Open Access

A novel FOXA1/ESR1 interacting pathway: A study of Oncomine™ breast cancer microarrays

  • Authors:
    • Sanjib Chaudhary
    • B. Madhu Krishna
    • Sandip K. Mishra
  • View Affiliations

  • Published online on: June 7, 2017     https://doi.org/10.3892/ol.2017.6329
  • Pages: 1247-1264
  • Copyright: © Chaudhary et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Forkhead box protein A1 (FOXA1) is essential for the growth and differentiation of breast epithelium, and has a favorable outcome in breast cancer (BC). Elevated FOXA1 expression in BC also facilitates hormone responsiveness in estrogen receptor (ESR)‑positive BC. However, the interaction between these two pathways is not fully understood. FOXA1 and GATA binding protein 3 (GATA3) along with ESR1 expression are responsible for maintaining a luminal phenotype, thus suggesting the existence of a strong association between them. The present study utilized the Oncomine™ microarray database to identify FOXA1:ESR1 and FOXA1:ESR1:GATA3 co‑expression co‑regulated genes. Oncomine™ analysis revealed 115 and 79 overlapping genes clusters in FOXA1:ESR1 and FOXA1:ESR1:GATA3 microarrays, respectively. Five ESR1 direct target genes [trefoil factor 1 (TFF1/PS2), B-cell lymphoma 2 (BCL2), seven in absentia homolog 2 (SIAH2), cellular myeloblastosis viral oncogene homolog (CMYB) and progesterone receptor (PGR)] were detected in the co‑expression clusters. To further investigate the role of FOXA1 in ESR1‑positive cells, MCF7 cells were transfected with a FOXA1 expression plasmid, and it was observed that the direct target genes of ESR1 (PS2, BCL2, SIAH2 and PGR) were significantly regulated upon transfection. Analysis of one of these target genes, PS2, revealed the presence of two FOXA1 binding sites in the vicinity of the estrogen response element (ERE), which was confirmed by binding assays. Under estrogen stimulation, FOXA1 protein was recruited to the FOXA1 site and could also bind to the ERE site (although in minimal amounts) in the PS2 promoter. Co‑transfection of FOXA1/ESR1 expression plasmids demonstrated a significantly regulation of the target genes identified in the FOXA1/ESR1 multi‑arrays compared with only FOXA1 transfection, which was suggestive of a synergistic effect of ESR1 and FOXA1 on the target genes. In summary, the present study identified novel FOXA1, ESR1 and GATA3 co‑expressed genes that may be involved in breast tumorigenesis.

Introduction

The majority of breast cancers (BCs) are generally hormone-related cancers, with estradiol (E2) essentially being the primary inducing factor (1,2). In women, E2 promotes cell proliferation, growth and development of the mammary epithelium (3,4). The mammary epithelium is composed of basal and myoepithelial/basal cell lineages (5). Approximately 15–25% of mammary epithelial cells express estrogen receptor 1 (ESR1) in the normal resting breast, and are considered to proliferate slowly and in a well-differentiated cell-type (6). However, the number of ESR1-positive mammary cells changes throughout the menstrual cycle (79). Notably, E2 induces the proliferation of ESR1-negative breast cells that surround the ESR1-positive cells, probably through the secretion of paracrine factors (6,7). E2 is also known to promote proliferation in a large number of BCs, with positive correlation between ESR1 positivity and endocrine therapy (10). In addition, the number of mammary epithelial cells and the expression of ESR1 increase to >50% during initial diagnosis, which suggests a transformation role that provides a target for therapy (8,9). Apart from cellular transformation, ESR1 also plays a pivotal role in cell proliferation and growth (11,12). Approximately 70% of BCs are ESR+ or E2-responsive (13). The presence of ESR1 is a good predictive and prognostic factor for BC patients, who are likely to respond to anti-hormone therapy with tamoxifen or aromatase inhibitors (8). The use of adjuvant therapy such as tamoxifen results in ~40–50% reduction in recurrence and prolonged disease-free and overall patient survival (14), and also provides a clinical benefit for >50% of all metastatic ESR1+ tumors (15). Although tamoxifen is initially effective, ~50% of breast tumors acquire tamoxifen resistance during the course of treatment (1618). Such a situation has resulted in the quest for developing novel selective ESR modulators.

Forkhead box A1 (FOXA1) is a forkhead family member protein encoded by the FOXA1 gene, which is located on chromosome 14q21.1 (19,20). FOXA1 was initially identified as a vital factor for liver development by transcriptionally activating the liver-specific transcripts albumin and transthyretin (21); however, its role in the development of the breast and other organs has also been reported (2225). FOXA proteins bind to DNA elements [A(A/T)TRTT(G/T)RYTY] as monomers to mediate their physiological response (6). These proteins are similar to histone linker proteins, but unlike histones, they lack basic amino acids that are essential for chromatin compaction (26). FOXA1 protein also has the potential to compact chromatin and reposition the nucleosome by recruiting itself to enhancer regions of the target genes (20). The repositioning of nucleosomes is considered to facilitate the temporal and spatial differential binding of transcription factors in a lineage-specific manner (27). As observed in rescue experiments in FOXA1-null mice, FOXA1 is responsible for post-natal development of mammary and prostate glands (25). Apart from development, FOXA1 was observed to be highly elevated in prostate cancer and BC (28,29). In ESR+ BC cells, FOXA1 facilitates hormone responsiveness by modulating ESR1 binding sites in the target genes (30,31). Thorat et al demonstrated that ~50% of ESR1-regulated target genes and E2-induced cell proliferation requires prior FOXA1 protein recruitment (32). Furthermore, FOXA1 expression is also associated with low breast tumor grade, exhibiting a positive correlation with the luminal A BC subtype (33). Such observation suggests a strong correlation between FOXA1 expression and luminal A breast tumor subtype; however, the co-regulatory partners of both molecules are still undefined.

GATA binding protein 3 (GATA3) is one of the six members of the zinc finger DNA binding protein family (22). It binds to the DNA sequence (A/T)GATA(A/G) in the target gene, and promotes cell proliferation, development and differentiation of different tissues and cell types (34,35), including the luminal glandular epithelial cells of the mammary gland (3638). The genes GATA3, FOXA1 and ESR1 are highly expressed in BC, with positive correlation between them (39). ESR1 messenger RNA (mRNA) is transcribed from ~6 promoter regions with different tissue specificity (40). The regulatory factors involved in GATA3 and FOXA1 expression may interact with the ESR1 promoter region, although this remains to be determined (28). However, a previous whole genome expression analysis demonstrated that FOXA1 and GATA3 protein express in close association with ESR1 (41).

Previous studies have utilized the Oncomine™ software (Thermo Fisher Scientific, Inc., Waltham, MA, USA) to correlate published microarray data (42,43) in order to confirm the authenticity of the correlation data. The Oncomine™ software enables to understand and analyze a number of microarray data (multi-array), which contain multiple clinical tumor samples and normal biopsies (44). The software function search tool allows the queried gene to be correlated in terms of its expression with other genes in the multi-arrays (www.oncomine.org). Such analyses will yield a significant overlap of co-expressed genes that can link proteins in the same molecular pathway.

The objective of the present study was to compare the co-expressed target genes of FOXA1 and to correlate them with ESR1 and GATA3 in order to determine the extent of overlap using Oncomine™ microarray data. For that purpose, an intensive individual meta-analysis of FOXA1, ESR1 and GATA3 (putative pathway partners that may be associated in BC tumorigenesis) was performed, followed by a comparison of the overlapping genes. Such comparisons would provide a highly significant number of genes that may be involved in the same pathway. Analyses of the Oncomine™ microarray data identified 115 co-regulated genes between FOXA1 and ESR1. Comparison of these genes with another co-related and co-regulated gene, GATA3, identified 79 genes that are co-expressed along with FOXA1 and ESR1 co-regulated genes, which are consistent with the previously reported estrogen- and ESR1-regulated pathway. Semiquantitative and quantitative polymerase chain reaction (qPCR) analysis also confirmed a number of the overlapping genes [PS2, B-cell lymphoma 2 (BCL2), progesterone receptor (PGR), seven in absentia homolog 2 (SIAH2), cellular myeloblastosis viral oncogene homolog (CMYB) and GATA3], which suggested a significant correlation. In silico analysis of one of the significantly associated genes, PS2, demonstrated the presence of two FOXA1 binding sites and an estrogen response element (ERE), which was observed to recruit FOXA1 upon E2 stimulation.

The present findings reveal novel co-expression partners and the existence of a molecular network involving interacting partners in the FOXA1, ESR1 and GATA3 signaling pathways.

Materials and methods

Oncomine™ analysis

Oncomine™ is an integrated cancer microarray database and web-based data-mining platform (44). Oncomine™ analysis was performed as previously described (42,43). The co-expressed genes correlated with FOXA1 and ESR1 were searched for in the Oncomine™ platform. A total of 24 microarrays were selected, 20 of which were ESR+ BC microarrays, while the remaining 4 were normal ESR+ breast microarrays (Table I) (4568). All the ESR+ microarrays were selected for co-expression analysis. The first 500 genes co-regulated with FOXA1 and ESR1 within each microarray were retrieved and compared separately. These 500 genes were selected based on a >2 fold-change expression level and in an adjusted threshold by gene rank for the top 10%. Such a threshold will return mRNA datasets having breast cancer clinical samples, with FOXA1 and ESR1 coexpression results ranked or grouped in the top 10% of the datasets. Therefore by examining these coexpression results we can determine genes that are coordinately expressed with FOXA1 and ESR1, which may help to identify potential targets in the same pathway. The repetitive genes within each study (FOXA1 and ESR1) were removed, keeping only a single representative of the gene in each microarray analysis. The gene names were derived from GeneCards® (http://www.genecards.org/). To understand the significant correlations, genes represented on >4 microarrays were considered significant (16% frequency), and those represented on >5 microarrays were considered highly significant (20% frequency). Genes from the FOXA1 and ESR1 microarrays were sorted and overlapped to identify overlapping co-expressed genes. Such microarray coexpression analysis may help to identify potential targets that function in the same regulatory pathway.

Table I.

Forkhead box protein A1:estrogen receptor 1 microarray used for the analysis.

Table I.

Forkhead box protein A1:estrogen receptor 1 microarray used for the analysis.

AuthorTypeaSample numbersRef.
Higgins et alNormal  34(45)
Roth et alNormal353(46)
Shyamsundar et alNormal123(47)
Tabchy et alBreast178(48)
Perou et alBreast  65(49)
Su et alNormal101(50)
Zhao et alBreast  64(51)
Yu et alBreast 3  96(52)
Wang et alBreast286(53)
Waddell et alBreast  85(54)
Van't Veer et alBreast117(55)
Schmidt et alBreast200(56)
Pollack et alBreast 2  41(57)
Minn et alBreast 2121(58)
Lu et alBreast129(59)
Korde et alBreast  61(60)
Kao et alBreast327(61)
Julka et alBreast  44(62)
Hatzis et alBreast508(63)
Gluck et alBreast158(64)
Farmer et alBreast  49(65)
Desmedt et alBreast198(66)
Bos et alBreast204(67)
Bonnefoi et alBreast160(68)

a According to the Oncomine database acronym.

Cell culture and transient transfection

The cell lines MCF7 and T47D were purchased from the National Center for Cell Sciences (Pune, India). The MCF7 and T47D cell lines were cultured in Dulbecco's modified Eagle medium (DMEM; PAN Biotech GmbH, Aidenbach, Germany) and RPMI 1640 medium (PAN Biotech GmbH) respectively, supplemented with 10% (v/v) fetal bovine serum (PAN Biotech GmbH) and 1% (v/v) penicillin/streptomycin (Sigma-Aldrich, St. Louis, MO, USA). The cells were maintained under a humidified atmosphere in 5% CO2 at 37°C. The plasmids pRB-HNF3α (expressing FOXA1) and pAcGFPC1-ESR1 (expressing ESR1) were provided by Professor Kenneth S. Zaret (Department of Cell and Developmental Biology, Smilow Center for Translational Research, Philadelphia, PA, USA) and Professor Ratna K. Vadlamudi (The Department of Obstetrics and Gynecology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA), respectively.

To investigate the role of FOXA1 in the transcriptional regulation of target genes, FOXA1 expression plasmid (1 µg) and empty vector (1 µg) were transfected in MCF7 and T47D cells cultured in 35-mm plates (BD Biosciences, Franklin Lakes, NJ, USA) using the TransPass D2 transfection reagent (New England BioLabs, Inc., Ipswich, MA, USA). Transfected and untransfected cell lines were harvested at 24 h post-transfection. Similarly, co-transfection was performed by transfecting FOXA1 (500 ng) and ESR1 (500 ng) expression plasmids. After 24 h of transfection, total RNA was isolated and processed.

RNA isolation, reverse transcription-PCR and qPCR

Total RNA was isolated from FOXA1-transfected and ESR1/FOXA1-co-transfected samples at 24 h post-transfection using TRI reagent (Sigma-Aldrich). RNA was digested with DNase I (Sigma-Aldrich) digested converted into complementary DNA (cDNA) using a first-strand cDNA synthesis kit (Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA). The qPCR conditions were as follows: 95°C for 2 min, followed by 40 cycles of 95°C for 30 sec and 56–58°C for 30 sec). GAPDH was used as a internal control. The relative quantification of gene expression was calculated by the 2−∆∆Cq method (69). The primers used for PCR are listed in Table II. qPCR was performed using SYBR® Green (Sigma-Aldrich) with an MJ Research thermocycler (Bio-Rad Laboratories, Inc., Hercules, CA, USA).

Table II.

Lists of primers used.

Table II.

Lists of primers used.

PrimersPrimer sequence (5′-3′)Amplicon size (bp)
RT-FOXA1F: GGGTGGCTCCAGGATGTTAGG194
R: GGGTCATGTTGCCGCTCGTAG
RT-GATA3F: CAGACCACCACAACCACACTCT124
R: GGATGCCTCCTTCTTCATAGTCA
RT-PGRF: CGCGCTCTACCCTGCACTC121
R: TGAATCCGGCCTCAGGTAGTT
RT-CMYBF: GAAGGTCGAACAGGAAGGTTATCT224
R: GTAACGCTACAGGGTATGGAACA
RT-SIAH2F: CCTCGGCAGTCCTGTTTCCCTGT124
R: CCAGGACATGGGCAGGAGTAGGG
RT-BCL2F: TGTGGATGACTGAGTACCTG116
R: GGAGAAATCAAACAGAGGCC
RT-PS2F: GAACAAGGTGATCTGCGCCC223
R: TTCTGGAGGGACGTCGATGG
RT-GAPDHF: AAGATCATCAGCAATGCCTC619
R: CTCTTCCTCTTGTGCTCTTG
FOXA1 chip (FOXA1 site1) PS2F: CATGTTGGCCAGGCTAGTCT165
R: CATTCCGTCTAGGCCTAAGC
FOXA1 chip (FOXA1 site2) PS2F: GCTTAGGCCTAGACGGAATG180
R: CTCATATCTGAGAGGCCCTC
PS2 chip F (ERE)F: TTAAGTGATCCGCCTGCTTT271
R: CTCCCGCCAGGGTAAATACT
FOXA1 consensus siteF: CTTATGCAATGTGTTGGTCTCACG
R: CGTGAGACCAACACATTGCATAAG
FOXA1 EMSA (FOXA1 site1) PS2 GGCCTCCCAAAGTGTTGGGATTACAGGCGT
ACGCCTGTAATCCCAACACTTTGGGAGGCC
FOXA1 EMSA (FOXA1 site2) PS2 CCCCGTGAGCCACTGTTGTCACGGCCAAG
CTTGGCCGTGACAACAGTGGCTCACGGGG

[i] RT, reverse transcription; FOXA1, forkhead box protein A1; EMSA; electrophoretic mobility shift assay; GATA3, GATA binding protein 3; PGR, progesterone receptor; BCL2, B-cell lymphoma 2; GAPDH, glyceraldehyde 3-phosphate dehydrogenase; ERE, estrogen response element; F, forward; R, reverse; PS2, trefoil factor 1; SIAH2, seven in absentia homolog 2; CMYB, cellular myeloblastosis viral oncogene homolog

Nuclear extract

Nuclear lysate was extracted from MCF7 cells. The cells were washed with ice-cold phosphate-buffered saline (PBS) and lysed with cell lysis buffer [20 mM 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid (HEPES) (pH 7.9), 50% (v/v) glycerol, 0.1% (v/v) Triton X-100, 10 mM NaCl, 1.5 mM MgCl2, 1 mM ethylene glycol-bis(β-aminoethyl ether)-N,N,N',N'-tetraacetic acid (EGTA), 1 mM ethylenediaminetetraacetic acid (EDTA) and 1X protease inhibitor cocktail] (Sigma-Aldrich) for 15 min in 4°C. Nuclear pellets were collected upon centrifugation at 500 × g for 15 min, and resuspended in chilled extraction buffer [20 mM HEPES (pH=7.9), 50% (v/v) glycerol, 420 mM NaCl, 10 mM MgCl2, 1 mM EGTA, 1 mM dithiothreitol (DTT) and 1X protease inhibitor cocktail] (Sigma-Aldrich). After 30 min of incubation on ice, the nuclear proteins were collected by centrifugation at 16,000 × g at 4°C for 30 min. The lysate prepared was stored at −80°C prior to use.

Electrophoretic mobility shift assay (EMSA)

In vitro DNA-protein interaction was performed using EMSA. Oligonucleotides consisting of FOXA1 binding sites present in the PS2 promoter were designed from −517 to −547 (EMSA1) and from −363 to −393 (EMSA2) residues upstream of the transcription start site. The oligonucleotide sequences are provided in Table II. The forward primers of EMSA1 and EMSA2 were kinase-labeled with γ32P adenosine triphosphate (BRIT, Hyderabad, India), and then annealed with reverse complementary oligonucleotide residues in annealing buffer [200 mM Tris-Cl (pH 7.5), 1,000 mM NaCl and 100 mM MgCl2]. The nuclear lysate was incubated in 10 µl binding buffer [1 M Tris-Cl (pH 7.5), 50% (v/v) glycerol, 0.5 M EDTA, 1 mM DTT and 50 mg/ml bovine serum albumin; Sigma-Aldrich) containing 0.2 pmol radiolabeled probe. Poly(deoxyinosinic-deoxycytidylic) acid was used as a nonspecific competitor. For specific competition, the radiolabeled probes were mixed to compete with various excess molar concentrations of unlabeled double-stranded FOXA1 consensus probe. After 25 min of incubation at room temperature, the samples were subjected to electrophoresis in a 6% polyacrylamide gel at 180 V in 0.5X Tris/borate/EDTA running buffer [40 mM Tris-Cl (pH 8.3), 45 mM boric acid and 1 mM EDTA] for 1 h. Subsequently, the gel was dried and autoradiographed.

Chromatin immunoprecipitation (ChIP) assay

For in vivo binding assays, ChIP was performed. Prior to E2 treatment, MCF7 cells were maintained in phenol-free DMEM (PAN Biotech GmbH) for 48 h. The cells were stimulated with 100 nM E2 (Sigma-Aldrich) for additional 24 h, fixed with 1% (v/v) formaldehyde for 10 min, washed twice with 1X PBS (10 mM PO43−, 137 mM NaCl and 2.7 mM KCl), lysed with cell lysis buffer [1% (v/v) sodium dodecyl sulfate (SDS), 10 mM EDTA, 50 mM Tris-Cl (pH 8.1) and 1X protease inhibitor cocktail] (Sigma-Aldrich) and sonicated at M2 amplitude strength (~250W intensity level) using a Bioruptor® ultrasonicator device (Diagenode S.A., Seraing, Belgium). The sonicated samples were pre-cleared using protein A-sepharose beads (GE Healthcare Life Sciences, Chalfont, UK) and incubated with 1 µg anti-FOXA1 (catalog no., sc101058; Santa Cruz Biotechnology, Inc., Dallas, TX, USA), anti-ESR1 (catalog no., 8644s; Cell Signaling Technology, Inc., Danvers, MA, USA), normal mouse immunoglobulin G (IgG) (catalog no., kch-819-015; Diagenode S.A.) and normal rabbit IgG (catalog no., sc-2027; Santa Cruz Biotechnology, Inc.) antibodies (diluted, 1:100) at 4°C for 1 h. The antibody-protein complexes were separated using protein A-sepharose beads for an additional 1 h, and washed with different washing buffers, including a low salt wash buffer [0.1% (v/v) SDS, 1% (v/v) Triton X-100, 2 mM EDTA, 20 mM Tris-HCl (pH 8.1) and 150 mM NaCl], a high salt wash buffer [0.1% (v/v) SDS, 1% (v/v) Triton X-100, 2 mM EDTA, 20 mM Tris-HCl (pH 8.1) and 500 mM NaCl], a LiCl wash buffer [0.25 M LiCl, 1% (v/v) NP-40, 1% (w/v) deoxycholic acid (sodium salt), 1 mM EDTA and 10 mM Tris-HCl (pH 8.1)] and 1X Tris/EDTA [10 mM Tris-HCl (pH 8.1) and 1 mM EDTA]. The samples were then eluted with elution buffer [1% (v/v) SDS and 0.1 M NaHCO3], reverse crosslinked with 5 mM NaCl for 6 h at 65°C and subjected to proteinase K digestion at 45°C for 1 h. The ChIP eluates were purified by phenol-chloroform, and the purified DNA fractions were used to perform PCR analysis to confirm the presence of ESR1 and FOXA1 binding in the PS2 promoter (Table II).

Statistical analysis

Data are shown as representative experiments performed in triplicates, and represented as the mean ± standard error. Differences were compared with the paired Student's t-test. All statistical tests were performed with GraphPad Prism version 5.01 (GraphPad Software, Inc., La Jolla, CA, USA). P<0.05 was considered to indicate a statistically significant difference.

Results and Discussion

Co-expression meta-analysis was performed using Oncomine™ (www.oncomine.org), which is a web-based interface cancer-profiling database containing published microarray data that have been collected, analyzed, annotated and maintained by Compendia Bioscience™ (Thermo Fisher Scientific, Inc., Waltham, MA, USA). The co-expression genes for ESR1 and FOXA1 were searched and analyzed in the multi-arrays (Table I). The first 500 highly co-expressed genes (exhibiting both significantly low and high expression) with a cut-off frequency of ≥4 (≥16%) studies in each microarray were selected (Tables III and IV). Approximately 16–20% of genes were observed to overlap with each other when the co-expressed genes of ESR1 and FOXA1 were combined (Fig. 1A and B). Under higher stringent conditions with a cut-off frequency of ≥5 (≥20%), ~115 genes overlapped in ESR1 and FOXA1 co-expression genes multi-arrays (Fig. 1B). Table V presents the overlapping genes of ESR1 and FOXA1 identified in the aforementioned multi-arrays.

Table III.

FOXA1 Oncomine™ meta-analysis.

Table III.

FOXA1 Oncomine™ meta-analysis.

GenePercentage of co-expression (%)
FOXA1100
ESR1  67
GATA3  67
MLPH  67
AGR2  63
CA12  63
TFF3  63
XBP1  63
NAT1  58
SLC39A6  58
TBC1D9  58
DNALI1  54
SCNN1A  54
SLC44A4  54
SPDEF  54
TSPAN1  54
ANXA9  50
DNAJC12  50
FBP1  50
GREB1  50
MAGED2  50
MAPT  50
MYB  50
TFF1  50
AR  46
FAM174B  46
INPP4B  46
KDM4B  46
SCUBE2  46
SIDT1  46
VAV3  46
ABAT  42
BCL2  42
GPD1L  42
IL6ST  42
RHOB  42
TTC39A  42
ACADSB  38
ERBB4  38
EVL  38
NME5  38
SYBU  38
TOX3  38
ZNF552  38
CACNA1D  33
DACH1  33
GALNT6  33
GAMT  33
GFRA1  33
RAB17  33
RBM47  33
SLC16A6  33
SLC7A833
STC233
TSPAN1333
ZMYND1033
AFF329
AKR7A329
C10orf11629
C9orf11629
CRIP129
CYB5A29
ELOVL529
GALNT729
KCNK1529
KIAA132429
LASS629
MCCC229
MTL529
PGR29
RAB2629
SERPINA529
SIAH229
SLC2A1029
AGR325
CAMK2N125
CYP2B7P125
FAM134B25
GPR16025
GSTM325
INPP5J25
KIF5C25
MAST425
MED13L25
NPDC125
PNPLA425
PP1457125
RABEP125
SCCPDH25
SEMA3B25
SEMA3F25
STARD1025
SYT1725
THSD425
UGCG25
ABCC821
ABLIM321
BCAS121
C5orf3021
C6orf9721
C9orf15221
CLSTN221
CYP2B621
DHCR2421
DUSP421
DYNLRB221
EFHC121
ERBB321
FAAH21
FSIP121
GDF1521
IRS121
KCTD321
KIAA004021
KIF16B21
KRT1821
LRBA21
METRN21
MREG21
MYO5C21
PECI21
PRR1521
PTPRT21
PVRL221
REEP121
REEP621
RERG21
RNF10321
SLC19A221
SLC22A521
SLC4A821
SYTL221
TBX321
TMC521
TMEM30B21
TP53TG121
TTC621
WFS121
ADCY917
ANKRD30A17
APBB217
AZGP117
BBS417
C17orf2817
C1orf2117
C1orf6417
C4A17
CACNA2D217
CASC117
CCNG217
CELSR217
CLGN17
COX6C17
CPB117
CREB3L417
CXXC517
CYP4B117
DEGS217
EEF1A217
FAM110C17
FUT817
HHAT17
HPN17
IGF1R17
KIAA023217
KIAA124417
KRT817
LRIG117
MEIS3P117
MKL217
MYST417
NBEA17
NPNT17
NRIP117
PBX117
PCSK617
RAB27B17
RALGPS217
RND117
SLC9A3R117
SPRED217
STK32B17
WWP117
ZNF70317

[i] FOXA1, forkhead box protein A1.

Table IV.

ESR1 Oncomine™ meta-analysis.

Table IV.

ESR1 Oncomine™ meta-analysis.

GenePercentage of co-expression (%)
ESR1100
CA12  79
GATA3  79
NAT1  71
SLC39A6  71
TBC1D9  71
DNALI1  67
FOXA1  67
ANXA9  63
DNAJC12  63
GREB1  63
MAPT  63
ABAT  58
SCUBE2  58
TFF3  58
ERBB4  54
KDM4B  54
MLPH  54
MYB  54
XBP1  54
AGR2  50
DACH1  50
FBP1  50
IL6ST  50
MAGED2  50
TFF1  50
VAV3  50
ACADSB  46
GFRA1  46
INPP4B  46
KIAA1324  46
PGR  46
SCNN1A  46
SLC44A4  46
SLC7A8  46
SPDEF  46
BCL2  42
C9orf116  42
CACNA1D  42
EVL  42
GAMT  42
GPD1L  42
NME5  42
SERPINA5  42
STC2  42
SYBU  42
TTC39A  42
ZMYND10  42
AFF3  38
AGR3  38
AR  38
FAM174B  38
SIDT138
THSD438
TSPAN138
CLSTN233
CYP2B633
CYP2B7P133
ELOVL533
FAM134B33
KCNK1533
RERG33
RHOB33
SLC16A633
SLC22A533
UGCG33
ZNF55233
ABCC829
C5orf3029
C6orf9729
CYB5A29
DYNLRB229
GSTM329
IRS129
MAST429
MCCC229
MTL529
PNPLA429
PTPRT29
RABEP129
SEMA3B29
SIAH229
SUSD329
SYT1729
TSPAN1329
ABLIM325
ADCY925
AKR7A325
C10orf11625
CACNA2D225
CASC125
CRIP125
CXXC525
ERBB325
FSIP125
GALNT625
HHAT25
INPP5J25
KCTD325
KIF5C25
MED13L25
NRIP125
RAB1725
RBM4725
SCCPDH25
SEMA3F25
SLC2A1025
TBX325
TOX325
WFS125
WWP125
ACOX221
ANKRD30A21
APBB221
C4A21
CAMK2N121
CCDC74B21
CCNG221
COX6C21
DEGS221
EEF1A221
EFHC121
FAAH21
FUT821
GALNT721
IGF1R21
KIAA004021
LASS621
LRBA21
LRIG121
MEIS3P121
METRN21
MREG21
NPDC121
NPNT21
PDZK121
PRSS2321
RAB2621
REPS221
RNF10321
SALL221
STK32B21
ZNF70321
ASTN217
AZGP117
BBS117
BBS417
BCAS117
C14orf4517
C16orf4517
C1orf6417
C6orf21117
CAPN817
CELSR217
CPB117
CYP4B117
DHCR2417
GDF1517
HPN17
KIAA023217
LONRF217
MKL217
MYO5C17
MYST417
NKAIN117
PARD6B17
PBX117
PCP217
PCSK617
PECI17
PLAT17
PLCD417
PP1457117
PP1R3C17
PREX117
PRLR17
RALGPS217
RARA17
REEP117
REEP617
SEC14L217
SEMA3C17
SERPINA317
SLC19A217
SLC22A1817
SLC27A217
SSH317
STARD1017
SYTL217
TCEAL117
TMEM2517
TMEM30B17
TP53TG117
TPRG117
WNK417

[i] ESR1, estrogen receptor 1.

Table V.

Overlapping meta-analysis of ESR1 and FOXA1 with a cut-off frequency of 5 (20%).

Table V.

Overlapping meta-analysis of ESR1 and FOXA1 with a cut-off frequency of 5 (20%).

Overlap of ESR1 and FOXA1 (≥5 studies, ESR1=143, FOXA1=138, overlapping genes=115)

GeneFOXA1 (%)ESR1 (%)Function
ESR1  67100Estrogen receptor 1
CA12  6379Carbonic anhydrase 12
GATA3  6779GATA binding protein 3
NAT1  5871NAT1 N-acetyltransferase 1
SLC39A6  5871Zinc transporter ZIP6
TBC1D9  5871TBC1 domain family member 9
DNALI1  5467Axonemal dynein light intermediate polypeptide 1
FOXA110067Forkhead box protein A1
ANXA9  5063Annexin A9
DNAJC12  5063DnaJ homolog subfamily C member 12
GREB1  5063Growth regulation by estrogen in breast cancer 1
MAPT  5063 Microtubule-associated protein tau
NPDC1  2563Neural proliferation differentiation and control protein 1
ABAT  42584-aminobutyrate aminotransferase
SCUBE2  4658Signal peptide, CUB domain, EGF-like 2
TFF3  6358Trefoil factor 3
ERBB4  3854Receptor tyrosine-protein kinase erbB-4
KDM4B  4654Lysine (K)-specific demethylase 4B
MLPH  6754Melanophilin
MYB  5054Myb proto-oncogene protein
XBP1  6354X-box binding protein 1
AGR2  6350Anterior gradient homolog 2
DACH1  3350Dachshund homolog 1
FBP1  5050 Fructose-1,6-bisphosphatase 1
IL6ST  4250Glycoprotein 130
MAGED2  5050Melanoma antigen fmily D, 2
TFF1  5050Trefoil factor 1
VAV3  4650Guanine nucleotide exchange factor
ACADSB  3846Acyl-CoA dehydrogenase, short/branched chain
GFRA1  3346GDNF family receptor alpha-1
INPP4B  4646Inositol polyphosphate-4-phosphatase
KIAA1324  2946Estrogen-induced gene 121
PGR  2946Progesterone receptor
SCNN1A  5446Sodium channel, non-voltage-gated 1 alpha subunit
SLC44A4  5446Choline transporter-like protein 4
SLC7A8  3346Solute carrier family 7 (amino acid transporter light chain, L system)
SPDEF  5446SAM pointed domain-containing ETS transcription factor
BCL2  4242B-cell lymphoma 2
C9orf116  2942Chromosome 9 open reading frame 116
CACNA1D  3342Calcium channel, voltage-dependent, L type, alpha 1D subunit
EVL  3842Enah/Vasp-like
GAMT  3342Guanidinoacetate N-methyltransferase
GPD1L  4242 Glycerol-3-phosphate dehydrogenase 1-like
NME5  3842NME/NM23 family member 5
SERPINA5  2942Serpin peptidase inhibitor, clade A (alpha-1 antiproteinase, antitrypsin), member 5
STC2  3342 Stanniocalcin-related protein
SYBU  3842Syntabulin (syntaxin-interacting)
TTC39A  4242Tetratricopeptide repeat domain 39A
ZMYND10  3342Zinc finger, MYND-type containing 10
AFF32938AF4/FMR2 family, member 3
AGR32538Anterior gradient 3 homolog (xenopus laevis)
AR4638Androgen receptor
FAM174B4638Family with sequence similarity 174, member B
SIDT14638SID1 transmembrane family, member 1
THSD42538Thrombospondin, type I, domain containing 4
TSPAN15438Tetraspanin 1
CLSTN22133Calsyntenin 2
CYP2B62133Cytochrome P450, family 2, subfamily B, polypeptide 6
CYP2B7P12533Cytochrome P450, family 2, subfamily B, polypeptide 7 pseudogene 1
ELOVL52933ELOVL fatty acid elongase 5
FAM134B2533Family with sequence similarity 134, member B
KCNK152933Potassium channel, subfamily K, member 15
RERG2133RAS-like, estrogen-regulated, growth inhibitor
RHOB4233Ras homolog family member B
SLC16A63333Solute carrier family 16, member 6 (monocarboxylic acid transporter 7)
SLC22A52133Solute carrier family 22 (organic cation/carnitine transporter), member 5
UGCG2533UDP-glucose ceramide glucosyltransferase
ZNF5523833Zinc finger protein 552
ABCC82129ATP-binding cassette transporter sub-family C member 8
C5orf302129Chromosome 5 open reading frame 30
C6orf972129Chromosome 6 open reading frame 97
CYB5A2929Cytochrome B5 type A (microsomal)
DYNLRB22129Dynein, light chain, roadblock-type 2
GSTM32529Glutathione S-transferase mu 3 (brain)
IRS12129Insulin Receptor Substrate 1
MAST42529Microtubule associated serine/threonine kinase family member 4
MCCC22929Methylcrotonoyl-CoA carboxylase 2 (beta)
MTL52929 Metallothionein-like 5, testis-specific (tesmin)
PNPLA42529Patatin-like phospholipase domain containing 4
PTPRT2129Protein tyrosine phosphatase, receptor type, T
RABEP12529Rabaptin, RAB GTPase binding effector protein 1
SEMA3B2529Sema domain, immunoglobulin domain (Ig), short basic domain, secreted, (semaphorin) 3B
SIAH22929Siah E3 ubiquitin protein ligase 2
SYT172529Synaptotagmin XVII
TSPAN133329Tetraspanin 13
ABLIM32125Actin binding LIM protein family, member 3
AKR7A32925Aldo-keto reductase family 7, member a3 (aflatoxin aldehyde reductase)
C10orf1162925Chromosome 10 open reading frame 116
CRIP12925Cysteine-rich protein 1 (intestinal)
ERBB32125V-Erb-B2 erythroblastic leukemia viral oncogene homolog 3 (avian)
FSIP12125Fibrous sheath interacting protein 1
GALNT63325Polypeptide N-acetylgalactosaminyltransferase 6
INPP5J2525Inositol polyphosphate-5-phosphatase J
KCTD32125Potassium channel tetramerisation domain containing 3
KIF5C2525Kinesin family member 5C
MED13L2525Mediator complex subunit 13-like
RAB173325Ras-related protein Rab-17
RBM473325RNA binding motif protein 47
SCCPDH2525Saccharopine dehydrogenase (putative)
SEMA3F2525Sema domain, immunoglobulin domain (Ig), short basic domain, secreted, (semaphorin) 3F
SLC2A102925Solute carrier family 2 (facilitated glucose transporter), member 10
TBX32125T-box protein 3
TOX33825TOX high mobility group box family Member 3
WFS12125Wolfram syndrome 1 (wolframin)
CAMK2N12521 Calcium/calmodulin-dependent protein kinase II inhibitor 1
EFHC12121EF-hand domain (C-terminal) containing 1
FAAH2121Fatty acid amide hydrolase
GALNT72921 UDP-N-acetyl-alpha-D-galactosamine:polypeptide N-acetylgalactosaminyltransferase 7 (GalNAc-T7)
KIAA00402121Uncharacterized protein KIAA0040
LASS62921LAG1 homolog, ceramide synthase 6
LRBA2121LPS-responsive vesicle trafficking, beach and anchor containing
METRN2121Meteorin, glial cell differentiation regulator
MREG2121Melanoregulin
RAB262921RAB26, member RAS oncogene family
RNF1032121Ring finger protein 103

[i] FOXA1, forkhead box protein A1; ESR1, estrogen receptor 1.

The transcription factor ESR is overexpressed in 70% of BCs, and is a major target for endocrine therapies for luminal A BC patients (13). Dimeric ESR binds to promoter and distant enhancer regions of E2-sensitive genes to regulate their expression. The binding of FOXA1 to enhancer regions of the compact chromatin facilitates remodeling at the ESR1 binding regions (23,30,70); therefore, FOXA1 is also known as ‘pioneer’ transcription factor (20). When the 115 overlapping genes from microarrays (cut-off frequency of 5) were compared with ESR1-stimulated genes (71), ~22% of ESR1 and 17% of FOXA1 genes were represented in the overlapping, co-expressed FOXA1:ESR1 microarray gene cluster (Table VI). Furthermore, comparisons were performed only for 51 of the ESR1-upregulated genes identified by Tozlu et al (71), but these 51 genes were not classified as such if they were regulated classically or in a non-genomic manner by ESR1 protein.

Table VI.

Comparison of ESR1 and FOXA1 co-expression Oncomine™ analysis with 51 estrogen-upregulated genes reported by Tozlu et al (71).

Table VI.

Comparison of ESR1 and FOXA1 co-expression Oncomine™ analysis with 51 estrogen-upregulated genes reported by Tozlu et al (71).

ESR1Co-expression Oncomine™FOXA1Co-expression Oncomine™
ESR1+FOXA1+
CA12+ESR1+
GATA3+GATA3+
NAT1+MLPH
SLC39A6+AGR2
TBC1D9 CA12+
DNALI1 TFF3+
FOXA1+XBP1+
ANXA9 NAT1+
DNAJC12+SLC39A6+
TFF3+SPDEF
ERBB4+TSPAN1
MLPH DNAJC12+
MYB+FBP1
XBP1+GREB1
FBP1 MYB+
IL6ST+TFF1+
MAGED2 AR+
TFF1+FAM174B
ACADSB+KDM4B
PGR+ABAT
SCNN1A BCL2+
SLC7A8 IL6ST+
BCL2+TTC39A
C9orf116 ACADSB+
CACNA1D ERBB4+
STC2+CACNA1D
AR+RBM47
THSD4 STC2+
CYP2B6+AFF3
RERG+CYB5A
C6orf97 PGR+
PTPRT+GPR160
RABEP1+GSTM3
SEMA3B+INPP5J
AKR7A3 RABEP1+
CACNA2D2 SEMA3B+
RAB17 CYP2B6+
CAMK2N1 KRT18+
CCDC74B LRBA+
FAAH PTPRT+
KIAA0040 RERG+
LRBA+SLC19A2
HPN+EEF1A2
MYO5C HPN+

[i] FOXA1, forkhead box protein A1; ESR1, estrogen receptor 1.

GATA3 is required for mammary gland morphogenesis and luminal cell differentiation, and is implicated in BC metastasis and progression (38,72). Additionally, GATA3 is also closely associated with ESR1 expression status, and its expression indicates favorable BC pathological outcome (73). Since GATA3 expression together with ESR1 and FOXA1 expression correlates strongly with luminal BC subtypes (33,74), GATA3 (43) was also observed to be overlapped with the ESR1:FOXA1 gene cluster. Approximately 79 genes were co-expressed in all the three microarrays: ESR1, FOXA1 and GATA3. Notably, in both co-expression overlaps (FOXA1:ESR1 and FOXA1:ESR1:GATA3), the majority of genes were involved in signal transduction (Figs. 1C and 2A), thus suggesting a prominent role of these genes in BC tumorigenesis. Lin et al demonstrated by whole-genome microarray analysis that 137 genes were regulated by ESR1 out of the ~19,000 genes surveyed (75). However, only 89 of the 137 ESR1-regulated genes were direct targets of ESR1. When the overlapping co-expression gene clusters (FOXA1:ESR1 or FOXA1:ESR1:GATA3) were compared with the Lin et al data (74), only 8 genes were observed to be direct target genes (Table VII). One of the possible reasons for such low detection of ESR-responsive genes may be the absence of a responsive DNA element or non-genomic binding through specificity protein 1, activator protein 1 or specificity protein 3 (7678). The pie chart and Venn diagram based on pathways of overlapping co-expression cluster genes of FOXA1:ESR1 and FOXA1:ESR1:GATA3 are shown in Fig. 1A-C and Fig. 2A and B, respectively.

Table VII.

Comparison of ESR1 and forkhead box protein A1 co-expression Oncomine™ analysis with the direct targets of ESR1(39).

Table VII.

Comparison of ESR1 and forkhead box protein A1 co-expression Oncomine™ analysis with the direct targets of ESR1(39).

GenesExpression pattern
STC2
GREB1
SIAH2
PGR
IL6ST
NRIP1
ADCY9
CCNG2

[i] ESR1, estrogen receptor 1; ↑, upregulation; ↓, downregulation.

FOXA1, also known as hepatocyte nuclear factor 3α, is a member of the forkhead class of DNA-binding proteins, and is co-expressed with ESR1 in BC luminal subtype A (49,79). Importantly, it has been previously reported that FOXA1-mediated chromatin changes were not influenced by E2 treatment, but contributed to the recruitment of ESR to chromatin by creating optimal binding conditions (70). The co-expression of ESR1 and FOXA1 is also associated with the luminal subtype of breast tumors and patient survival (33). Approximately 50% of ESR-E2 responsive genes require prior FOXA1 binding for their optimal expression (32,33). As illustrated in luminal A BC cells MCF7, there is a reduced E2-dependent gene expression and proliferation during FOXA1 depletion in the cells (30,31). In addition, RNA interference-mediated depletion of FOXA1 in MCF7 cells leads to a decreased expression of the PS2, BCL2, SIAH2 and CMYB genes (25). By contrast, in the present study, ectopic FOXA1 expression was able to regulate the ESR1 target genes PS2, BCL2, PGR, SIAH2, CMYB and GATA3 in both MCF7 and T47D BC cells (Fig. 3A and B). The ectopic expression of FOXA1 is shown in Fig. 3A and C.

The secretory protein trefoil factor (TFF) 1 or PS2 is abnormally expressed in ~50% of BCs (80). In mammary carcinoma, forced PS2 expression resulted in increased cell proliferation and survival in mammary carcinoma cells with anchorage-independent growth, migration and invasion in a xenograft model (81). The present study identified that the PS2 gene co-expresses with ESR1 and FOXA1, but the molecular pathway involved is not clearly understood. Bioinformatic analysis of the PS2 promoter indicated the presence of two FOXA1 binding sites at 8 bp downstream and 132 bp upstream, respectively, of a molecularly characterized ERE site in the PS2 promoter (Fig. 4A). EMSA confirmed that FOXA1 binds to the PS2 promoter at FOXA1 site 1 (−546 to 534 nucleotide position) and FOXA1 site 2 (−390 to −378 nucleotide position) (Fig. 4B and C). To confirm the specificity of EMSA binding, cold probe (non-radioactively labeled) competition with FOXA1 consensus sequence was performed for both EMSA1 and EMSA2 sequences. With increasing concentrations of cold probe (100–150-fold) there was a clear indication of cold probe competition, as observed by the decreased protein-DNA complex (Fig. 4B). In vivo ChIP assay also confirmed FOXA1 binding in both sites using an anti-FOXA1 antibody (Fig. 4C). A similar in vitro assay for the ERE site in PS2 was not performed, as it was confirmed previously by Amiry et al (81). Notably, an enhanced recruitment of FOXA1 to its site was also observed during E2 stimulation. Subsequently, enhanced FOXA1 recruitment to the FOXA1 site also resulted in elevated levels of ESR1 recruitment to the ERE site of the PS2 gene. In addition, there was also a slight recruitment of FOXA1 to the ERE site during E2 stimulation (Fig. 5). To understand the effect of ESR1 and FOXA1 co-expression on the PS2 gene and other FOXA/ESR1 co-regulated genes, transient transfection was performed in ESR1+ T47D BC cells. PS2 along with CMYB, BCL2 and SIAH2 were significantly regulated by FOXA1, and co-transfection with ESR1 expression plasmid suggested an interaction between these genes. Importantly, the regulation was significantly enhanced during ESR1 and FOXA1 co-transfection compared with only FOXA1-transfected cells (Fig. 6). For example, the target genes CMYB, SIAH2 and PS2 were significantly upregulated upon co-transfection with ESR1/FOXA1 expression plasmids, thus suggesting a co-regulatory function of ESR1/FOXA1 on the above target genes. In the case of the PS2 gene, FOXA1 and ESR1 responsive elements were observed to be separated by ~122 nucleotides (Fig. 4A). Therefore, one of the probable reasons for enhanced PS2 transcription during FOXA1/ESR1 co-transfection may be the recruitment of ESR1 and FOXA1 to their respective responsive sites, thereby causing a synergistic effect. However, the presence of FOXA1 sites adjacent to ERE in the promoter of other target genes remains to be determined. In addition to PS2, the established target gene of ESR1, other genes such as BCL2, PGR, SIAH2 and CMYB were also detected in both the co-expression overlapping genes and in individual microarrays with ESR1 and FOXA1, which suggests the validity of the present meta-analysis.

In addition to extrapolating highly correlated overlapping genes, the present study also enabled the comparison of genes that may not always have high correlation coefficient values, and provide an advantage in clustering co-expression overlapping genes based on their pathway (Figs. 1C and 2B). In addition to the ESR-established pathway genes (GATA3, growth regulation by estrogen in breast cancer 1, TFF1, TFF3, epidermal growth factor receptor 4, MYB, PGR and BCL2), novel pathways can be proposed according to the results of the present study, including protein folding (DnaJ heat shock protein family 40 member C12), development and differentiation (neural proliferation, differentiation and control 1, anterior gradient 2, metallothionein-like 5, semaphorin 3B, actin-binding LIM protein 3, chromosome 10 open reading frame 116, T-box 3 and meteorin) and metabolism (solute carrier family 39, member 6, 4-aminobutyrate aminotransferase, elongation of very long chain fatty acids protein 5, methylcrotonoyl-CoA carboxylase 2 and cytochrome P450 2B6), which have a direct and indirect influence during tumorigenesis.

In the present study, co-expression analysis has been used to depict overlapping co-regulatory genes in known pathways; however, this analysis has certain caveats. First, the overlapping genes were clustered based on gene ontology data. Second, the clustered meta-analysis genes are only a predictive hypothesis, which requires experimental validation. Third, it may be possible that a number of true FOXA1:ESR1 pathways interacting partners are lost due to the stringency used in the analysis. However, the present analysis provides novel pathways for assessing the FOXA1:ESR1 and FOXA1:ESR1:GATA3 signaling pathway axes, particularly in breast tumorigenesis.

In conclusion, Oncomine™ co-expression meta-analysis provided a cluster of genes with definitive pathways based on stronger co-expression co-efficient analysis using different microarrays, which may be of higher significance than a single microarray. To the best of our knowledge, the present is the first study to provide insight into FOXA1:ESR1 and FOXA1:ESR1:GATA3 co-expressed genes involved in BC tumorigenesis. The microarray analysis also provides information on novel intricate pathways, including protein folding, metabolism, development and differentiation. To understand the role of these predictive pathways, a future experimental model is required to further validate the present findings.

Acknowledgements

The present study was supported by a core grant from the Institute of Life Sciences, Department of Biotechnology, Government of India (New Delhi, India) awarded to S.C. and B.M.K. The authors thank Kathi Downs (Sr. Inside Sales and Support Specialist, Compendia Bioscience; Thermo Fisher Scientific, Inc.) for providing the trail version of Oncomine™ and Dr Karen L. (American Journal Experts, LLC, Durham, NC, USA) for editing the present manuscript.

References

1 

Colditz GA: Relationship between estrogen levels, use of hormone replacement therapy, and breast cancer. J Natl Cancer Inst. 90:814–823. 1998. View Article : Google Scholar : PubMed/NCBI

2 

Nomura Y, Tashiro H, Hamada Y and Shigematsu T: Relationship between estrogen receptors and risk factors of breast cancer in Japanese pre- and postmenopausal patients. Breast Cancer Res Treat. 4:37–43. 1984. View Article : Google Scholar : PubMed/NCBI

3 

Borellini F and Oka T: Growth control and differentiation in mammary epithelial cells. Environ Health Perspect. 80:85–99. 1989. View Article : Google Scholar : PubMed/NCBI

4 

Zhang HZ, Bennett JM, Smith KT, Sunil N and Haslam SZ: Estrogen mediates mammary epithelial cell proliferation in serum-free culture indirectly via mammary stroma-derived hepatocyte growth factor. Endocrinology. 143:3427–3434. 2002. View Article : Google Scholar : PubMed/NCBI

5 

Richert MM, Schwertfeger KL, Ryder JW and Anderson SM: An atlas of mouse mammary gland development. J Mammary Gland Biol Neoplasia. 5:227–241. 2000. View Article : Google Scholar : PubMed/NCBI

6 

Clarke RB, Howell A, Potten CS and Anderson E: Dissociation between steroid receptor expression and cell proliferation in the human breast. Cancer Res. 57:4987–4991. 1997.PubMed/NCBI

7 

Russo J, Ao X, Grill C and Russo IH: Pattern of distribution of cells positive for estrogen receptor alpha and progesterone receptor in relation to proliferating cells in the mammary gland. Breast Cancer Res Treat. 53:217–227. 1999. View Article : Google Scholar : PubMed/NCBI

8 

Ali S and Coombes RC: Endocrine-responsive breast cancer and strategies for combating resistance. Nat Rev Cancer. 2:101–112. 2002. View Article : Google Scholar : PubMed/NCBI

9 

Jiang Y, Zou L, Lu WQ, Zhang Y and Shen AG: Foxo3a expression is a prognostic marker in breast cancer. PloS One. 8:e707462013. View Article : Google Scholar : PubMed/NCBI

10 

Gross JM and Yee D: How does the estrogen receptor work? Breast Cancer Res. 4:62–64. 2002. View Article : Google Scholar : PubMed/NCBI

11 

Power KA and Thompson LU: Ligand-induced regulation of ERalpha and ERbeta is indicative of human breast cancer cell proliferation. Breast Cancer Res Treat. 81:209–221. 2003. View Article : Google Scholar : PubMed/NCBI

12 

Akaogi K, Nakajima Y, Ito I, Kawasaki S, Oie SH, Murayama A, Kimura K and Yanagisawa J: KLF4 suppresses estrogen-dependent breast cancer growth by inhibiting the transcriptional activity of ERalpha. Oncogene. 28:2894–2902. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Lumachi F, Brunello A, Maruzzo M, Basso U and Basso SM: Treatment of estrogen receptor-positive breast cancer. Curr Med Chem. 20:596–604. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Breast Cancer Trialists' Collaborative Group, . Systemic treatment of early breast cancer by hormonal, cytotoxic, or immune therapy. 133 randomised trials involving 31,000 recurrences and 24,000 deaths among 75,000 women. Early Breast Cancer Trialists' Collaborative Group. Lancet. 339:71–85. 1992.PubMed/NCBI

15 

Osborne CK: Tamoxifen in the treatment of breast cancer. N Engl J Med. 339:1609–1618. 1998. View Article : Google Scholar : PubMed/NCBI

16 

Osborne CK and Schiff R: Mechanisms of endocrine resistance in breast cancer. Annu Rev Med. 62:233–247. 2011. View Article : Google Scholar : PubMed/NCBI

17 

Garcia-Becerra R, Santos N, Diaz L and Camacho J: Mechanisms of resistance to endocrine therapy in breast cancer: Focus on signaling pathways, miRNAs and genetically based resistance. Int J Mol Sci. 14:108–145. 2013. View Article : Google Scholar

18 

Kabel AM, Altalhi D, Alsharabi H, Qadi O and Ad Khan M: Tamoxifen-resistant breast cancer: Causes of resistance and possible management. Journal of Cancer Research and Treatment. 4:37–40. 2016.

19 

Lin L, Miller CT, Contreras JI, Prescott MS, Dagenais SL, Wu R, Yee J, Orringer MB, Misek DE, Hanash SM, et al: The hepatocyte nuclear factor 3 alpha gene, HNF3alpha (FOXA1), on chromosome band 14q13 is amplified and overexpressed in esophageal and lung adenocarcinomas. Cancer Res. 62:5273–5279. 2002.PubMed/NCBI

20 

Sekiya T, Muthurajan UM, Luger K, Tulin AV and Zaret KS: Nucleosome-binding affinity as a primary determinant of the nuclear mobility of the pioneer transcription factor FoxA. Genes Dev. 23:804–809. 2009. View Article : Google Scholar : PubMed/NCBI

21 

Costa RH, Grayson DR and Darnell JE Jr: Multiple hepatocyte-enriched nuclear factors function in the regulation of transthyretin and alpha 1-antitrypsin genes. Mol Cell Biol. 9:1415–1425. 1989. View Article : Google Scholar : PubMed/NCBI

22 

Tilghman SM and Belayew A: Transcriptional control of the murine albumin/alpha-fetoprotein locus during development. Proc Natl Acad Sci USA. 79:pp. 5254–5257. 1982; View Article : Google Scholar : PubMed/NCBI

23 

Kaestner KH: The hepatocyte nuclear factor 3 (HNF3 or FOXA) family in metabolism. Trends Endocrinol Metab. 11:281–285. 2000. View Article : Google Scholar : PubMed/NCBI

24 

Lee CS, Friedman JR, Fulmer JT and Kaestner KH: The initiation of liver development is dependent on Foxa transcription factors. Nature. 435:944–947. 2005. View Article : Google Scholar : PubMed/NCBI

25 

Bernardo GM and Keri RA: FOXA1: A transcription factor with parallel functions in development and cancer. Biosci Rep. 32:113–130. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Cirillo LA, McPherson CE, Bossard P, Stevens K, Cherian S, Shim EY, Clark KL, Burley SK and Zaret KS: Binding of the winged-helix transcription factor HNF3 to a linker histone site on the nucleosome. EMBO J. 17:244–254. 1998. View Article : Google Scholar : PubMed/NCBI

27 

Lupien M, Eeckhoute J, Meyer CA, Wang Q, Zhang Y, Li W, Carroll JS, Liu XS and Brown M: FoxA1 translates epigenetic signatures into enhancer-driven lineage-specific transcription. Cell. 132:958–970. 2008. View Article : Google Scholar : PubMed/NCBI

28 

Lacroix M and Leclercq G: About GATA3, HNF3A, and XBP1, three genes co-expressed with the oestrogen receptor-alpha gene (ESR1) in breast cancer. Mol Cell Endocrinol. 219:1–7. 2004. View Article : Google Scholar : PubMed/NCBI

29 

Mirosevich J, Gao N, Gupta A, Shappell SB, Jove R and Matusik RJ: Expression and role of Foxa proteins in prostate cancer. Prostate. 66:1013–1028. 2006. View Article : Google Scholar : PubMed/NCBI

30 

Carroll JS and Brown M: Estrogen receptor target gene: An evolving concept. Molecular Endocrinol. 20:1707–1714. 2006. View Article : Google Scholar

31 

Laganiere J, Deblois G, Lefebvre C, Bataille AR, Robert F and Giguère V: From the Cover: Location analysis of estrogen receptor alpha target promoters reveals that FOXA1 defines a domain of the estrogen response. Proc Natl Acad Sci USA. 102:pp. 11651–11656. 2005; View Article : Google Scholar : PubMed/NCBI

32 

Thorat MA, Marchio C, Morimiya A, Savage K, Nakshatri H, Reis-Filho JS and Badve S: Forkhead box A1 expression in breast cancer is associated with luminal subtype and good prognosis. J Clin Pathol. 61:327–332. 2008. View Article : Google Scholar : PubMed/NCBI

33 

Badve S, Turbin D, Thorat MA, Morimiya A, Nielsen TO, Perou CM, Dunn S, Huntsman DG and Nakshatri H: FOXA1 expression in breast cancer - correlation with luminal subtype A and survival. Clinical Cancer Res. 13:4415–4421. 2007. View Article : Google Scholar

34 

Burch JB: Regulation of GATA gene expression during vertebrate development. Semin Cell Dev Biol. 16:71–81. 2005. View Article : Google Scholar : PubMed/NCBI

35 

Zheng R and Blobel GA: GATA transcription factors and cancer. Genes Cancer. 1:1178–1188. 2010. View Article : Google Scholar : PubMed/NCBI

36 

Yoon NK, Maresh EL, Shen D, Elshimali Y, Apple S, Horvath S, Mah V, Bose S, Chia D, Chang HR and Goodglick L: Higher levels of GATA3 predict better survival in women with breast cancer. Hum Pathol. 41:1794–1801. 2010. View Article : Google Scholar : PubMed/NCBI

37 

Kouros-Mehr H, Slorach EM, Sternlicht MD and Werb Z: GATA3 maintains the differentiation of the luminal cell fate in the mammary gland. Cell. 127:1041–1055. 2006. View Article : Google Scholar : PubMed/NCBI

38 

Asselin-Labat ML, Sutherland KD, Barker H, Thomas R, Shackleton M, Forrest NC, Hartley L, Robb L, Grosveld FG, van der Wees J, et al: GATA3 is an essential regulator of mammary-gland morphogenesis and luminal-cell differentiation. Nat Cell Biol. 9:201–209. 2007. View Article : Google Scholar : PubMed/NCBI

39 

Schaner ME, Ross DT, Ciaravino G, Sorlie T, Troyanskaya O, Diehn M, Wang YC, Duran GE, Sikic TL, Caldeira S, et al: Gene expression patterns in ovarian carcinomas. Mol Biol Cell. 14:4376–4386. 2003. View Article : Google Scholar : PubMed/NCBI

40 

Flouriot G, Griffin C, Kenealy M, Sonntag-Buck V and Gannon F: Differentially expressed messenger RNA isoforms of the human estrogen receptor-alpha gene are generated by alternative splicing and promoter usage. Mol Endocrinol. 12:1939–1954. 1998. View Article : Google Scholar : PubMed/NCBI

41 

Albergaria A, Paredes J, Sousa B, Milanezi F, Carneiro V, Bastos J, Costa S, Vieira D, Lopes N, Lam EW, et al: Expression of FOXA1 and GATA3 in breast cancer: The prognostic significance in hormone receptor-negative tumours. Breast Cancer Res. 11:R402009. View Article : Google Scholar : PubMed/NCBI

42 

Wilson BJ and Giguère V: Identification of novel pathway partners of p68 and p72 RNA helicases through Oncomine™ meta-analysis. BMC Genomics. 8:4192007. View Article : Google Scholar : PubMed/NCBI

43 

Wilson BJ and Giguère V: Meta-analysis of human cancer microarrays reveals GATA3 is integral to the estrogen receptor alpha pathway. Mol Cancer. 7:492008. View Article : Google Scholar : PubMed/NCBI

44 

Rhodes DR, Yu J, Shanker K, Deshpande N, Varambally R, Ghosh D, Barrette T, Pandey A and Chinnaiyan AM: Oncomine™: A cancer microarray database and integrated data-mining platform. Neoplasia. 6:1–6. 2004. View Article : Google Scholar : PubMed/NCBI

45 

Higgins JPT, Wang L, Kambham N, Montgomery K, Mason V, Vogelmann SU, Lemley KV, Brown PO, Brooks JD and van de Rijn M: Gene expression in the normal adult human kidney assessed by complementary DNA microarray. Mol Biol Cell. 15:1–656. 2004.PubMed/NCBI

46 

Roth RB, Hevezi P, Lee J, Willhite D, Lechner SM, Foster AC and Zlotnik A: Gene expression analyses reveal molecular relationships among 20 regions of the human CNS. Neurogenetics. 7:67–80. 2006. View Article : Google Scholar : PubMed/NCBI

47 

Shyamsundar R, Kim YH, Higgins JP, Montgomery K, Jorden M, Sethuraman A, van de Rijn M, Botstein D, Brown PO and Pollack JR: A DNA microarray survey of gene expression in normal human tissues. Genome Biol. 6:R222005. View Article : Google Scholar : PubMed/NCBI

48 

Tabchy A, Valero V, Vidaurre T, Lluch A, Gomez H, Martin M, Qi Y, Barajas-Figueroa LJ, Souchon E, Coutant C, et al: Evaluation of a 30-gene paclitaxel, fluorouracil, doxorubicin and cyclophosphamide chemotherapy response predictor in a multicenter randomized trial in breast cancer. Clinc Cancer Res. 16:5351–5361. 2010. View Article : Google Scholar

49 

Perou CM, Sørlie T, Eisen MB, van de Rijn M, Jeffrey SS, Rees CA, Pollack JR, Ross DT, Johnsen H, Akslen LA, et al: Molecular portraits of human breast tumours. Nature. 406:747–752. 2000. View Article : Google Scholar : PubMed/NCBI

50 

Su AI, Cooke MP, Ching KA, Hakak Y, Walker JR, Wiltshire T, Orth AP, Vega RG, Sapinoso LM, Moqrich A, et al: Large-scale analysis of the human and mouse transcriptomes. Proc Natl Acad Sci USA. 99:pp. 4465–4470. 2002; View Article : Google Scholar : PubMed/NCBI

51 

Zhao H, Langerød A, Ji Y, Nesland JM, Tibshirani R, Bukholm IK, Kåresen R, Botstein D, Børresen-Dale A and Jeffrey SS: Different gene expression patterns in invasive Lobular and ductal carcinomas of the breast. Mol Biol Cell. 15:2523–2536. 2004. View Article : Google Scholar : PubMed/NCBI

52 

Yu K, Ganesan K, Miller LD and Tan P: A modular analysis of breast cancer reveals a novel low-grade molecular signature in estrogen receptor-positive tumors. Clin Cancer Res. 12:3288–3296. 2006. View Article : Google Scholar : PubMed/NCBI

53 

Wang Y, Klijn JG, Zhang Y, Sieuwerts AM, Look MP, Yang F, Talantov D, Timmermans M, Meijer-van Gelder ME, Yu J, et al: Gene-expression profiles to predict distant metastasis of lymph-node-negative primary breast cancer. Lancet. 365:671–679. 2005. View Article : Google Scholar : PubMed/NCBI

54 

Waddell N, Cocciardi S, Johnson J, Healey S, Marsh A, Riley J, da Silva L, Vargas AC, Reid L; kConFab Investigators, ; Simpson PT, Lakhani SR and Chenevix-Trench G: Gene expression profiling of formalin-fixed, paraffin-embedded familial breast tumours using the whole genome-DASL assay. J Pathol. 221:452–461. 2010.PubMed/NCBI

55 

van 't Veer LJ1, Dai H, van de Vijver MJ, He YD, Hart AA, Mao M, Peterse HL, van der Kooy K, Marton MJ, Witteveen AT, et al: Gene expression profiling predicts clinical outcome of breast cancer. Nature. 415:530–536. 2002. View Article : Google Scholar : PubMed/NCBI

56 

Schmidt M, Böhm D, von Törne C, Steiner E, Puhl A, Pilch H, Lehr HA, Hengstler JG, Kölbl H and Gehrmann M: The humoral immune system has a key prognostic impact in node-negative breast cancer. Cancer Res. 68:5405–5413. 2008. View Article : Google Scholar : PubMed/NCBI

57 

Pollack JR, Sørlie T, Perou CM, Rees CA, Jeffrey SS, Lonning PE, Tibshirani R, Botstein D, Børresen-Dale AL and Brown PO: Microarray analysis reveals a major direct role of DNA copy number alteration in the transcriptional program of human breast tumors. Proc Natl Acad Sci USA. 99:pp. 12963–12968. 2002; View Article : Google Scholar : PubMed/NCBI

58 

Minn AJ, Gupta GP, Siegel PM, Bos PD, Shu W, Giri DD, Viale A, Olshen AB, Gerald WL and Massagué J: Genes that mediate breast cancer metastasis to lung. Nature. 436:518–524. 2005. View Article : Google Scholar : PubMed/NCBI

59 

Lu X, Lu X, Wang ZC, Iglehart JD, Zhang X and Richardson AL: Predicting features of breast cancer with gene expression patterns. Breast Cancer Res Treat. 108:191–201. 2008. View Article : Google Scholar : PubMed/NCBI

60 

Korde LA, Lusa L, McShane L, Lebowitz PF, Lukes L, Camphausen K, Parker JS, Swain SM, Hunter K and Zujewski JA: Gene expression pathway analysis to predict response to neoadjuvant docetaxel and capecitabine for breast cancer. Breast Cancer Res Treat. 119:685–699. 2010. View Article : Google Scholar : PubMed/NCBI

61 

Kao KJ, Chang KM, Hsu HC and Huang AT: Correlation of microarray-based breast cancer molecular subtypes and clinical outcomes: implications for treatment optimization. BMC Cancer. 11:1432011. View Article : Google Scholar : PubMed/NCBI

62 

Julka PK, Chacko RT, Nag S, Parshad R, Nair A, Oh DS, Hu Z, Koppiker CB, Nair S, Dawar R, et al: A phase II study of sequential neoadjuvant gemcitabine plus doxorubicin followed by gemcitabine plus cisplatin in patients with operable breast cancer: prediction of response using molecular profiling. Br J Cancer. 98:1327–1335. 2008. View Article : Google Scholar : PubMed/NCBI

63 

Hatzis C, Pusztai L, Valero V, Booser DJ, Esserman L, Lluch A, Vidaurre T, Holmes F, Souchon E, Wang H, et al: A genomic predictor of response and survival following taxane-anthracycline chemotherapy for invasive breast cancer. JAMA. 305:1873–1881. 2011. View Article : Google Scholar : PubMed/NCBI

64 

Glück S, Ross JS, Royce M, McKenna EF Jr, Perou CM, Avisar E and Wu L: TP53 genomics predict higher clinical and pathologic tumor response in operable early-stage breast cancer treated with docetaxel-capecitabine ± trastuzumab. Breast Cancer Res Treat. 132:781–791. 2012. View Article : Google Scholar : PubMed/NCBI

65 

Farmer P, Bonnefoi H, Becette V, Tubiana-Hulin M, Fumoleau P, Larsimont D, Macgrogan G, Bergh J, Cameron D, Goldstein D, et al: Identification of molecular apocrine breast tumours by microarray analysis. Oncogene. 24:4660–4771. 2005. View Article : Google Scholar : PubMed/NCBI

66 

Desmedt CI, Piette F, Loi S, Wang Y, Lallemand F, Haibe-Kains B, Viale G, Delorenzi M, Zhang Y, d'Assignies MS, et al: TRANSBIG Consortium: Strong 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. 2007. View Article : Google Scholar : PubMed/NCBI

67 

Bos PD, Zhang XH, Nadal C, Shu W, Gomis RR, Nguyen DX, Minn AJ, van de Vijver MJ, Gerald WL, Foekens JA and Massagué J: Genes that mediate breast cancer metastasis to the brain. Nature. 459:1005–1009. 2009. View Article : Google Scholar : PubMed/NCBI

68 

Bonnefoi H, Potti A, Delorenzi M, Mauriac L, Campone M, Tubiana-Hulin M, Petit T, Rouanet P, Jassem J, Blot E, et al: Validation of gene signatures that predict the response of breast cancer to neoadjuvant chemotherapy: a substudy of the EORTC 10994/BIG 00–01 clinical trial. Lancet Oncol. 8:1071–1078. 2007. View Article : Google Scholar : PubMed/NCBI

69 

Livak and Schmittgen: Analysis of relative gene expression data using real-time quantitative PCR and the 2−ΔΔCT method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

70 

Jozwik KM and Carroll JS: Pioneer factors in hormone-dependent cancers. Nat Rev Cancer. 12:381–385. 2012. View Article : Google Scholar : PubMed/NCBI

71 

Tozlu S, Girault I, Vacher S, Vendrell J, Andrieu C, Spyratos F, Cohen P, Lidereau R and Bieche I: Identification of novel genes that co-cluster with estrogen receptor alpha in breast tumor biopsy specimens, using a large-scale real-time reverse transcription-PCR approach. Endocr Relat Cancer. 13:1109–1120. 2006. View Article : Google Scholar : PubMed/NCBI

72 

Cimino-Mathews A, Subhawong AP, Illei PB, Sharma R, Halushka MK, Vang R, Fetting JH, Park BH and Argani P: GATA3 expression in breast carcinoma: Utility in triple-negative, sarcomatoid, and metastatic carcinomas. Hum Pathol. 44:1341–1349. 2013. View Article : Google Scholar : PubMed/NCBI

73 

Voduc D, Cheang M and Nielsen T: GATA3 expression in breast cancer has a strong association with estrogen receptor but lacks independent prognostic value. Cancer Epidemiol Biomarkers Prev. 17:365–373. 2008. View Article : Google Scholar : PubMed/NCBI

74 

Mehra R, Varambally S, Ding L, Shen R, Sabel MS, Ghosh D, Chinnaiyan AM and Kleer CG: Identification of GATA3 as a breast cancer prognostic marker by global gene expression meta-analysis. Cancer Res. 65:11259–11264. 2005. View Article : Google Scholar : PubMed/NCBI

75 

Lin CY, Ström A, Vega VB, Kong SL, Yeo AL, Thomsen JS, Chan WC, Doray B, Bangarusamy DK, Ramasamy A, et al: Discovery of estrogen receptor alpha target genes and response elements in breast tumor cells. Genome Biol. 5:R662004. View Article : Google Scholar : PubMed/NCBI

76 

Kim K, Barhoumi R, Burghardt R and Safe S: Analysis of estrogen receptor alpha-Sp1 interactions in breast cancer cells by fluorescence resonance energy transfer. Mol Endocrinol. 19:843–854. 2005. View Article : Google Scholar : PubMed/NCBI

77 

Lambertini E, Tavanti E, Torreggiani E, Penolazzi L, Gambari R and Piva R: ERalpha and AP-1 interact in vivo with a specific sequence of the F promoter of the human ERalpha gene in osteoblasts. J Cell Physiol. 216:101–110. 2008. View Article : Google Scholar : PubMed/NCBI

78 

Li L and Davie JR: Association of Sp3 and estrogen receptor alpha with the transcriptionally active trefoil factor 1 promoter in MCF-7 breast cancer cells. J Cell Biochem. 105:365–369. 2008. View Article : Google Scholar : PubMed/NCBI

79 

Oh DS, Troester MA, Usary J, Hu Z, He X, Fan C, Wu J, Carey LA and Perou CM: Estrogen-regulated genes predict survival in hormone receptor-positive breast cancers. J Clin Oncol. 24:1656–1664. 2006. View Article : Google Scholar : PubMed/NCBI

80 

Buache E, Etique N, Alpy F, Stoll I, Muckensturm M, Reina-San-Martin B, Chenard MP, Tomasetto C and Rio MC: Deficiency in trefoil factor 1 (TFF1) increases tumorigenicity of human breast cancer cells and mammary tumor development in TFF1-knockout mice. Oncogene. 30:3261–3273. 2011. View Article : Google Scholar : PubMed/NCBI

81 

Amiry N, Kong X, Muniraj N, Kannan N, Grandison PM, Lin J, Yang Y, Vouyovitch CM, Borges S, Perry JK, et al: Trefoil factor-1 (TFF1) enhances oncogenicity of mammary carcinoma cells. Endocrinology. 150:4473–4483. 2009. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2017
Volume 14 Issue 2

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Chaudhary S, Krishna BM and Mishra SK: A novel FOXA1/ESR1 interacting pathway: A study of Oncomine™ breast cancer microarrays. Oncol Lett 14: 1247-1264, 2017
APA
Chaudhary, S., Krishna, B.M., & Mishra, S.K. (2017). A novel FOXA1/ESR1 interacting pathway: A study of Oncomine™ breast cancer microarrays. Oncology Letters, 14, 1247-1264. https://doi.org/10.3892/ol.2017.6329
MLA
Chaudhary, S., Krishna, B. M., Mishra, S. K."A novel FOXA1/ESR1 interacting pathway: A study of Oncomine™ breast cancer microarrays". Oncology Letters 14.2 (2017): 1247-1264.
Chicago
Chaudhary, S., Krishna, B. M., Mishra, S. K."A novel FOXA1/ESR1 interacting pathway: A study of Oncomine™ breast cancer microarrays". Oncology Letters 14, no. 2 (2017): 1247-1264. https://doi.org/10.3892/ol.2017.6329