oestrogen which causes
ERaexpression to be suppressed and downregulated [31,32].
Treatment of MCF7 cells with ICI did not affect ORF expression
or their correlation with ESR1 (Figure 2c). To confirm that the
observed correlation was not being influenced by RNA transcribed
prior to the addition of ICI, we also measured newly synthesised
nascent RNA using PCR amplicons designed to cross an exon/
intron boundary [33]. This analysis revealed that nascent
transcripts for ESR1 and the C6orfs remained correlated in both
the presence and absence of ICI. The observation that
transcription of the genes remains strongly correlated in the
presence of ICI suggests that transcriptional regulation by ERais
not the main driver of the ESR1/C6ORF co-expression.
Knockdown of C6ORF211 by siRNA induces a reduction in
proliferation in MCF7cells
The effect of reducing expression of each C6orf on cell
proliferation was determined by transfecting siRNA SMART-
POOLs directed against each ORF into MCF7 cells. In cells
grown in both E2-containing media and without E2, all three
siRNAs reduced transcript levels of their target ORF to ,30% of
levels in cells transfected with the control non-targeting siRNA
pool. Levels of ESR1, and the non-targeted ORFs were unaffected
by the SMARTpool’s (Figure S2) while ESR1-SMARTpool
siRNA led to a reduction in levels of all three C6orfs (Figure
S3). Immunoblotting with a polyclonal antibody raised against a
polypeptide of the predicted product of C6ORF211 showed an
86% reduction at the protein level (Figure S4). Cells transfected
with C6ORF211 siRNA showed a mean 36% reduction in cell
number (p,0.0001) over four separate repeat experiments
(Figure 3A). C6ORF211 knockdown had no effect on oestrogen-
dependent proliferation (Figure 3B). Deconvolution of the
SMARTPOOL showed that the four constituent siRNAs had a
reproducible anti-proliferative effect when compared with scram-
bled control siRNA (Figure S5). No consistent alteration in
proliferation was observed in cells transfected with siRNAs
directed against C6ORF96 or C6ORF97 (Figure 3A).
C6ORF211 correlates with proliferation and clinical
outcome in tumours
To determine whether the association between C6ORF211
expression and proliferation seen in cultured cells is reflected in
tumours, the relationship between C6ORF211 expression and a
metagene composed of known proliferation-associated genes [34]
was investigated. In baseline biopsies, levels of C6ORF211 but not
ESR1 correlated significantly with proliferation (C6ORF211,
Rs = 0.23, p = 0.04; ESR1,Rs=20.01, p = ns) (Figure 4a), suggest-
ing that C6ORF211 is more strongly associated with proliferation
than ESR1. Correlations were also observed with a number of well-
known proliferation-associated genes (Table S2). The relationship
with proliferation was validated in data from a set of 354 ER+ve
tumours [35] (Rs = 0.18, p = 0.0008) (Figure 4b) and the 209 ER+ve
tumours from the Wang dataset [24] (Rs = 0.21, p = 0.004).
Consistent with the findings in our own data, ESR1 was not
significantly correlated with the proliferation metagene in either of
the publicly available datasets (Loi, Rs = 20.03, p = ns; Wang, Rs =
0.02, p = ns). In contrast, C6ORF97 showed an independent,
reproducible negative correlation with proliferation, in our dataset
(Rs = 20.19, p = 0.05) and in the Loi (Rs = 20.22, p,0.0001)
(Figure 4c) and ER+ve Wang datasets (Rs = 20.24, p = 0.0007).
Table 1. Genes positively correlated with ESR1 gene
expression ranked according to Spearman correlation.
GB acc Gene symbol Cytoband
Correlation
coefficient
Pre-treatment
1NM_000125 ESR1 6q25.1 1
2NM_025059 C6orf97 6q25.1 0.672
3NM_024573 C6orf211 6q25.1 0.637
4NM_152437 ZNF664 12q24.31 0.608
5NM_019000 FLJ20152 5p15.1 0.562
6NM_015391 ANAPC13 3q22.1 0.552
7NM_018718 TSGA14 7q32 0.547
8NM_017909 C6orf96 6q25.1 0.546
9NM_021627 SENP2 3q27.2 0.545
10 NM_012319 SLC39A6 18q12.2 0.544
11 NM_004496 FOXA1 14q12-q13 0.537
12 NM_005001 NDUFA7 19p13.2 0.534
13 NM_207118 GTF2H5 6q25.3 0.532
14 NM_004703 RABEP1 17p13.2 0.528
15 NM_016058 TPRKB 2p24.3-p24.1 0.528
16 NM_005375 MYB 6q22-q23 0.527
17 NM_175924 ILDR1 3q13.33 0.526
18 NM_173079 RUNDC1 17q21.31 0.526
19 NM_032918 RERG 12p12.3 0.523
20 NM_002051 GATA3 10p15 0.523
2 weeks post-treatment
1NM_000125 ESR1 6q25.1 1
2NM_025059 C6orf97 6q25.1 0.741
3NM_017909 C6orf96 6q25.1 0.718
4NM_024573 C6orf211 6q25.1 0.705
5NM_004703 RABEP1 17p13.2 0.688
6NM_006452 PAICS 4q12 0.658
7NM_004496 FOXA1 14q12-q13 0.637
8NM_020784 KIAA1344 14q22.1 0.632
9NM_018199 EXDL2 14q24.1 0.629
10 NM_002222 ITPR1 3p26-p25 0.629
11 NM_181656 C17orf58 17q24.2 0.625
12 NM_002051 GATA3 10p15 0.623
13 NM_005080 XBP1 22q12.1|22q12 0.621
14 NM_012319 SLC39A6 18q12.2 0.62
15 NM_015575 TNRC15 2q37.1 0.619
16 NM_173079 RUNDC1 17q21.31 0.615
17 NM_015130 TBC1D9 4q31.21 0.608
18 NM_138809 LOC134147 5p15.2 0.598
19 NM_006405 TM9SF1 14q11.2 0.592
20 NM_152416 C8orf38 8q22.1 0.587
All genes shown have parametric p-value and false discovery rates ,1e-07.
doi:10.1371/journal.pgen.1001382.t001
ESR1 Is Co-Expressed with Closely Adjacent Genes
PLoS Genetics | www.plosgenetics.org 3 April 2011 | Volume 7 | Issue 4 | e1001382
To determine whether the relationship of the ORFs with
proliferation is related to clinical outcome, recurrence free survival
(RFS) in tamoxifen-treated patients was investigated for association
with C6ORF97 and C6ORF211 expression. Despite the fact that in
the Loi dataset ESR1 was not predictive of a significant difference in
survival over 5 years [36], the lowest quartile of C6ORF97 was
associated with significantly higher risk of recurrence (HR = 3.1,
p = 0.0014) (Figure 4d). A similar trend was observed in untreated
ER+ve tumours from the Wang dataset [24], although this was not
significant (HR = 1.6, p = 0.16) (Figure S6a). C6ORF211 was not
significantly associated with RFS (Figure S6b and S6c).
Discussion
Our observation of a previously unreported transcriptional
activity hub in the ESR1/C6ORF region of 6q25.1 has implications
for recently identified associations between SNPs in the ESR1
region and breast cancer risk, as well as broader implications for
the biological and clinical importance of ERain established breast
cancer. A number of SNPs, including rs3757318 within intron 7 of
C6ORF97 [7], have been associated with breast cancer risk but the
causative variant and mechanism remain undefined [6–10]. In an
attempt to identify the pathogenic variant, Stacey and colleagues
recently reported that GG homozygotes at rs9397435, located
immediately downstream of C6ORF97, may express higher mean
levels of ESR1 and that the rs9397435 [G] allele conferred
significant risk of both hormone receptor positive and hormone
receptor negative breast cancer in European and Taiwanese
patients [8]. The association of a SNP in this region with ER
expression is consistent with findings from our own group which
have revealed that the variant genotype of SNP rs2046210 is
associated with increased ERaexpression as measured by
Figure 1. Correlation of
ESR1
expression and oestrogen-responsive gene expression. a. Scatterplot of relationship between expression of
ESR1 and C6ORF97 in baseline biopsies. b. Correlation between expression of ESR1 and the mean of C6ORF96,C6ORF97 and C6ORF211 in baseline
biopsies. c. Correlation between ESR1 and the mean of C6ORF96,C6ORF97 and C6ORF211 with samples with measured copy number variations shown
omitted. d. Scatterplot of relationship between change in ESR1 and the mean change in C6ORF96,C6ORF97 and C6ORF211 e. Location of open
reading frames, ESR1 and breast cancer associated SNPs on chromosome 6q25.1.
doi:10.1371/journal.pgen.1001382.g001
ESR1 Is Co-Expressed with Closely Adjacent Genes
PLoS Genetics | www.plosgenetics.org 4 April 2011 | Volume 7 | Issue 4 | e1001382
immunohistochemistry [37]. The findings reported in this paper
suggest that, due to their high degree of correlation with ESR1,
levels of C6ORF97,C6ORF96 and C6ORF211 are also likely to
correlate with the rs2046210 and rs9397435 genotype. Conse-
quently, these genes may be involved in the pathogenesis of the
variant SNPs and could explain the apparent anomaly noted by
Stacey and colleagues in that the SNPs predispose to both
hormone receptor positive and negative disease.
To date, analysis of ESR1 co-expressed genes has focussed on
genes which are also downstream targets of the oestradiol-
activated transcription factor activity of ERasuch as FOXA1,
TFF1 and GATA3. High throughput technologies have identified
numerous classical and novel ERa-dependent targets of oestradiol
[11,17]. This association with the expression of ORFs has,
however, not been reported other than by ourselves in abstract
form [38].
The transcriptional correlation between ESR1 and these ORFs
is highly statistically significant in our dataset, and in all of the
publicly available datasets we examined. In our own patient
cohort, we showed that two weeks’ treatment with anastrozole
induces a concomitant change in ESR1 and the C6orfs and a yet
stronger correlation in their expression. Genomic amplification
does not account for the correlations. This suggests that
transcriptional co-regulation rather than major genomic rear-
rangement is likely to underlie their co-expression. To our
knowledge, a transcriptional activity hub surrounding a major
cancer related gene has not previously been identified.
The observation that the four transcripts remain correlated over
a short timecourse in MCF7 and BT474 cells further supports the
idea that the co-regulation of these genes is likely to occur at a
transcriptional level. Given that ERacan autoregulate its own
transcription by binding to an oestrogen responsive element (ERE)
in its promoter [17,39], the possibility that ERacould co-regulate
itself and the C6orfs provides an attractive potential explanation
for the correlation. We tested this hypothesis by treating MCF7
cells with the ERaantagonist ICI in the absence of E2. Our
finding that the nascent transcripts of ESR1 and the three C6orfs
remain correlated in the presence of ICI (Figure 2c) suggests that
this co-regulation is not dependent on ERatranscriptional
activation.
Regulation of the steady-state level of ERain breast cancer cells
is a complex phenomenon that includes transcriptional and post-
transcriptional mechanisms [40–42]. C6ORF96 is transcribed off
the opposite DNA strand to ESR1 (Figure 1e), therefore excluding
the possibility that ESR1 and the ORFs are transcribed as a single
polycistronic mRNA. Recent genome-wide mapping experiments
have revealed the importance of chromatin organisation for gene
expression [18,43] suggesting that 3-D chromatin arrangement
could represent a potential explanation for C6ORF/ESR1 co-
expression. However, analysis of the data produced by Fullwood
and colleagues [18] shows that C6ORF96,C6ORF97 and
C6ORF211 are not encompassed by an ERa-bound long-range
chromatin loop. Nevertheless, it remains possible that a loop
driven by an alternative transcription factor could explain the
transcriptional activity in this area.
At the nucleotide level, all three ORFs show some homology
with ESR1, suggesting they may have arisen from gene duplication
events [44]. C6ORF97 encodes a 715 amino acid coiled-coil
domain-containing protein that is conserved across 11 species [45]
while C6ORF211 is a member of the UPF0364 protein family of
unknown function and is also conserved across multiple species
[45]. Confocal analysis revealed that the protein encoded by
C6ORF211 was expressed mainly in the cytoplasm and did not co-
localize with ER (Figure S7). In a proteomic screen it has been
found to interact with SAP18, a Sin3A-associated cell growth
inhibiting protein [46].
This reported interaction with a growth inhibitory protein could
explain our observation that knockdown of C6ORF211 induces
suppression of proliferation in cultured cells. This association is
mirrored in tumours, where a proliferation metagene correlates
significantly with C6ORF211. Conversely, C6ORF97 expression
correlates negatively with expression of the proliferation metagene
and high C6ORF97 predicts for improved disease-free survival in a
tamoxifen-treated published dataset, independently of ESR1
(Figure 4d). As high ESR1 has previously been shown to be
associated with improved outcome on endocrine therapy [47], this
raises the possibility that, given the observed correlation of
C6ORF97 with ESR1, some of this association with outcome could
be attributable to C6ORF97.
The high degree of correlation between ESR1 and the C6orfs
has significant potential implications for our interpretation of ER
levels and therapy of ER+ve breast cancers. As a transducer of
mitogenic oestrogen signalling, disruption of ER represents a key
target of therapies for ER+ve breast cancer, including tamoxifen
and fulvestrant. Our data shows that C6ORF211 and C6ORF97
may contribute to the proliferative phenotype of ER+ve tumours,
yet these proteins are unlikely to be affected by therapies targeted
directly at ERa. Consequently, these proteins may represent
potential targets for synergistic therapies in patients with high
levels of C6orf expression or targets for breast cancer prevention.
In addition, along with further research these relationships could
shed light on recent associations between breast cancer risk and
SNPs in the region.
Materials and Methods
Patient samples
Core-cut tumor biopsies (14-gauge) were obtained from 112
postmenopausal women with stage I to IIIB ER+early breast
cancer before and after two-weeks’ anastrozole treatment in a
neoadjuvant trial [23]. This study received approval from an
institutional review board at each site and was conducted in
accordance with the 1964 Declaration of Helsinki [48] and
International Conference on Harmonization/Good Clinical
Practice guidelines. Written informed consent was obtained from
each patient before participation. Tissue was stored in RNAlater
at 220uC. Two 4 mm sections from the core were stained with
Table 2. Correlations in other breast cancer datasets.
Study Number of samples
C6ORF96 C6ORF97 C6ORF211
TransBig [52] 198 breast tumours 0.607 0.776 0.656
Wang – All
tumours [24]
286 breast tumours 0.524 0.558 0.769
-ER+ve 209 breast tumours 0.388 0.418 0.608
-ER2ve 65 breast tumours 0.056 0.189 0.087
Loi [35] 354 breast tumours 0.468 0.555 0.588
Huang [53] 23 primary cell lines 0.759 0.759 0.878
Miller [51] 251 breast tumours 0.623 0.547 0.674
Data from five large, publicly available breast cancer datasets performed on
Affymetrix U133A arrays which contained probes for ESR1,C6ORF96,C6ORF97,
and C6ORF211 were examined. The mean of all probes for ESR1 was correlated
with each of the three C6ORFs. Correlation co-efficients for each of the genes
versus ESR1 is shown.
doi:10.1371/journal.pgen.1001382.t002
ESR1 Is Co-Expressed with Closely Adjacent Genes
PLoS Genetics | www.plosgenetics.org 5 April 2011 | Volume 7 | Issue 4 | e1001382
hematoxylin and eosin to confirm the presence of cancerous tissue
and the histopathology and six 8 mm sections were retained for
microarray CGH analysis (see below). Total RNA was extracted
using RNeasy Mini kits (Qiagen, Sussex, UK). RNA quality was
checked using an Agilent Bioanalyser (Santa Clara, CA, USA):
samples with RNA integrity values of less than 5 were excluded
from further analysis. ER status and Ki67 values by immunohis-
tochemistry were already available [23].
Gene expression analysis and data pre-processing
RNA amplification, labelling and hybridization on HumanWG-
6 v2 Expression BeadChips were performed according to the
manufacturer’s instructions (http://www.illumina.com) at a single
Illumina BeadStation facility. Tumor RNA of sufficient quality
and quantity was available to generate expression data from 104
pre-treatment biopsies. Data was extracted using BeadStudio
software and normalized with variance-stabilizing transformation
(VST) and Robust Spline Normalisation method (RSN) in the
Lumi package [49]. Probes that were not detected in any samples
(detection p value .1%) were discarded from further analysis.
Data analysis
Multiple correlation analysis was performed in BRB-Array
Tools (http://linus.nci.nih.gov/BRB-ArrayTools.html). A statisti-
cal significance level for each gene for testing the hypothesis that
the Spearman’s correlation between expression of ESR1 and other
genes was zero was calculated and p-values were then used in a
multivariate permutation test [50] from which false discovery rates
were computed. Other statistical analyses were performed in SPSS
for Windows (SPSS Inc., Chicago, IL), S-PLUS (TIBCO Software
Inc., Palo Alto, CA) and Graphpad Prism (Graphpad Software
Inc., La Jolla, CA).
Multivariable analysis was performed in a forward stepwise
fashion, the most significant additional variable (satisfying p,0.05)
being added at each stage. Cases with missing values for any of the
variables in the model were excluded from analysis.
Figure 2. Correlation of C6orf expression
in vitro
.a. Timecourse of expression of ESR1,C6ORF96,C6ORF97 and C6ORF211 in MCF7 cells cultured
in the absence of oestradiol. Each gene is normalized to the mean of two housekeeping genes, TBP and FKBP15. b. Timecourse of expression of ESR1,
C6ORF96,C6ORF97 and C6ORF211 in BT-474 cells after addition of lapatinib. c. Timecourse of expression of ESR1,C6ORF96,C6ORF97 and C6ORF211 in
MCF7 cells cultured with the addition of ICI 182,780. d. Analysis of expression of nascent ESR1,C6ORF96,C6ORF97 and C6ORF211 in MCF7 cells.
e. Analysis of expression of nascent ESR1,C6ORF96,C6ORF97 and C6ORF211 in MCF7 cells treated with ICI. Points represent the mean of three
triplicate samples 6SEM.
doi:10.1371/journal.pgen.1001382.g002
ESR1 Is Co-Expressed with Closely Adjacent Genes
PLoS Genetics | www.plosgenetics.org 6 April 2011 | Volume 7 | Issue 4 | e1001382
Figure 3. Exploration of the function of the C6orfs in MCF7 cells. a. Wild type-MCF7 cells were stripped of steroid for 48 hours then
transfected with either control siRNA, siRNA SMARTpool for C6ORF96,C6ORF97 or C6ORF211. b. Stripped MCF7 cells were transfected with C6ORF211
siRNA SMARTpool and 48 hours post transfection these were treated with increasing concentrations of oestradiol. After 6 days, proliferation in
response to siRNA knockdown was established by change in cell number using a Coulter counter. Bars represent the mean 6SEM of four separate
repeats of the experiment. Oestradiol-dependent proliferation is shown as fold change relative to cells with no added oestradiol.
doi:10.1371/journal.pgen.1001382.g003
ESR1 Is Co-Expressed with Closely Adjacent Genes
PLoS Genetics | www.plosgenetics.org 7 April 2011 | Volume 7 | Issue 4 | e1001382
Analysis of publicly available datasets
For analysis of the breast cancer datasets from public resources
the publicly available normalised gene expression data and clinical
data were retrieved from Gene Expression Omnibus (http://www.
ncbi.nlm.nih.gov/geo/) (‘Wang’ dataset [24], n = 286;GEO,
accession number GSE2034) or obtained from the authors (‘Loi’
dataset [35], n = 354 tamoxifen-treated tumours composed of
GEO, accession numbers GSE9195, GSE6532 and GSE2990;
combined normalised dataset received courtesy of Dr Christos
Sotiriou). Correlations between ESR1 and the C6orfs in the
‘Miller’ [51] (n = 251), ‘TransBig’ (n = 198) [52] and Huang [53]
(n = 23 cell lines) were calculated using the correlation analysis tool
in Oncomine (http://www.oncomine.org).
Data from the 72 genes comprising the proliferation metagene
was retrieved from tumours from the Wang and Loi datasets and
proliferation metagene scores were calculated as described
previously [54]. Spearman correlation between the proliferation
metagene and ESR1 and the C6orfs was calculated in Graphpad
Prism. Survival analysis was carried out in these datasets using the
quartiled expression of the C6orfs and the endpoints of recurrence
free survival or time to relapse, according to the original publication.
DNA extraction
Five 8 mm sections from frozen core biopsies were mounted
onto Superfrost glass slides, stained with nuclear fast red, and
microdissected with a sterile needle under a stereomicroscope to
obtain a percentage of tumor cells .75% as described previously
[55]. Genomic DNA was extracted as described previously [55].
The concentration of the DNA was measured with Picogreen
according to the manufacturer’s instructions (Invitrogen).
Array CGH analysis
The 32K bacterial artificial chromosome (BAC) re-array
collection (CHORI) tiling path aCGH platform used for this study
was constructed in the Breakthrough Breast Cancer Research
Centre [55]. DNA labelling, array hybridisations, image acquisition
and filtering were performed as described in Natrajan et al. [56].
Data were smoothed using the circular binary segmentation (cbs)
algorithm [27]. A categorical analysis was applied to the BACs after
classifying them as representing gain, loss or no-change according to
their smoothed Log2 ratio values as defined [56].
Cell culture
MCF7 cells were routinely maintained in phenol red free
RPMI1640 (Invitrogen, Paisley, UK) supplemented with 10%
foetal bovine serum and oestradiol (1 nM). Cells were passaged
weekly and medium replenished every 48–72 hours. In the case of
BT474, cell monolayers were cultured in phenol red containing
medium supplemented with 10% foetal bovine serum. Cell lines
were shown to be free of mycoplasma by routine testing.
Figure 4. Association between C6orf expression, proliferation, and outcome in tumours. a. Relationship between C6ORF211 expression
and expression of proliferation metagene in 104 breast cancers. b. Relationship between C6ORF211 expression and expression of proliferation
metagene in 354 breast cancers from the Loi dataset. c. Relationship between C6ORF97 expression and expression of proliferation metagene in the
Loi dataset. d. Kaplan–Meier curve representing the fraction relapse-free survival comparing the lowest quartile of C6ORF97 expression with the
highest in the Loi dataset.
doi:10.1371/journal.pgen.1001382.g004
ESR1 Is Co-Expressed with Closely Adjacent Genes
PLoS Genetics | www.plosgenetics.org 8 April 2011 | Volume 7 | Issue 4 | e1001382
Real-time quantitative PCR
Total RNA from treated MCF7 and BT-474 cells was extracted
using the RNeasy Mini Kit (Qiagen) according to the manufactur-
er’s instructions. All RNA quantification was performed using the
Agilent 2100 Bioanalyzer with RNA Nano LabChip Kits (Agilent
Technologies, Wokingham, Berkshire, UK). RNA was reverse
transcribed using SuperScript III (Invitrogen), and random primers.
Twenty nanograms of resulting cDNA of each sample was analyzed
in triplicates by qRT-PCR using the ABI Perkin-Elmer Prism
7900HT Sequence detection system (Applied Biosystems). Taqman
gene expression assays (Applied Biosystems) were used to quantitate
processed transcripts of ESR1 (Hs01046818_m1), C6ORF96
(Hs00215537_m1), C6ORF97 (Hs01563344_m1), C6ORF211
(Hs00226188_m1), which were normalized to two housekeeping
genes, FKBP15 (Hs00391480_m1) and TBP (Hs00427620_m1).
These housekeepers were selected from a previously published list of
appropriate reference genes for breast cancer [57]. Custom assays
using primers designed to span intron-exon boundaries were used to
measure nascent RNA (Table S3). Gene expression was quantified
using a standard curve generated from serial dilutions of reference
cDNA from a pooled breast cancer cell line RNA.
Immunoblots
Cell monolayers were washed with cold PBS twice and collected by
scraping. Cell pellets were lysed in extraction buffer, resolved by SDS-
PAGE and transferred to nitrocellulose membranes as described
previously [30]. Membranes were blocked and probed with a
polyclonal antibody directed against the predicted peptide (amino
acids 368–382) of C6orf211 (Eurogentec, Southampton, UK) and anti
b-actin (Sigma-Aldrich, Poole, UK) using the methods described
previously [58]. Quantification of immunoblots was performed using
the NIH ImageJ software, and immunoblots were normalized to actin.
Immunofluorescence and confocal studies
Cells were grown on glass coverslips in standard growth
medium. Cells were fixed and incubated in the presence of
primary antibodies as described previously [58]. Coverslips were
washed with PBS and cells were incubated in the presence of
appropriate Alexa Fluor 555 (red) or Alexa Fluor 488 (green)-
labeled secondary antibodies (Molecular Probes, Invitrogen,
Paisley, UK) diluted 1:1000 for 1 hr. Cells were washed in PBS
and nuclei (DNA) were counterstained with 4,6-diamidino-2-
phenylindole (DAPI; Invitrogen) diluted 1:10000. Coverslips were
mounted onto glass slides using Vectashield mounting medium
(Vector Laboratories, Peterborough, UK). Images were collected
sequentially in three channels on a Zeiss LSM710 (Carl Zeiss Ltd,
Welwyn Garden City, UK) laser scanning confocal microscope at
the same magnification (663 oil immersion objective).
Cell proliferation assays
Cell lines were depleted of steroids for 3 days by culturing in DCC-
medium [59], seeded into 12-well plates at a density of 1610
4
cells/
well for MCF7 and 4610
4
cells per well for BT474, monolayers were
allowed to acclimatize for 24 h before treatment with drug
combinations indicated for 6 d with daily changes. Cell number
was determined using a Z1 Coulter Counter (Beckman Coulter).
Results were confirmed in a minimum of three independent
experiments, and each experiment was performed in triplicate.
Effect of oestradiol and ICI182780 on ORF RNA
expression
Wt-MCF7 cells were stripped of steroid for 3 days as described
above. Cells were subsequently seeded into 12 well plates at a
density of 1610
5
cells/well. After 24 hours monolayers were treated
with vehicle (0.01% v/v ethanol), oestradiol (1 nM) or ICI182780
(10 nM) for the time intervals indicated. RNA was extracted using
RNeasy Mini kit (Qiagen) and subjected to qRT-PCR as described.
SiRNA knockdown of ORFs
Wt-MCF7 cells were stripped of steroid for 24 hours in DCC-
medium. Stripped cells were subsequently seeded into 12 well
plates at a density of 2610
4
cells/well for proliferation assays or
1610
5
cells/well for RNA expression analysis. After 24 hours
monolayers were transfected with 100 nM of either siRNA against
C6ORF96,C6ORF97,C6ORF211 or control siRNA using
DharmaFECT 1 reagent (Dharmacon, Thermo Fisher Scientific,
UK). Medium was then replenished the following day and cells
were allowed to acclimatise for a further 24 hours. After 24 hours
samples were taken for RNA expression analysis. For analysis of
oestrogen-dependent proliferation, the monolayers were treated
with increasing concentrations of oestradiol (0.01, 0.1 or 1 nM)
48 hours post transfection. The remaining plates were treated
daily with the treatments indicated for 6 days before carrying out
cell counts as described above.
Supporting Information
Figure S1 Correlation between ESR1 and the mean of
C6ORF96,C6ORF97, and C6ORF211 showing tumours with
measured copy number variations shown in colour.
Found at: doi:10.1371/journal.pgen.1001382.s001 (0.18 MB
DOC)
Figure S2 Validation of C6ORF gene silencing by siRNA. MCF7
cells were grown in either media containing stripped serum or
stripped serum plus 1 nM oestradiol and transfected with siRNA.
After 48 h, RNA was extracted from cells and complementary DNA
synthesized using standard methods. Using Assay-on-Demand
primer/probe sets (Applied Biosystems, UK), we performed real-
time quantitative PCR. Gene expression was calculated relative to
expression of TBP and FKBP15 and adjusted relative to expression
in cells transfected with a non-targeting siRNA (siControl). Error
bars represent the standard error of the mean (SEM). MCF7 cells
were transfected with siRNA against C6ORF96,C6ORF97,
C6ORF211 or control siRNA in A. DCC or B. 1 nM oestradiol.
Found at: doi:10.1371/journal.pgen.1001382.s002 (0.78 MB
DOC)
Figure S3 Validation of C6ORF211 gene silencing in deconvolu-
tion of siRNA SMARTPOOL. MCF7 cells were grown in media
containing stripped serum and transfected with individual siRNAs.
After 48 h, RNA was extracted from cells and complementary DNA
synthesized using standard methods. Using Assay-on-Demand
primer/probe sets (Applied Biosystems, UK), we performed real-
time quantitative PCR. Gene expression was calculated relative to
expression of TBP and FKBP15 and adjusted relative to expression
in cells transfected with a non-targeting siRNA (siRNA Control).
Error bars represent the standard error of the mean (SEM).
Found at: doi:10.1371/journal.pgen.1001382.s003 (0.84 MB
DOC)
Figure S4 Validation of C6ORF protein knockdown by siRNA.
MCF7 cells were transfected with siRNA against C6ORF97,
C6ORF211 or control siRNA. 72 h after siRNA transfection, cell
lysates were generated and immunoblotted using a. a polyclonal
antibody generated against C6orf211 and b. anti-b-actin as a
loading control.
Found at: doi:10.1371/journal.pgen.1001382.s004 (0.06 MB
DOC)
ESR1 Is Co-Expressed with Closely Adjacent Genes
PLoS Genetics | www.plosgenetics.org 9 April 2011 | Volume 7 | Issue 4 | e1001382
Figure S5 Validation of proliferation changes induced by
individual siRNAs. WT-MCF7 cells were stripped of steroid for
24 hours in DCC-medium. Stripped cells were seeded into 12 well
plates at a density of 20,000 cells/well for proliferation assays or
100,000 cells/well for RNA expression analysis. After 24 hours
monolayers were transfected with 100 nM of single siRNAs against
C6ORF211 or control siRNA (SMARTPool). Medium was
replenished the following day and cells were allowed to acclimatise
for a further 24 hours. Monolayers were subsequently treated with
fresh DCC medium. The remaining plates were treated with DCC
medium for 6 days. Proliferation in response to individual siRNA
knockdown were established by change in cell number using a
coulter counter (Beckman Scientific UK). Data presented is
expressed as absolute cell number or fold change over siControl
(SMARTpool). All data is from triplicate wells, each well read twice.
Found at: doi:10.1371/journal.pgen.1001382.s005 (0.27 MB
DOC)
Figure S6 a. Kaplan-Meier curve comparing proportion relapse-
free survival in the lowest quartile of C6ORF97 expression versus the
highest in 142 untreated ER+ve tumours from the Wang dataset. b.
Kaplan-Meier curve comparing the proportion relapse-free survival
in the lowest quartile of C6ORF211 expression versus the highest in
345 tamoxifen-treated ER+ve tumours from the Loi dataset. c.
Kaplan-Meier curve comparing the proportion relapse-free survival
in the lowest quartile of C6ORF211 expression versus the highest in
142 untreated ER+ve tumours from the Wang dataset.
Found at: doi:10.1371/journal.pgen.1001382.s006 (0.69 MB
DOC)
Figure S7 Confocal analysis of C6orf211 localisation. To
determine the subcellular localization of C6orf211 protein,
confocal analysis was carried out using a polyclonal antibody
directed against the predicted peptide (amino acids 368–381).
MCF-7 cells were plated onto coverslips and stained. a. Nuclei
were visualized using DAPI and stained with antibodies against
C6ORF211 (b) and oestrogen receptor (c). An overlay of all three
images is shown in (d).
Found at: doi:10.1371/journal.pgen.1001382.s007 (0.07 MB
DOC)
Table S1 Correlation of expression of genes in the region of
amplification surrounding ESR1 as defined by Reis-Filho et al.
(2008) [26] with expression of ESR1 in baseline biopsies from 104
patients with ER+ve breast cancer.
Found at: doi:10.1371/journal.pgen.1001382.s008 (0.06 MB
DOC)
Table S2 Correlation expression of the C6ORFs and ESR1 with
expression of well-known proliferation genes. Correlations signif-
icant at p,0.05 are indicated with an asterisk.
Found at: doi:10.1371/journal.pgen.1001382.s009 (0.04 MB
DOC)
Table S3 Custom assays designed to measure nascent RNA.
Found at: doi:10.1371/journal.pgen.1001382.s010 (0.04 MB
DOC)
Author Contributions
Conceived and designed the experiments: AKD HA LAM MD. Performed
the experiments: AKD HA ELK SP RR SD KS AL LAM. Analyzed the
data: AKD HA ZG ELK SP SD LAM. Contributed reagents/materials/
analysis tools: AL. Wrote the paper: AKD MD.
References
1. Parkin DM, Bray F, Ferlay J, Pisani P (2005) Global cancer statistics, 2002. CA
Cancer J Clin 55: 74–108.
2. Dowsett M, Dunbier AK (2008) Emerging biomarkers and new understanding
of traditional markers in personalized therapy for breast cancer. Clin Cancer
Res 14: 8019–8026.
3. Perou CM, Sorlie T, Eisen MB, van de Rijn M, Jeffrey SS, et al. (2000)
Molecular portraits of human breast tumours. Nature 406: 747–752.
4. Hammes SR, Levin ER (2007) Extranuclear steroid recept ors: nature and
actions. Endocr Rev 28: 726–741.
5. Sorlie T, Tibshirani R, Parker J, Hastie T, Marron JS, et al. (2003) Repeated
observation of breast tumor subtypes in independent gene expression data sets.
Proc Natl Acad Sci U S A 100: 8418–8423.
6. Zheng W, Long J, Gao YT, Li C, Zheng Y, et al. (2009) Genome-wide
association study identifies a new breast cancer susceptibility locus at 6q25.1. Nat
Genet 41: 324–328.
7. Turnbull C, Ahmed S, Morrison J, Pernet D, Renwick A, et al. (2010) Genome-
wide association study identifies five new breast cancer susceptibility loci. Nat
Genet 42: 504–7.
8. Stacey SN, Sulem P, Zanon C, Gudjonsson SA, Thorleif sson G, et al. (2010)
Ancestry-Shift Refinement Mapping of the C6orf97-ESR1 Breast Cancer
Susceptibility Locus. PLoS Genet 6: e1001029. doi: 10.1371/journal.
pgen.1001029.
9. Fletcher O, Johnson N, Orr N, Hosking FJ, Gibson LJ, et al. (2011) Novel Breast
Cancer Susceptibility Locus at 9q31.2: Results of a Genome-Wide Association
Study. J Natl Cancer Inst 103: 425–35.
10. Cai Q, Wen W, Qu S, Li G, Egan KM, et al. (2011) Replication and Functional
Genomic Analyses of t he Breast Cancer Su sceptibility Locus at 6q 25.1
Generalize Its Importance in Women of Chinese, Japanese, and European
Ancestry. Cancer Res 71: 1344–1355.
11. Frasor J, Danes JM, Komm B, Chang KC, Lyttle CR, et al. (2003) Profiling of
estrogen up- and down-regulated gene expression in human breast cancer cells:
insights into gene networks and pathways underlying estrogenic control of
proliferation and cell phenotype. Endocrinology 144: 4562–4574.
12. Oh DS, Troester MA, Usary J, Hu Z, He X, et al. (2006) Estrogen-regu lated
genes predict survival in hormone receptor-positive breast cancers. J Clin Oncol
24: 1656–1664.
13. Yu J, Yu J, Cordero KE, Johnson MD, Ghosh D, et al. (2008) A transcriptional
fingerprint of estrogen in human breast cancer predicts patient survival.
Neoplasia 10: 79–88.
14. Miller WR, Larionov AA, Renshaw L, Anderson TJ, White S, et al. (2007)
Changes in breast cancer transcriptional profiles after treatment with the
aromatase inhibitor, letrozole. Pharmacogenet Genomics 17: 813–826.
15. Mackay A, Urruticoechea A, Dixon JM, Dexter T, Fenwick K, et al. (2007)
Molecular response to aromatase inhibitor treatment in primary breast cancer.
Breast Cancer Res 9: R37.
16. Mello-Grand M, Singh V, Ghimenti C, Scatolini M, Regolo L, et al. (2010)
Gene expression profiling and prediction of response to hormonal neoadjuvant
treatment with anastrozole in surgically resectable breast cancer. Breast Cancer
Res Treat.
17. Carroll JS, Meyer CA, Song J, Li W, Geistlinger TR, et al. (2006) Gen ome-wide
analysis of estrogen receptor binding sites. Nat Genet 38: 1289–1297.
18. Fullwood MJ, Liu MH, Pan YF, Liu J, Xu H, et al. (2009) An oestrogen-
receptor-alpha-bound human chromatin interactome. Nature 462: 58–64.
19. Rae JM, Johnson MD, Scheys JO, Cordero KE, Larios JM, et al. (2005) GREB
1 is a critical regulator of hormone dependent breast cancer growth. Breast
Cancer Res Treat 92: 141–149.
20. Shang Y, Hu X, DiRenzo J, Lazar MA, Brown M (2000) Cofactor dynamics and
sufficiency in estrogen receptor-regulated transcription. Cell 103: 843–852.
21. Musgrove EA, Sutherland RL (2009) Biological determinants of endocrine
resistance in breast cancer. Nat Rev Cancer 9: 631–643.
22. Ali S, Coombes RC (2002) Endocrine-responsive breast cancer and strategies for
combating resistance. Nat Rev Cancer 2: 101–112.
23. Smith IE, Walsh G, Skene A, Llombart A, Mayordomo JI, et al. (2007) A phase
II placebo-controlled trial of neoadjuvant anastrozole alone or with gefitinib in
early breast cancer. J Clin Oncol 25: 3816–3822.
24. Wang Y, Klijn JG, Zhang Y, Sieuwerts AM, Look MP, et al. (2005) Gene-
expression profiles to predict distant metastasis of lymph-node-negative primary
breast cancer. Lancet 365: 671–679.
25. Holst F, Stahl PR, Ruiz C, Hellwinkel O, Jehan Z, et al. (2007) Estrogen
receptor alpha (ESR1) gene amplification is frequent in breast cancer. Nat Genet
39: 655–660.
26. Reis-Filho JS, Drury S, Lambros MB, Marchio C, Johnson N, et al. (2008) ESR1
gene amplification in breast cancer: a common phenomenon? Nat Genet 40:
809–810; author reply 810-802.
27. Mackay A, Tamber N, Fenwick K, Iravani M, Grigoriadis A, et al. (2009) A
high-resolution integrated analysis of genetic and expression profiles of breast
cancer cell lines. Breast Cancer Res Treat 118: 481–498.
28. Xia W, Bacus S, Hegde P, Husain I, Strum J, et al. (2006) A model of acquired
autoresistance to a potent ErbB2 tyrosine kinase inhibitor and a therapeutic
ESR1 Is Co-Expressed with Closely Adjacent Genes
PLoS Genetics | www.plosgenetics.org 10 April 2011 | Volume 7 | Issue 4 | e1001382
strategy to prevent its onset in breast cancer. Proc Natl Acad Sci U S A 103:
7795–7800.
29. Leary AF, Martin L, Thornhill A, Dowsett M, Johnston S (2010) Combining or
Sequencing Targeted Therapies in ER+/HER2 Amplified Breast Cancer (BC):
In Vitro and In Vivo Studies of Letrozole and Lapatinib in an ER+/HER2+
Aromatase-Transfected BC Model. Cancer Res 70: 292s.
30. Martin LA, Farmer I, Johnston SR, Ali S, Marshall C, et al. (2003) Enhanced
estrogen receptor (ER) alpha, ERBB2, and MAPK signal transduction pathways
operate during the adaptation of MCF-7 cells to long term estrogen deprivation.
J Biol Chem 278: 30458–30468.
31. Wakeling AE, Bowler J (1992) ICI 182,780, a new antioestrogen with clinical
potential. J Steroid Biochem Mol Biol 43: 173–177.
32. McClelland RA, Gee JM, Francis AB, Robertson JF, Blamey RW, et al. (1996)
Short-term effects of pure anti-oestrogen ICI 182780 treatment on oestrogen
receptor, epidermal growth factor receptor and transforming growth factor-
alpha protein expression in human breast cancer. Eur J Cancer 32A: 413–416.
33. Li GJ, Zhao Q, Zheng W (2005) Alteration at translational but not
transcriptional level of transferrin receptor expression following manganese
exposure at the blood-CSF barrier in vitro. Toxicol Appl Pharmacol 205:
188–200.
34. Ghazoui Z, Buffa FM, Dunbier AK, Anderson H, Dexter T, et al. (2011) Close
and stable relationship between proliferation and a hypoxia metagene in
aromatase inhibitor treated ER-positive breast cancer. Clin Cancer Res.
35. Loi S, Haibe-Kains B, Desmedt C, Wirapati P, Lallemand F, et al. (2008)
Predicting prognosis using molecular profiling in estrogen receptor-positive
breast cancer treated with tamoxifen. BMC Genomics 9: 239.
36. Loi S, Haibe-Kains B, Desmedt C, Lallemand F, Tutt AM, et al. (2007)
Definition of clinically distinct molecular subtypes in estrogen receptor-positive
breast carcinomas through genomic grade. J Clin Oncol 25: 1239–1246.
37. Drury S, Johnson N, Hills M, Salter J, Dunbier A, et al. (2010) A breast cancer-
associated SNP adjacent to ESR1 correlates with oestrogen receptor-alpha (ER
alpha) level in invasive breast tumours. Cancer Res 69: 4138.
38. Dunbier AK, Anderson H, Ghazoui Z, Pancholi S, Sidhu K, et al. (2010) ESR1
is co-expressed with closely adjacent novel genes in estrogen receptor positive
breast cancer. Presented at the 101st American Association for Cancer Research
Annual Meeting, Washington DC, USA Abstract number 2916.
39. Lazennec G, Huignard H, Valotaire Y, Kern L (1995) Characterization of the
transcription start point of the trout estrogen receptor-encoding gene: evidence
for alternative splicing in the 59untranslated region. Gene 166: 243–247.
40. Martin MB, Saceda M, Garcia-Morales P, Gottardis MM (1994) Regulation of
estrogen receptor expression. Breast Cancer Res Treat 31: 183–189.
41. Martin MB, Angeloni SV, Garcia-Morales P, Sholler PF, Castro-Galache MD,
et al. (2004) Regulation of estrogen receptor-alpha expression in MCF-7 cells by
taxol. J Endocrinol 180: 487–496.
42. Angeloni SV, Martin MB, Garcia-Morales P, Castro-Galache MD, Ferragut JA,
et al. (2004) Regulation of estrogen receptor-alpha expression by the tumor
suppressor gene p53 in MCF-7 cells. J Endocrinol 180: 497–504.
43. Lieberman-Aiden E, van Berkum NL, Williams L, Imakaev M, Ragoczy T, et al.
(2009) Comprehensive mapping of long-range interactions reveals folding
principles of the human genome. Science 326: 289–293.
44. http://www.genecards.org/.
45. http://www.ncbi.nlm.nih.gov/homologene.
46. Ewing RM, Chu P, Elisma F, Li H, Taylor P, et al. (2007) Large-scale mapping
of human protein-protein interactions by mass spectrometry. Mol Syst Biol 3: 89.
47. Early Breast Cancer Trialists’ Collaborative Group (1998) Tamoxifen for early
breast cancer: an overview of the randomised trials. Lancet 351: 1451–1467.
48. The World Medical Association: Declaration of Helsinki. http://www .wma.net/
e/policy/b3htm.
49. Du P, Kibbe WA, Lin SM (2008) lumi: a pipeline for processing Illumina
microarray. Bioinformatics 24: 1547–1548.
50. Korn EL, Troendle JF, McShane LM, Simon R (2004) Controlling the number
of false discoveries: application to high-dimensional genomic data. Journal of
Statistical Planning and Inference 124: 379–398.
51. Miller LD, Smeds J, George J, Vega VB, Vergara L, et al. (2005) An expression
signature for p53 status in human breast cancer predicts mutation status,
transcriptional effects, and patient survival. Proc Natl Acad Sci U S A 102:
13550–13555.
52. Desmedt C, Piette F, Loi S, Wang Y, Lallemand F, et al. (2007) Stro ng time
dependence of the 76-gene prognostic signature for node-negative breast cancer
patients in the TRANSBIG multicenter independent validation series. Clin
Cancer Res 13: 3207–3214.
53. Huang F, Reeves K, Han X, Fairchild C, Platero S, et al. (2007) Identification of
candidate molecular markers predicting sensitivity in solid tumors to dasatinib:
rationale for patient selection. Cancer Res 67: 2226–2238.
54. Ghazoui Z, Buffa FM, Dunbier AK, Anderson H, Dexter T, et al. (2009)
Aromatase Inhibitors Reduce the Expression of a Hypoxia Metagene in
Oestrogen Receptor Positive Breast Cancer in Postmenopausal Women. Cancer
Res 69: 408.
55. Marchio C, Iravani M, Natrajan R, Lambros MB, Savag e K, et al. (2008)
Genomic and immunophenotypical characterization of pure micropapillary
carcinomas of the breast. J Pathol 215: 398–410.
56. Natrajan R, Weigelt B, Mackay A, Geyer FC, Grigoriadis A, et al. (2010) An
integrative genomic and transcriptomic analysis reveals molecular pathways and
networks regulated by copy number aberrations in basal-like, HER2 and luminal
cancers. Breast Cancer Res Treat 121: 575–589.
57. Drury S, Anderson H, Dowsett M (2009) Selection of REFERENCE genes for
normalization of qRT-PCR data derived from FFPE breast tumors. Diagn Mol
Pathol 18: 103–107.
58. Pancholi S, Lykkesfeldt AE, Hilmi C, Banerjee S, Leary A, et al. (2008) ERBB2
influences the subcellular localization of the estrogen receptor in tamoxifen-
resistant MCF-7 cells leading to the activation of AKT and RPS6KA2. Endocr
Relat Cancer 15: 985–1002.
59. Darbre P, Yates J, Curtis S, King RJ (1983) Effect of estradiol on human breast
cancer cells in culture. Cancer Res 43: 349–354.
ESR1 Is Co-Expressed with Closely Adjacent Genes
PLoS Genetics | www.plosgenetics.org 11 April 2011 | Volume 7 | Issue 4 | e1001382
... [GRCh38/hg38]). CCDC170 was found to be coexpressed with ESR1 in breast cancer tissues [6], and an ESR1-CCDC170 rearrangement was discovered in luminal B breast tumors [7]. ERα, the protein encoded by ESR1, is well known to be an independent prognostic factor in breast cancer, and thus is a target of endocrine therapy [8]. ...
... Several single-nucleotide polymorphisms around human CCDC170 have been identified as important breast cancer risk indicators in Chinese women [25][26][27]. In addition, CCDC170 was found to be tightly coexpressed with ESR1 in breast tumor biopsies and cells [6]. A previous study demonstrated that CCDC170 was fused to ESR1 and employed the constitutively active ESR1 promoter to induce the expression of a truncated form of CCDC170. ...
... However, it has been proposed that CCDC170 can function as either an oncogene or a tumor suppressor [28]. Indeed, higher CCDC170 expression has been associated with a better prognosis in certain breast cancer subtypes, but with a poorer prognosis in others [6,29]. Though these data have highlighted the importance of the CCDC170 gene and its fusion protein in breast cancer, the pathobiology and clinical relevance of CCDC170 have remained unclear. ...
Article
Full-text available
Genome-wide association studies have revealed that multiple single-nucleotide polymorphisms in the intergenic region between estrogen receptor 1 and coiled-coil domain containing 170 (CCDC170) are associated with breast cancer risk. We performed microarray and bioinformatics analyses to identify genes that were induced upon CCDC170 overexpression, and confirmed our findings by evaluating paraffin-embedded breast cancer tissues and conducting cellular assays. In CCDC170-overexpressing MCF7 breast cancer cells, microarray analyses revealed that inositol-requiring enzyme 1 (IRE1) was the most elevated gene in enriched pathways. In breast cancer tissues, IRE1 expression correlated positively with CCDC170 and X-box binding protein 1 expression at both the mRNA and protein levels. In a survival analysis, patients with higher CCDC170 levels exhibited better disease-free survival. Western blotting indicated that overexpressing CCDC170 in MCF7 cells increased protein levels of IRE1α, estrogen receptor α and X-box binding protein 1, while silencing CCDC170 reduced them. CCDC170 overexpression promoted apoptosis in MCF7 cells, and this effect was more obvious under endoplasmic reticulum stress. MCF7 cells overexpressing CCDC170 were more sensitive to paclitaxel. Our study showed that higher CCDC170 expression is associated with a better prognosis in breast cancer patients and that CCDC170 may promote apoptosis through the IRE1α pathway.
... Neighboring genes in close proximity in humans can be expressed concurrently (Hurst et al., 2002;Takai and Jones, 2004;Trinklein et al., 2004). There is also evidence that RMND1, CCDC170 and ARMT1 expression, immediately upstream of the ESR1 are correlated with ESR1 expression in tumor biopsies taken from postmenopausal women with stage I to IIIB ERþ breast cancer (Dunbier et al., 2011). Therefore, genes in the ESR1 region may be coregulated and not just menstrual cycle-dependent. ...
... Variants located in regulatory elements might activate promoters affecting more than one gene. Variants associated with breast cancer have been reported to regulate ESR1 in reporter assays and may regulate other genes RMND1, CCDC170 and ARMT1 supporting coregulation of genes in this region (Dunbier et al., 2011;Dunning et al., 2016). Other approaches will be required to understand how independent genetic risk factors in the region of ESR1 increase risk for multiple diseases and traits. ...
Article
Full-text available
The aetiology and pathogenesis of endometriosis are complex with both genetic and environmental factors contributing to disease risk. Genome-wide association studies (GWAS) have identified multiple signals in the estrogen receptor 1 (ESR1) region associated with endometriosis and other reproductive traits and diseases. In addition, candidate gene association studies identified signals in the ESR1 region associated with endometriosis risk suggesting genetic regulation of genes in this region may be important for reproductive health. This study aimed to investigate hormonal and genetic regulation of genes in the ESR1 region in human endometrium. Changes in serum oestradiol and progesterone concentrations and expression of hormone receptors ESR1 and progesterone receptor (PGR) were assessed in endometrial samples from 135 women collected at various stages of the menstrual cycle. Correlation between hormone concentrations, receptor expression and expression of genes in the ESR1 locus was investigated. The effect of endometriosis risk variants on expression of genes in the region was analysed to identify gene targets. Hormone concentrations and receptor expression varied significantly across the menstrual cycle. Expression of genes in the ESR1 region correlated with progesterone concentration, however, they were more strongly correlated with expression of ESR1 and PGR suggesting co-regulation of genes. There was no evidence that endometriosis risk variants directly regulated expression of genes in the region. Limited sample size and cellular heterogeneity in endometrial tissue may impact the ability to detect significant genetic effects on gene expression. Effects of these variants should be validated in a larger dataset and in relevant individual cell types.
... However, techniques that assess long-range chromatin interactions show how transcription factors and distal regulatory elements act in concert to regulate gene expression (techniques reviewed in [70]). Evidence of long-range chromatin interactions involving ESR1 was described by Dunbier et al. in which ESR1 appeared to be co-regulated with neighboring genes CCDC170, ARMT1, and SYNE1 in ER-positive breast cancer patient samples [71]. Dunning et al. also utilized genome-wide association studies and found enhancer elements between CCDC170 and ESR1 that co-regulated ESR1, RMND1, and CCDC170 [72]. ...
... As described earlier, Bailey et al. also observed co-repression of RMND1, ARMT, CCDC170, and ESR1 following CRISPR knockout of a genomic region that is commonly mutated in breast cancer patients [60]. CCDC170 (Coiled-Coil Domain-Containing Protein 170) and ARMT1 (Acidic Residue Methyltransferase 1) have been implicated in breast cancers by causing Golgi reorganization and promoting cell proliferation, respectively [71,73,74]. The functions of RMND1 (Required for Meiotic Nuclear Division 1) and SYNE1 (Spectrin Repeat Containing Nuclear Envelope Protein) in breast cancer are unknown, but RMND1 has been reported to affect mitochondria translation in human subjects [75]. ...
Article
Transcriptional regulation of ESR1, the gene that encodes for estrogen receptor α (ER), is critical for regulating the downstream effects of the estrogen signaling pathway in breast cancer such as cell growth. ESR1 is a large and complex gene that is regulated by multiple regulatory elements, which has complicated our understanding of how ESR1 expression is controlled in the context of breast cancer. Early studies characterized the genomic structure of ESR1 with subsequent studies focused on identifying intrinsic (chromatin environment, transcription factors, signaling pathways) and extrinsic (tumor microenvironment, secreted factors) mechanisms that impact ESR1 gene expression. Currently, the introduction of genomic sequencing platforms and additional genome-wide technologies has provided additional insight on how chromatin structures may coordinate with these intrinsic and extrinsic mechanisms to regulate ESR1 expression. Understanding these interactions will allow us to have a clearer understanding of how ESR1 expression is regulated and eventually provide clues on how to influence its regulation with potential treatments. In this review, we highlight key studies concerning the genomic structure of ESR1, mechanisms that affect the dynamics of ESR1 expression, and considerations towards affecting ESR1 expression and hormone responsiveness in breast cancer.
... Several genes are located within a 1 Mb region of the 6q25.1 locus, including ZBTB2, RMND1 [C6orf96], C6orf97, C6orf211, and AKAP12 [5]. Recently, genome-wide studies have identified relationships in the expression patterns between genes and the SNPs in this region, which is upstream of the gene encoding ER (ESR1), which is associated with breast cancer susceptibility [20][21][22][23][24]. Dunbier and colleagues have previously suggested that some of the biological effects caused by ESR1 could be mediated and modified by these co-expressed genes [21]. ...
... Several genes are located within a 1 Mb region of the 6q25.1 locus, including ZBTB2, RMND1 [C6orf96], C6orf97, C6orf211, and AKAP12 [5]. Recently, genome-wide studies have identified relationships in the expression patterns between genes and the SNPs in this region, which is upstream of the gene encoding ER (ESR1), which is associated with breast cancer susceptibility [20][21][22][23][24]. Dunbier and colleagues have previously suggested that some of the biological effects caused by ESR1 could be mediated and modified by these co-expressed genes [21]. ...
Article
Full-text available
Chronic myeloid leukemia (CML) is a clonal myeloproliferative disorder. Our previous study reported novel loci as genetic markers associated with increased susceptibility to CML. The present study conducted an expression quantitative trait loci (eQTL) analysis to confirm that the single nucleotide polymorphisms (SNPs) at these loci affect the expression of candidate CML-susceptible genes. We identified that three SNPs (rs963193, rs6931104, and rs9371517) were related to the gene expression pattern of RMND1 (Required For Meiotic Nuclear Division 1 Homolog) in both granulocytes and mononuclear cells from 83 healthy donors. Furthermore, reduced expression of RMND1 expression was noted in CML patients compared with that in healthy individuals. We used the eQTL browsing tool to assess the regulatory information on the three associated significant SNPs, out of which rs6931104 showed strong evidence of regulatory effects. Chromatin immunoprecipitation (ChIP) assays demonstrated that A alleles of rs6931104 could significantly change the binding affinity of transcription factor (TF) RFX3 compared to the G alleles. Then, we performed in vitro experiments on BCR-ABL1-positive (BCR-ABL1+) cell lines. We found that expression of the CML-susceptible gene RMND1 is affected by the binding affinity of TF RFX3, suggesting that RFX3 plays a role in RMND1 expression. Our findings suggest potential target genes for associations of genetic susceptibility risk loci and provide further insights into the pathogenesis and mechanism of CML.
... Because R-loop accumulation also contributes to genome instability (25), BRCA1 mutation-carrying luminal progenitor cells with accumulated R-loops and compromised ability of maintaining genome integrity (28,(64)(65)(66) could be the prime target for further oncogenic events that ultimately lead to basallike BRCA1-associated breast cancer. ESR1 and its two neighboring genes affected by BRCA1 deficiency, CCDC170 and RMND1, have been implicated in sporadic breast cancer (67)(68)(69)(70)(71). For example, CCDC170 protein is involved in Golgi-associated microtubules dynamics. ...
... Its truncation in breast cancer has been suggested to alter cell polarity/motility and drive tumor initiation/progression (70). In addition, co-expression and co-regulation of ESR1, CCDC170 and RMND1 have been reported in breast cancer specimens (69), and their allele- specific expression could account for the association between variants at the ESR1 locus and ER␣ negative breast cancer (68,71). Furthermore, tumor-specific ESR1-CCDC170 rearrangements have been reported in breast cancer (67,72,73), accentuating the clinical relevance of this genomic region to tumorigenesis. ...
Article
Full-text available
BRCA1-associated basal-like breast cancer originates from luminal progenitor cells. Breast epithelial cells from cancer-free BRCA1 mutation carriers are defective in luminal differentiation. However, how BRCA1 deficiency leads to lineage-specific differentiation defect is not clear. BRCA1 is implicated in resolving R-loops, DNA-RNA hybrid structures associated with genome instability and transcriptional regulation. We recently showed that R-loops are preferentially accumulated in breast luminal epithelial cells of BRCA1 mutation carriers. Here, we interrogate the impact of a BRCA1 mutation-associated R-loop located in a putative transcriptional enhancer upstream of the ERα-encoding ESR1 gene. Genetic ablation confirms the relevance of this R-loop-containing region to enhancer-promoter interactions and transcriptional activation of the corresponding neighboring genes, including ESR1, CCDC170 and RMND1. BRCA1 knockdown in ERα+ luminal breast cancer cells increases intensity of this R-loop and reduces transcription of its neighboring genes. The deleterious effect of BRCA1 depletion on transcription is mitigated by ectopic expression of R-loop-removing RNase H1. Furthermore, RNase H1 overexpression in primary breast cells from BRCA1 mutation carriers results in a shift from luminal progenitor cells to mature luminal cells. Our findings suggest that BRCA1-dependent R-loop mitigation contributes to luminal cell-specific transcription and differentiation, which could in turn suppress BRCA1-associated tumorigenesis.
... In humans, CCDC170 is coexpressed with its neighboring gene ESR1 (22). We hypothesized that the physical genetic linkage could cause higher expression in females than in males and therefore explain why the polymorphism is limited to females. ...
Article
Full-text available
Animal coloration is often expressed in periodic patterns that can arise from differential cell migration, yet how these processes are regulated remains elusive. We show that a female-limited polymorphism in dorsal patterning (diamond/chevron) in the brown anole is controlled by a single Mendelian locus. This locus contains the gene CCDC170 that is adjacent to, and coexpressed with, the Estrogen receptor-1 gene, explaining why the polymorphism is female limited. CCDC170 is an organizer of the Golgi-microtubule network underlying a cell's ability to migrate, and the two segregating alleles encode structurally different proteins. Our agent-based modeling of skin development demonstrates that, in principle, a change in cell migratory behaviors is sufficient to switch between the two morphs. These results suggest that CCDC170 might have been co-opted as a switch between color patterning morphs, likely by modulating cell migratory behaviors.
... We validated our predictions by the following several independent datasets. Specifically, we observed that (i)four clusters have different functions, i.e., cluster 6 cells exhibit a variety of biological functions such as activation of Stat3 protein to modulate cancer stemness 30 ; cluster 10 cells are related to cell adhesion and cell division; cluster 3 cells participate in cell growth and proliferation; and cluster 5 cells mediate cell migration and apoptotic process (Supplementary Fig.8); (ii) by re-analyzing 24,489 epithelial cells from 20 breast cancer patients, the higher expression of ARMT1, SCUBE2, and CCNO is in scRNA-seq data from ER-positive patients, compared with those from ER-negative patients(Figs.4f and 5a), which is consistent with the previous conclusion: ER and ARMT1 are co-expressed, and ARMT1 affects cell proliferation31 , as well as clusters 10 and 3 cells existing in breast cancer; (iii) there is a trajectory between three clusters identified from 2,352 epithelial cells of CID4067 (a representative luminal B patient) through analyzing its scRNA-seq by Monocle 2 32 , and with the estimated pseudo-time, the expression of PDZK1IP1 for cluster 6 decreases, ARMT1 and SCUBE2 for clusters 10 and 3 increase, supporting our trajectory inference between clusters 6, 10, and 3(Fig.5c); and (iv) the expression of CCNO (p=0.044) and SCUBE2 (p=0.0018) is significantly correlated with ...
Preprint
Full-text available
Spatially resolved transcriptomics (SRT) technology enables us to gain novel insights into tissue architecture and cell development, especially tumors. However, the lack of effective methods for exploiting biological contexts (e.g., global position information) and multi-view features has severely hindered the disentangling ability for tissue heterogeneity. Here, we proposed stMVC, a multi-view graph collaborative learning model that integrates histology, gene expression, spatial location, and biological contexts in analyzing SRT data by attention. Specifically, stMVC adopting semi-supervised graph attention autoencoder separately learns view-specific representations for each of two graphs, i.e., histological similarity graph by visual features and spatial location graph by physical coordinates, and then simultaneously integrates two-view graphs for robust representations via learning weights of different views with attention in a semi-supervision manner from biological contexts. Benchmark studies of stMVC on 12 slices from the human cortex, demonstrate its superior capability in detecting tissue structure, visualizing trajectory relationships between different layers, and denoising data. In particular, in the breast cancer study, stMVC identified new disease-related cell-states and their transition cell-states, which were further validated by the functional and survival analysis of independent clinical data. Those results not only provided novel biological insights into tumor heterogeneity but also demonstrated clinical and prognostic applications from SRT data. The software is available at https://github.com/cmzuo11/stMVC.
... In addition, consistent with our results, the high expression of TBC1D 9 reduced the mortality of breast cancer patients and prolonged their survival time [35]. Although the mechanism of TBC1D 9 in breast cancer is not clear, it is significantly correlated with ESR 1, which is a risk factor for breast cancer [36]. ...
Article
Full-text available
Breast cancer patients at the same stage may show different clinical prognoses or different therapeutic effects of systemic therapy. Differentially expressed genes of breast cancer were identified from GSE42568. Through survival, receiver operating characteristic (ROC) curve, random forest, GSVA and a Cox regression model analyses, genes were identified that could be associated with survival time in breast cancer. The molecular mechanism was identified by enrichment, GSEA, methylation and SNV analyses. Then, the expression of a key gene was verified by the TCGA dataset and RT-qPCR, Western blot, and immunohistochemistry. We identified 784 genes related to the 5-year overall survival time of breast cancer. Through ROC curve and random forest analysis, 10 prognostic genes were screened. These were integrated into a complex by GSVA, and high expression of the complex significantly promoted the recurrence-free survival of patients. In addition, key genes were related to immune and metabolic-related functions. Importantly, we identified methylation of MEX3A and TBC1D 9 and mutations events. Finally, the expression of UGCG was verified by the TCGA dataset and by experimental methods in our own samples. These results indicate that 10 genes may be potential biomarkers and therapeutic targets for long-term survival in breast cancer, especially UGCG.
Article
Full-text available
Spatially resolved transcriptomics (SRT) technology enables us to gain novel insights into tissue architecture and cell development, especially in tumors. However, lacking computational exploitation of biological contexts and multi-view features severely hinders the elucidation of tissue heterogeneity. Here, we propose stMVC, a multi-view graph collaborative-learning model that integrates histology, gene expression, spatial location, and biological contexts in analyzing SRT data by attention. Specifically, stMVC adopting semi-supervised graph attention autoencoder separately learns view-specific representations of histological-similarity-graph or spatial-location-graph, and then simultaneously integrates two-view graphs for robust representations through attention under semi-supervision of biological contexts. stMVC outperforms other tools in detecting tissue structure, inferring trajectory relationships, and denoising on benchmark slices of human cortex. Particularly, stMVC identifies disease-related cell-states and their transition cell-states in breast cancer study, which are further validated by the functional and survival analysis of independent clinical data. Those results demonstrate clinical and prognostic applications from SRT data.
Article
Metabolite damage control is a critical but poorly defined aspect of cellular biochemistry, which likely involves many of the so far functionally uncharacterized protein domain (domains of unknown function; DUFs). We have determined the crystal structure of the human DUF89 protein product of the C6ORF211 gene to 1.85 Å. The crystal structure shows that the protein contains a core α-β-α fold with an active site-bound metal ion and α-helical bundle N-terminal cap, which are both conserved features of subfamily III DUF89 domains. The biochemical activities of the human protein are conserved with those of a previously characterized budding yeast homolog, where an in vitro phosphatase activity is supported by divalent cations that include Co²⁺, Ni²⁺, Mn²⁺ or Mg²⁺. Full steady-state kinetics parameters of human DUF89 using a standard PNPP phosphatase assay revealed a six times higher catalytic efficiency in presence of Co²⁺ compared to Mg²⁺. The human enzyme targets a number of phosphate substrates similar to the budding yeast homolog, while it lacks a previously indicated methyltransferase activity. The highest activity on substrate was observed with fructose-1-phosphate, a potent glycating agent, and thus human DUF89 phosphatase activity may also play a role in limiting the buildup of phospho-glycan species and their related damaged metabolites.
Article
Full-text available
s: Thirty-Second Annual CTRC‐AACR San Antonio Breast Cancer Symposium‐‐ Dec 10‐13, 2009; San Antonio, TX Aim: To identify whether breast cancer-associated SNP rs2046210 is associated with ERα level in invasive breast tumours. Background A recent genome-wide association study identified SNP rs2046210 at 6q25.1 as having a strong association with breast cancer risk1. The SNP is located 29kb upstream of the first untranslated exon of ESR1 and 180kb upstream of the transcription start site2. Rs2046210 is not in linkage disequilibrium (LD) with two of the most widely studied polymorphisms in ESR1 . We test here the hypothesis that rs2046210 may be associated with altered ESR1 expression. Methods Patients with both leukocyte DNA and invasive breast tumour paraffin blocks available were identified from two of our ongoing tissue collections: Femara Anastrazole Clinical Evaluation (FACE) and the British Breast Cancer (BBC) study. Germline DNA was extracted from bloods using the QIAamp DNA Blood Mini Kit. PCR was performed with primers spanning the SNP site, with a single basepair mismatch in the reverse sequence to generate an HhaI restriction site in the presence of the C allele. PCR product was run on a 3% agarose gel to confirm presence of a single 120bp band. Restriction digest with HhaI was then performed and products run on a 4% Metaphor agarose gel. Genotype was assigned as follows: 97bp band only = wild-type (C/C); 97bp and 120bp band = heterozygote (C/T); 120bp band only = variant (T/T). From corresponding invasive breast tumour, ERα was assessed on 4μm whole sections using clone 6F11 (Vector Labs) and quantified by H-score. Mean H-scores (left and right invasive breast tumours) were used for the BBC group. ERα- samples (H-score≤1.0) were excluded. Analysis was by ANOVA using non-parametric bias-corrected and accelerated 95% bootstrap confidence intervals (2000 replications), with genotype fitted as a score and study as a stratifying co-variate. Results Minor allele frequency was 33%, which is similar to the 38% previously reported in patients of European ancestry1. Within FACE, increased ERα was seen with presence of variant SNP. In the BBC group, variant SNP patients had higher ERα than both wild-type and heterozygote. Overall, there was a significant difference in ERα score per genotype group of 7.05, (95% CI 0.7-13.5, p=0.035). This was circa 4% difference in H-score per variant allele. Conclusion The variant genotype of SNP rs2046210 is associated with increased ERα expression. While the increase contributed by the variant allele is relatively modest, this may partly explain why the SNP is associated with increased breast cancer risk. Similar studies should be conducted in normal breast tissue.1Zheng et al. (2009) Nat. Gen . 41 (3): 324-3282Kos et al. (2001) Mol. Endocrinol . 15 : 2057-2063 View this table: H-score (mean) by genotype Citation Information: Cancer Res 2009;69(24 Suppl):Abstract nr 4138.
Article
The results presented here demonstrate that p53 upregulates estrogen receptor- (ER) expression in the human breast cancer cell line MCF-7. Two approaches were used to alter the activity of p53 in the cells. In the first approach, stable transfectants expressing an antisense p53 were established. In the stable clones, expression of antisense p53 resulted in a decrease in the expression of ER protein. In the second approach, MCF-7 cells were transiently transfected with wild-type p53. Overexpression of p53 increased the amount of ER. To determine whether the effects of p53 on the expression of ER were due to changes in transcription, deletion mutants of the ER promoter were used. This experimental approach demonstrated that p53 up-regulates ER gene expression by increasing transcription of the gene through elements located upstream of promoter A. Transfection assays using p53 mutants further demonstrated that the p53-induced increase in ER gene transcription was not dependent on the ability of p53 to bind to DNA but on its ability to interact with other proteins.
Article
s: Thirty-Second Annual CTRC‐AACR San Antonio Breast Cancer Symposium‐‐ Dec 10‐13, 2009; San Antonio, TX Aims: To (1) define the effect of oestrogen deprivation on genes related to hypoxia in oestrogen receptor positive (ER+) breast cancer and (2) identify any link between hypoxia and proliferation. Background: The majority of breast cancer patients are postmenopausal women with ER+ tumours and at some point receive an aromatase inhibitor (AI) as part of their treatment. Hypoxia and proliferation are important factors in the progression of ER+ tumours. Proliferation is profoundly reduced in most ER+ cancers after treatment with AIs[1], however little is known on the effects of AIs on hypoxia. Materials and methods: 81 pre- and 2-week post-treatment core-cut tumor biopsies were obtained from postmenopausal women with ER+ breast cancer who received single agent neoadjuvant anastrozole (AI)[2], and from 20 of these patients after 16 weeks of AI treatment. RNA was extracted and analysed on Illumina 48K microarrays. A hypoxia metagene (MG) was developed by identification of genes whose expression clustered with the expression of classical hypoxia-regulated genes[3]. Genes associated with proliferation were removed from the MG. A proliferation MG was derived by selecting the intersection of proliferation clusters from three public breast cancer datasets. Results: Spearman correlations revealed a strong relationship between the hypoxia and proliferation MGs prior to AI treatment (r =0.61, p<10-3), and persisted after 2 weeks (r =0.77, p<10-3) and after 16 weeks of treatment (r=0.72, p =0.002). Baseline expression of the hypoxia MG was (1) positively correlated with 2-week Ki67 (Spearman r =0.37, p =0.002), (2) showed a trend for a positive correlation with poor 2-week Ki67 change (Spearman r=0.22, p =0.06) and with poor reduction in the mean expression of four classical oestrogen dependant genes ( TFF1/pS2 , GREB1, PDZK1 and PGR ) known as AVERG[4] (Spearman r =0.22, p=0.06). Expression of the hypoxia MG was significantly down-regulated after 2 weeks of oestrogen deprivation using AI treatment (p<10-3). The 2-week change in hypoxia showed a positive correlation with the 2-week change in proliferation (Spearman r=0.58, p<10-3), and with the 2-week change in Ki67 (Spearman r=0.35, p=0.005). Conclusions : The expression of a hypoxia MG decreases after oestrogen deprivation. The hypoxia MG is strongly associated with proliferation prior to and after AI treatment. The data are consistent with hypoxia being a secondary effect of proliferation in ER+ breast cancer and could contribute in understanding the need to combine anti-proliferative drugs with anti-angiogenic agents for these patients. There may be a weak effect of hypoxia on de-novo resistance to AIs.1. Dowsett M, et al., Clin Cancer Res 2006; 12(3): p.1024-302. Smith, I.E, et al., J Clin Oncol 2007; 25(25): p.3816-223. Winter, S.C, et al., Cancer Res 2007; 67(7): p.3441-94. Dunbier, A.K, et al., Cancer Res 2009; 69(Suppl.), 78sSupported by The Mary-Jean Mitchell Green Foundation and Breakthrough Breast Cancer. Citation Information: Cancer Res 2009;69(24 Suppl):Abstract nr 408.
Article
Rationale: ER+/HER2+ BC may be resistant to endocrine therapies, but sensitive to HER2 inhibitors such as lapatinib. There is evidence that lapatinib may reactivate ERα expression1,2 raising the possibility that lapatinib may sensitize BC to subsequent endocrine therapy. Whether targeting the ER and HER2 in combination or in sequence in ER+/HER2+ BC may enhance anti-tumour effect is unknown. Aims: (1) To determine in ER+/HER2+ aromatase-expressing breast cancer models in vitro and in vivo.whether the combination of lapatinib with letrozole or tamoxifen is synergistic, and (2) to investigate whether using lapatinib and letrozole in sequence rather than in combination may provide a superior strategy. Methods: ER+/HER2+ BT474 cells were stably transfected with aromatase (BTArom). Combination growth studies, immunoblots and gene expression assays were conducted in BTArom cells. BTArom xenograft studies were performed over 18 weeks under androstenedione support and mice randomized to daily treatment with vehicle, lapatinib, letrozole, or lapatinib and letrozole (L+L). In addition, mice in the lapatinib alone arm, were randomized at 6 weeks to continued lapatinib, vs. switch to letrozole, vs switch to L+L. Results: Formal combination studies in aromatase-transfected BT474 (BTArom) cells demonstrated synergy between lapatinib and letrozole (combination index, CI=0.4, p<10-5), but antagonism between lapatinib and 4-OH-tamoxifen (CI=1.34, p=0.05). In BTArom cells, lapatinib increased ESR1 2.8-fold (P<0.0001) and ERα protein expression 2- fold. As expected, androstenedione upregulated both ER-controlled genes, PGR and GREB1, however lapatinib treatment increased PGR and GREB1 further, by 4.6- and 1.8—fold over androstenedione alone (p=0.0001 and p=0.045, respectively). In long-term BTArom xenografts the combination of lapatinib and letrozole was no more effective at controlling tumour growth than either treatment alone (Kruskall-Wallis comparing treatment arms, P>0.05). However for the group of mice treated with lapatinib for 6 weeks followed by the combination of lapatinib and letrozole for 12 weeks, tumour volume increased only 2.2-fold, compared to 4.5-fold for those continuing on lapatinib, 5.7-fold for those switching to letrozole and 5.2-fold for those on lapatinib and letrozole from the start. Conclusion: We have demonstrated that HER2 targeted therapy enhanced responsiveness to aromatase inhibition in an ER+/HER2+ breast cancer model in vitro and in vivo. In contrast, the interaction between lapatinib and 4-OH-tamoxifen was antagonistic. Lapatinib increased ER expression and function, and the most effective tumour control was achieved with lapatinib alone followed by the combination of lapatinib and letrozole. Maximizing benefit from ER and HER2 targeted therapies in ER+/HER2+ BC may involve sequential rather than concurrent treatment to exploit lapatinib-induced ER reactivation. 1 Leary et al, Clin Canc Res 2010 2Xia et al, PNAS 2006 Citation Information: Cancer Res 2010;70(24 Suppl):Abstract nr P3-14-03.
Article
Results presented in this study demonstrate that treatment of MCF-7 cells with taxol resulted in induction of estrogen receptor-alpha (ER alpha) gene transcription with a subsequent increase in ER alpha mRNA; this effect was promoter specific since taxol did not affect total transcription in MCF-7 cells and lacked an effect on transcription of the human acidic ribosomal phosphoprotein protein PO, progesterone receptor, and pS2 genes. In contrast to the increase in transcription of the ER alpha gene, taxol inhibited translation of the ER alpha mRNA. This effect is also transcript specific since taxol did not alter total protein synthesis and did not affect the concentration of progesterone receptor protein in the cell. The overall result of taxol treatment was to decrease the concentration of ER alpha protein in the MCF-7 cells. Evidence is presented that the effects of taxol on ER alpha gene transcription may be mediated through the induction of p53.