Skip to main content
Endocrinology logoLink to Endocrinology
. 2008 Jun 12;149(10):4809–4820. doi: 10.1210/en.2008-0035

Transcriptional Responses to Estrogen and Progesterone in Mammary Gland Identify Networks Regulating p53 Activity

Shaolei Lu 1, Klaus A Becker 1, Mary J Hagen 1, Haoheng Yan 1, Amy L Roberts 1, Lesley A Mathews 1, Sallie S Schneider 1, Hava T Siegelmann 1, Kyle J MacBeth 1, Stephen M Tirrell 1, Jeffrey L Blanchard 1, D Joseph Jerry 1
PMCID: PMC2582927  PMID: 18556351

Abstract

Estrogen and progestins are essential for mammary growth and differentiation but also enhance the activity of the p53 tumor suppressor protein in the mammary epithelium. However, the pathways by which these hormones regulate p53 activity are unknown. Microarrays were used to profile the transcriptional changes within the mammary gland after administration of either vehicle, 17β-estradiol (E), or progesterone (P) individually and combined (EP). Treatment with EP yielded 1182 unique genes that were differentially expressed compared to the vehicle-treated group. Although 30% of genes were responsive to either E or P individually, combined treatment with both EP had a synergistic effect accounting for 60% of the differentially regulated genes. Analysis of protein-protein interactions identified p53, RelA, Snw1, and Igfals as common targets of genes regulated by EP. RelA and p53 form hubs within a network connected by genes that are regulated by EP and that may coordinate the competing functions of RelA and p53 in proliferation and survival of cells. Induction of early growth response 1 (Egr1) and Stratifin (Sfn) (also known as 14–3-3σ) by EP was confirmed by reverse transcription-quantitative PCR and shown to be p53 independent. In luciferase reporter assays, Egr1 was shown to enhance transcriptional activation by p53 and inhibit nuclear factor κB activity. These results identify a gene expression network that provides redundant activation of RelA to support proliferation as well as sensitize p53 to ensure proper surveillance and integration of their competing functions through factors such as Egr1, which both enhance p53 and inhibit RelA.


ESTROGENS and progestins induce a broad spectrum of changes within the mammary epithelium that are essential for both normal development and function. The sustained increases in levels of 17β-estradiol (E) and progesterone (P) during pregnancy induce differentiation of terminal end buds and terminal ducts (1,2,3,4,5). In addition, pregnancy levels of exogenous E and P are sufficient to render the mammary gland resistant to mammary tumorigenesis (6,7,8).

The p53 tumor suppressor pathway appears to be a critical target of hormone-mediated prevention of breast cancer. Basal levels of p53 protein are normally below the limit of detection due to its rapid degradation, but it is stabilized and accumulates rapidly after DNA damage (9,10). However, ionizing radiation induces only modest levels of p53-dependent apoptosis in the mammary epithelium of nulliparous mice (11). Radiation-induced apoptosis increases dramatically within the first 4 d of pregnancy (12) in concert with the increasing levels of proliferation stimulated by estrogens and progestins (13). Treatment with E and P for 4 d is sufficient to increase p53-dependent responses to ionizing radiation (14). The increase in p53 activity during pregnancy appears to persist in mammary epithelium of parous mice (15). The hormone-induced increase in p53 activity appears critical for parity induced protection from mammary tumors because the protective effect of parity was diminished markedly in mammary tissues from p53-deficient mice (16,17).

As the responsiveness of p53 to ionizing radiation increases rapidly after exposure to E and P (14), the transcriptional responses in the mammary gland after acute stimulation with these hormones provide a method to elucidate hormone-responsive pathways that regulate p53 function. In these experiments mice were treated with E and P, individually and combined (EP), for 4 d to define the transcriptional changes that are associated with the enhanced sensitivity of p53. Although transcriptional responses to estrogen or P alone were significant, 60% of the differentially expressed genes required combined treatment with E and P, indicating synergistic interactions between these signaling pathways. The expression profiles showed an up-regulation of genes associated with proliferation and differentiation, whereas expression of genes involved in lipid metabolism and mitochondrial respiration were diminished. Protein interaction networks identified RelA as a common target of genes induced by EP, which is consistent with the essential role of nuclear factor κB (NF-κB) in proliferation of the mammary epithelium. Despite the pronounced effects on proliferation, p53 itself was also overrepresented as a common target of genes that were up-regulated by EP. The protein interaction networks identified targets that may mediate cross talk between these pathways to balance the proliferative responses with the need to ensure genomic integrity in the mammary epithelium. Reporter assays demonstrated that early growth response 1 (Egr1) simultaneously inhibits the transcriptional activity of NF-κB while enhancing the activity of p53 in MCF-7 cells. Thus, the transcriptional profiles induced by E and P reveal redundant networks that initiate proliferation while sensitizing p53 to ensure proper genomic surveillance.

Materials and Methods

Animal husbandry and surgery

There were 17 (8 wk old) virgin BALB/c mice ovariectomized followed by a period of 1 wk to clear endogenous hormones. Hormones were administered by ip injections in a total volume of 100 μl repeated daily for 4 d, and included four animals receiving 2 μg E, four animals receiving 1 mg P, five animals receiving both E and P (EP), and four animals receiving sesame oil (vehicle). To distinguish responses in the stroma, three mice with epithelial-free fat pads [referred to as “cleared fat pads” (CFP)] were prepared by surgically removing the primary duct at 21 d of age (16,18,19) and treated with EP when mice reached 8 wk of age. Additional BALB/c animals were treated with either E, P, EP, or vehicle for confirmation of gene expression. These treatments were also administered to ovariectomized BALB/c-Trp53−/− mice to determine whether responses to hormones were p53 dependent. Lymph nodes were removed from mammary glands, and the tissue was flash frozen in liquid nitrogen. The hormones were purchased from Sigma-Aldrich (St. Louis, MO). All procedures involving animals were in accordance with institutional and national guidelines for the use of animals, and were approved by the Institutional Animal Care and Use Committee at the University of Massachusetts-Amherst.

Microarray hybridization and data normalization

The total RNA from each lymph node-free mammary gland was extracted and purified using UltraSpec (Biotecx Laboratories, Inc., Houston, TX). Of total RNA, 10 μg were biotinylated, then fragmented into approximately 35–200 bases. Fragmented, biotinylated cRNA was hybridized to the Affymetrix U74v2 mouse genome chipset using standard protocols (Affymetrix, Inc., Santa Clara, CA). Background correction and normalization of gene expression data were computed directly from the Affymetrix .CEL files using the Bioconductor package for R [R 2.4.0; (20)] implementation of GC-Robust Multichip Average (21). The resulting expression values for each sample were compared with the average expression value of the appropriate treatment group by calculating their correlation coefficients. All samples had correlations of R > 0.9 and were included in the analysis. The data discussed in this publication have been deposited in the National Center for Biotechnology Information Gene Expression Omnibus (http://www.ncbi.nlm.nih.gov/geo/) and are accessible through Gene Expression Omnibus Series accession no. GSE5483.

Data filtering

ANOVA for each of the 36,899 probe sets among all treatments was used to select those that were differentially expressed (P < 0.05). Initial analysis identified one animal in the E-treatment group (no. 3021) that formed a unique cluster and had an overrepresentation of immune-related genes suggesting a subclinical infection. Therefore, data from this animal were excluded from further analysis. ANOVA was repeated using the 16 remaining animals, and 2899 probe sets were identified as differentially expressed (P < 0.05). Hierarchical clustering was conducted using the 2899 probe sets. Pairwise comparisons of each treatment relative to the vehicle-treated group were performed on these 2899 probe sets by applying a t test. Distribution of expression values for each treatment data set was computed using 1000 permutations. Treatment with E or P individually identified differential expression of 1605 probe sets, but no difference in expression was detected in mammary glands from EP-treated mice. Because EP is necessary for the induction of p53 responsiveness, the 1294 probe sets differentially expressed in response to EP (P < 0.05) were selected for further analysis. ANOVA, t tests, and hierarchical clustering were implemented using the Institute for Genomic Research’s MeV package, version 4.0.1 (22).

Classification of differentially expressed genes

Average expression signals for each treatment group were calculated for the 1294 differentially expressed probe sets. Ratios of the treatment averages relative to the vehicle-treated group were calculated to describe the direction and fold change in expression for each probe set. The fold change identified genes that were regulated by either E alone, P alone, or required both EP. Mean log2 expression ratios for E-responsive, P-responsive, EP-responsive, and E- or P-responsive probe sets are provided in supplemental data, which are published as supplemental data on The Endocrine Society’s Journals Online web site at http://endo.endojournals.org.

Analysis of biological relationships

The 1294 probe sets that were differentially expressed in the EP treatment represent 1182 unique genes. Biological relationships among the genes that were up-regulated or down-regulated by hormone treatments were assessed. Statistical overrepresentation of Gene Ontology (GO) terms (23), Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways (24), and first-neighbor interactions among proteins (ftp://ftp.ncbi.nih.gov/gene/GeneRIF; accessed May 30, 2006) within a study set of genes was obtained using the hypergeometric distribution with a modified Bonferroni correction (e score) as implemented in GeneMerge (25). Only unique genes with a National Center for Biotechnology Information gene identification (ID) number were used in the analysis. Identifiers for the GO and KEGG analyses were unique gene IDs for Mus musculus, whereas identifiers for the first-neighbor protein interactions were unique gene IDs for Homo sapiens. The Homologene database (build 5.1) was used to convert mouse gene IDs to human gene IDs. The population to which the study sets were compared was all unique genes with a gene ID on the U74v2 chip set.

Northern analysis

Total RNA from mammary glands was isolated using UltraSpec and RNA Tack Resin (Biotecx Laboratories). Northern blotting was performed following standard procedures using 32P-labeled probes generated by random priming. Template cDNAs were amplified by RT-PCR using primers described in Table 1 and were cloned into pCR2.1-TOPO plasmids (Invitrogen Corp., Carlsbad, CA). Each template sequence was confirmed by at least two different restriction digests. The blots were scanned by the Cyclone phosphorimager system (PerkinElmer Life And Analytical Sciences, Inc., Waltham, MA) and quantified by OptiQuant software (Parkard Bioscience, Downers Grove, IL). Statistical differences between means were calculated using paired t tests.

Table 1.

Primers used for RT-PCR

Transcript Primers
Gapdh gapdh5F: 5′-TTC ACC ACC ATG GAG AAG GC-3′
gapdh3R: 5′-GGC ATG GAC TGT GGT CAT GA-3′
Areg Areg-3−5 F: 5′-CAA CTG GGC ATC TGG AAC C-3′
Areg-3–3R: 5′-GCA GAG ACC GAG ACG CT-3′
Gpx3 Gtpx-1−5 F: 5′-GGA CCA CAG TCA GCA ACG T-3′
Gtpx-1–3R: 5′-AGT GAG AGG ATA GCA TGT CCT-3′
Rbp1 Rbp-1−5 F: 5′-CAG CAA CCG TCC GGG C-3′
Rbp-1–3R: 5′-GTG GGT ATG CGT TTC GGT CC-3′
Gsn Gels-1−5 F: 5′-TTC TGT ACA ACT ACC GCC ACG-3′
Gels-1–3R: 5′-TTT TCC AAC CCA GAC AAA GAC C-3′

F, Forward; Gapdh, glyceraldehyde-3-phosphate dehydrogenase; R, reverse. 

Quantitative RT-PCR

Total RNA was extracted with QIAzol (QIAGEN, Inc., Valencia, CA) following the manufacturer’s instructions. RNA concentration and purity were determined spectrophotometrically, and RNA quality was confirmed visually on an agarose gel. The RNA was heated to 65 C for 10 min, then the reverse transcription (RT) (20 μl reactions) was performed on 2 μg RNA using Transcriptor First Strand cDNA synthesis kit (Roche, Mannheim, Germany) following the manufacturer’s instructions for transcription using both random hexamers and oligo(deoxythymidine) primers. Real-time PCR [quantitative PCR (qPCR)] was performed in duplicate using Fast Start Universal Probe Master (Roche). Each 20-μl reaction contained a final concentration of 1 μm each of forward and reverse primers, 0.2 μm probe (Universal Probe Library; Roche), 1× master mix, and 2 μl cDNA. cDNA was diluted 1:10 for Pgk1 reactions. Standard curves were run for each transcript to ensure exponential amplification, and “no RT” controls were run to exclude nonspecific amplification. Expression of genes was normalized to phosphoglycerate kinase (Pgk1) expression. Primers and probes used are listed in Table 2. The reactions were run on an MX300 real-time PCR machine (Stratagene, La Jolla, CA) with a temperature profile of 95 C for 10 min, then 45 cycles of 95 C for 15 sec and 57 C for 1 min.

Table 2.

Primers and probes used for RT-qPCR

Transcript Primers Probe
Sfn Sfn-new-left: 5′-CCA CTG GCG ATG ACA AGAA-3′ 79
Sfn-new-right: 5′-GTT GGT AGG CGG CAT CTC-3′
Egr1 Egr1-left: 5′-CCT ATG AGC ACC TGA CCA CA-3′ 22
Egr-right: 5′-TCG TTT GGC TGG GAT AAC TC-3′
Pgk1 Pgk1-left: 5′-TAC CTG CTG GCT GGA TGG-3′ 108
Pgk1-right: 5′-CAC AGC CTC GGC ATA TTT CT-3′

Luciferase reporter assays of transcriptional activity

Telomerase-immortalized breast epithelial cells, hME-CC (26), were used as the source of mRNA to obtain the full-length human Egr1 cDNA by RT-PCR. The PCR primers were 5′-GGTACCAAGCTTTCGCCGCCTGCACGCTTCTCAGTGT-3′ and 5′-CTCGAGGCGGCCGCACCCCAAGAAAAACGAAATCCAT-3′. The cDNA was cloned into the pcDNA3.1/V5-His-TOPO (Invitrogen) to generate the pcDNA-Egr1 expression plasmid. The cDNA sequence was confirmed by sequencing. The p53-Luc and pNFκB-Luc reporter plasmids were purchased from Stratagene. pRL-CMV was purchased from Promega Corp. (Madison, WI). MCF-7 cell were cultured in DMEM:F12 supplemented with 25 mm HEPES, 1.2 g/liter NaHCO3, 10 μg/ml insulin, 10% adult bovine serum, 100 U/ml penicillin, 100 μg/ml streptomycin, and 0.25 μg/ml amphotericin B. Cells were plated in 12-well plates at 300,000 cells per well 24 h before transfection. Mixtures of the pcDNA-Egr1 expression plasmid together with reporter plasmids (0.5 μg p53-Luc or pNFκB-Luc) and 0.05 μg pRL-null were transfected into MCF-7 cells using Lipofectamine 2000 (Invitrogen). At 6 h after transfection, the media were replaced with DMEM:F12 medium containing 0.1% serum. At 48 h the cells were harvested in 1× luciferase lysis buffer. Luciferase activity was determined and normalized to the pRL-null luciferase activity using the Dual Luciferase Assay Kit (Promega) to account for differences in transfection efficiency.

Results

Classification of transcriptional responses

E and P initiate a host of changes in the mammary gland with the onset of pregnancy. The transcriptional changes may simply reflect additive effects of each hormone individually but may also require more complex interactions. Hierarchical clustering provides a Euclidean distance measure and, thus, can be used to evaluate relationships among treatments. Clustering was performed on the 2899 probe sets that were differentially expressed among treatments (Fig. 1A). Although each hormonal treatment formed distinct branches, P was closely related to vehicle. Tissues from E-treated mice were distinct but remained on the same branch of the dendrogram. In contrast, tissues from mice treated with both EP formed a distinct branch.

Figure 1.

Figure 1

Gene expression patterns induced by E and P. A, Hierarchical clustering was performed using the probe sets differentially expressed among treatments. The results for probe sets with more than 4-fold difference in expression are shown. Red and green indicate more than 4-fold increase and decrease in expression levels, respectively. Mouse IDs and treatment are indicated for each sample: E, 17ß-estradiol; EP, estrogen plus P; V, vehicle. B, Classification of differentially regulated genes by hormone responses. “E-responsive” indicates genes that were increased or decreased to similar extents in both E and EP treatments. “P-responsive” indicates genes that were increased or decreased to similar extents by both P and EP treatments. “EP-responsive” indicates genes that were not altered by either hormone alone but were significantly increased or decreased when both hormones were administered. “E or P-responsive” indicates genes showing significant increase or decrease in levels when either E or P was administered. “Complex interactions” indicates a small group of genes for which expression was increased by one treatment but reversed by another treatment. C, Heat maps of average expression levels for each treatment (E, P, EP) relative to the vehicle for genes that were differentially expressed.

The genes differentially expressed by treatment with EP were of primary interest because both hormones are necessary to enhance the responsiveness of p53 (14). A total of 1294 probe sets were differentially expressed in response to treatment with both hormones (EP). The proportions of these genes that were altered by treatment with estrogen or P individually were 19 and 11%, respectively (Fig. 1B). Although similar in the proportion of genes affected, comparing the heat maps for these treatments (Fig. 1A) indicates that the fold changes in expression was much less for P compared with the responses to E. Promiscuous responses were detected for 9% of the genes with either E or P being sufficient to induce changes in expression. Complex interactions were detected for a small number of the genes (1%, Fig. 1B). In contrast, 60% of the genes required both hormones for the transcriptional responses (EP-responsive, Fig. 1B). Therefore, E and P act cooperatively to regulate a large proportion of the genes.

Steroid hormone action is mediated by transcriptional activation as well as repression mediated by their receptors. Indeed, the EP-induced changes in mRNA levels were balanced with gene expression being both enhanced and inhibited to similar extents. Among the genes classified as E responsive, treatment with E induced expression of 11% and decreased expression of 8% of the genes (Fig. 1C, panel i). Among the genes classified as P responsive, treatment with P induced expression of 5% and decreased expression of 6% of the genes (Fig. 1C, panel ii). A small group of genes was sensitive to stimulation with either hormone (Fig. 1C, panel iv) with 4% of genes up-regulated and 5% down-regulated. Therefore, treatment with E or P transcriptionally activated and repressed near equal numbers of targets. In contrast, among the EP-responsive genes, transcriptional activation was far more prevalent, with 39% showing increased expression compared with 21% that were diminished (Fig. 1C, panel iii).

The expression of genes representing E-responsive and EP-responsive classes was validated using a separate set of animals (Fig. 2). The amphiregulin (Areg) gene contains an estrogen-responsive element (ERE) and is known to be estrogen responsive in the mammary gland (27). Indeed, expression was increased by 3.2-fold by E, and similar levels were detected when both EP were administered (Fig. 2A). This represents the classical estrogen-response pathway. Because glutathione peroxidase (Gpx) had been shown previously to be p53 responsive (28), the increase in Gpx3 detected by microarray was of interest. Although not known to be regulated by estrogen, levels were increased by 1.7-fold compared with vehicle by E and EP (Fig. 2B). Among the large group of genes requiring the combined effects of EP, retinol binding protein 1 (Rbp1) was selected because its expression is increased among parous mice (29). Levels of Rbp1 mRNA were increased modestly by E but were increased 2.6-fold in mammary tissue by EP compared with vehicle-treated mice (Fig. 2C). Conversely, expression of gelsolin (Gsn) was decreased in the microarray data by EP and was confirmed by Northern blot (Fig. 2D). Treatment with EP also reduced expression of inhibitor of DNA binding 1 (Id1) by 40 and 70% in the microarray data and RT-qPCR, respectively (data not shown).

Figure 2.

Figure 2

Validation of gene expression patterns. Genes were selected representing the estrogen responsive (E-responsive) or those requiring both E and P (EP-responsive) patterns as described in Fig. 1C. Both Areg and Gpx3 are E responsive being induced to similar levels by estrogen and unaffected by the presences of P (A and B). EP-responsive genes such as Rbp1 and Gsn required both EP for induction or repression (C and D, respectively).

Because hormones were administered for 4 d, these genes include both direct targets of E and P receptors, as well as secondary targets. To define which may be direct transcriptional targets of estrogen receptors, the list of EP genes were compared with the transcriptional responses in MCF-7 cells during the first 3 h after treatment with E (30). A total of 24 genes was common to both data sets (Table 3). Of this group of 24 genes, only MYC had an ERE within the 1-kilobase promoter region. In addition to MYC, seven additional genes (ACTN4, DMPK, PFDN1, SLC7A2, VDR, HBP1, and SLC25A36) showed increased levels of mRNA in mouse mammary glands after EP treatment and bound estrogen receptor within the 1-kilobase promoter region of MCF-7 cells (Table 3). Given that only a small fraction of the EP-responsive genes in mouse mammary gland were E responsive in MCF-7 cells, it is likely that EP treatment induces expression of transcriptional activators and/or repressors that, in turn, alter expression of a much broader class of genes that are secondary targets. Actions of EP in the stroma are also likely to contribute to the modest overlap between these data sets. Overall, these results identify a large class of genes that require both E and P for their regulation, and underscore the extensive cross talk and collaboration among these hormones and their signaling pathways.

Table 3.

Estrogen receptor targets conserved targets in mouse mammary gland and human MCF-7 cells

Human gene name Human gene ID Increased at 0-3 ha Promoter EREb Chromosome Strand Transcript start Transcript end
ACTN4 81 + 19 1 43,830,166 43,913,010
BMF 90427 + 15 −1 38,167,386 38,188,367
C15ORF38 348110 + 15 −1 88,244,836 88,257,163
CCND1 595 + 11 1 69,165,054 69,178,422
CD164 8763 + 6 −1 109,794,416 109,810,340
CKS1B 1163 + 1 1 153,213,781 153,218,346
CXCL12 6387 + 10 −1 44,186,808 44,200,548
DMPK 1760 + 19 −1 50,964,818 50,977,655
DNAPTP6 26010 + 2 1 200,985,153 201,051,475
DNTTIP1 116092 + 20 1 43,853,983 43,873,471
DPP7 29952 + 9 −1 139,124,814 139,129,016
FAM100B 283991 + 17 1 71,773,003 71,778,944
FOS 2353 + 14 1 74,815,284 74,818,685
HBP1 26959 + 7 1 106,596,696 106,630,209
HMGCL 3155 + 1 −1 24,000,955 24,024,536
IKBKG 8517 + X 1 153,423,673 153,446,437
IRX2 153572 + 5 −1 2,799,880 2,804,776
JDP2 122953 + 14 1 74,968,590 75,006,908
LSR 51599 + 19 1 40,431,399 40,450,703
MYC 4609 + + 8 1 128,817,498 128,822,853
PFDN1 5201 + 5 −1 139,604,819 139,662,873
PHLDA1 22822 + 12 −1 74,705,494 74,711,823
RAB43 339122 + 3 −1 130,289,108 130,323,309
SLC25A36 55186 + 3 1 142,143,378 142,178,843
SLC38A2 54407 + 12 −1 45,038,239 45,052,824
SLC7A2 6542 + 8 1 17,440,683 17,472,296
TAF9 6880 + 5 −1 68,696,327 68,701,596
TFRC 7037 + 3 −1 197,260,553 197,293,343
TMEM64 169200 + 8 −1 91,704,778 91,727,309
VDR 7421 + 12 −1 46,521,589 46,585,081
WWC1 23286 + 5 1 167,651,670 167,829,334
a

Transcripts induced in MCF-7 cells at 3 h after treatment with E (30) and in mouse mammary glands after treatment for 4 d with EP are indicated as ″+.″ 

b

Genes for which estrogen receptor was shown to bind by chromatin immunoprecipitation (30) and were increased in mouse mammary glands after treatment for 4 d with EP are indicated as ″+.″ 

Analysis of biological relationships

Global changes in transcriptional profiles in response to EP provide insights into the alterations in cellular and metabolic pathways that regulate p53 activity in the mammary epithelium. Therefore, the entire set of genes regulated by EP was analyzed for overrepresentation of specific GO terms or KEGG pathways. The genes up-regulated by EP show strong changes associated with cellular proliferation (Table 4). GO annotations for biological processes showed changes in genes associated with DNA replication, cell cycle, and cell division. The protein synthetic capacity also was prominently up-regulated. Similarly, the cellular components, molecular functions, and KEGG pathways also reflect synthesis of nucleotides and proteins associated with proliferation. These were expected given the potent mitogenic roles of estrogen and P. In contrast, the genes down-regulated by E and P revealed metabolic changes (Table 5). Genes involved in lipid metabolism and electron transport and localized in the mitochondria were all decreased. The decrease among genes associated with the KEGG pathway for oxidative phosphorylation was especially striking. Therefore, E and P appear to promote proliferation while diminishing aerobic metabolism.

Table 4.

Analysis of biological relationships among genes up-regulated by estrogen and/or P

Functional category Term Population fraction Subset fraction e score
Biological process Protein biosynthesis 285/17730 31/687 0.0001
DNA replication 98/17730 16/687 0.0006
Cell cycle 330/17730 32/687 0.0010
Cell division 169/17730 19/687 0.0162
Ribosome biogenesis 90/17730 13/687 0.0217
Cellular component Ribonucleoprotein complex 245/17730 29/687 1.32e-05
Nucleus 3209/17730 197/687 4.14e-10
Ribosome 168/17730 22/687 8.35e-05
Chromatin 58/17730 11/687 0.0017
Cytoplasm 1081/17730 68/687 0.0075
Cytosolic ribosome 28/17730 7/687 0.0106
Chromosome 95/17730 13/687 0.0106
Chromosome, pericentric region 24/17730 6/687 0.0350
KEGG Ribosome 70/17730 16/687 1.03e-06
Cell cycle 103/17730 16/687 0.0178
PPI First-neighbor of p53 155/13627 20/604 0.0240
First-neighbor of Snw1 15/13627 6/604 0.0375
First-neighbor of RelA 78/13627 13/604 0.0496

GO terms, KEGG pathways, and first-neighbor protein interactions statistically overrepresented within the E- and/or P-up gene set (subset fraction) relative to all unique genes on the U74v2 chips (population fraction) are shown. 

Table 5.

Analysis of biological relationships among genes down-regulated by estrogen and/or P

Functional category Term Population fraction Subset fraction e score
Biological process Lipid metabolism 152/17730 15/463 0.0039
Antigen presentation 36/17730 7/463 0.0123
Glycogen biosynthesis 9/17730 4/463 0.0187
Cellular component Mitochondrion 703/17730 48/463 1.16e-07
MHC class I protein complex 26/17730 7/463 0.0003
Membrane 3520/17730 129/463 0.0016
Mitochondrial inner membrane 220/17730 18/463 0.0019
Integral to membrane 3108/17730 116/463 0.0023
Plasma membrane 429/17730 27/463 0.0024
Mitochondrial envelope 22/17730 5/463 0.0210
Molecular function MHC class I receptor activity 35/17730 8/463 0.0008
KEGG Oxidative phosphorylation 105/17730 14/463 7.39e-05
Cell adhesion molecules 206/17730 18/463 0.0010
PPAR signaling pathway 65/17730 9/463 0.0057
Type 1 diabetes mellitus 52/17730 8/463 0.0070
Antigen processing and presentation 71/17730 9/463 0.0116
PPI First-neighbor of Igfals 3/13627 3/391 0.0167

GO terms, KEGG pathways, and first-neighbor protein interactions statistically overrepresented within the E- and/or P-down gene set (subset fraction) relative to all unique genes on the U74v2 chips (population fraction) are shown. MHC, Major histocompatibility complex; PPAR, peroxisome proliferator-activated receptor. 

These changes represent the global responses to E and P but can obscure the modest changes in regulatory pathways that may determine p53 activity in mammary tissue. Furthermore, gaps in signaling pathways can result from both low levels of expression of any particular gene and events that rely on posttranslational modifications of proteins rather than transcriptional responses. Therefore, analysis of protein-protein interactions (PPIs) among genes that are differentially expressed was used to identify common targets and pathways. This approach identified an overrepresentation of genes that were first neighbors interacting with p53, Snw1, and RelA among the up-regulated genes (Table 4) and IGF binding protein (IGFBP), labile subunit (Igfals) among the down-regulated genes (Table 5). These genes form hubs within an interaction network (Fig. 3). Although E and P stimulated expression of numerous proteins that interact with RelA, Snw1, and Igfals, the expression levels of these were unchanged by the hormonal treatments. One probe set in the microarray data showed induction of p53 mRNA by E and EP. However, this does not appear to be reproducible because no difference in expression was detected by three other probe sets for p53 mRNA, and no difference in expression was detected by Northern blot (14). Because overrepresentation of proteins interacting with p53 had the highest significance level (e score = 0.0240), it suggests that EP induces a complex of genes that interact with p53 and augment its activity.

Figure 3.

Figure 3

Network of protein interactions among genes that are up-regulated by E and P. Analysis of PPIs among the genes up-regulated by E and P identified p53, RelA, and Snw1 as common targets that are statistically overrepresented, as described in Tables 4 and 5. The relationships among the genes were graphed to demonstrate interaction networks. TP53, RelA, and Snw1 form nodes but are connected by genes that are coregulated, providing pathways for cross talk. Genes connecting p53 and RelA include topoisomerase II α (TOP2a), Egr1, heat shock 70-kDa protein 8 (HSPA8), tumor protein p53 binding protein 2 (TP53BP2), and coactivator-associated arginine methyltransferase 1 (CARM1). Genes connecting RelA and Snw1 include vitamin D (1,25-dihydroxyvitamin D3) receptor (VDR), v-ski sarcoma viral oncogene homolog (SKI), jun dimerization protein 2 (JDP2), notch gene homolog 1 (NOTCH1), v-fos FBJ murine osteosarcoma viral oncogene homolog (FOS), and nuclear receptor co-repressor 2 (NCOR2). The colors indicate the relative expression level of the mRNA in EP-treated compared with the vehicle-treated controls, and range from white (no difference from control) to dark red (>3-fold increase compared with control).

Hormone-induced expression of Egr1 and Stratfinin (Sfn) in Trp53+/+ and Trp53−/− tissues

The fold changes in mRNA for p53-interacting genes and their responses to each of the treatments are summarized in Table 6. These genes represent potential effectors that are downstream of E and P. Egr1 and Sfn (also known as 14-3-3σ) were among the highest fold increases and have been implicated previously as enhancing p53 activity. Therefore, these genes were examined in greater detail. Pgk1 was used as a housekeeping gene to normalize the microarray expression data. Expression of Egr1 was increased 3-fold after treatment with EP compared with vehicle (Fig. 4A). The induction by EP was primarily via the mammary epithelium because no induction by EP was observed in epithelium-free mammary fat pads (CFP). Sfn expression was increased 2-fold over vehicle when treated with EP (Fig. 4A). Expression of Sfn was decreased significantly in the epithelium-free fat pads, suggesting that the majority of Sfn is expressed in the mammary epithelium.

Table 6.

Genes that are up-regulated by estrogen and/or P and interact directly with p53

Probe set ID Gene symbol Regulatory patterna
E plus P/sham
E P EP Expression ratio P value
98579_at EGR1 3.025 0.001
100128_at CDC2 2.787 0.001
99578_at TOP2A 2.592 0.032
96704_at SFN 2.071 0.023
102001_at RRM2 1.896 0.001
97925_at CSNK1E 1.716 0.012
104275_g_at TP53 1.621 0.001
93559_at APEX1 1.617 0.001
96696_at PRMT1 1.611 0.012
94396_at ING1 1.503 0.036
113636_at TADA3 liter 1.409 0.012
94376_s_at MRE11A 1.388 0.012
93918_at TAF9 1.312 0.018
96564_at HSPA8 1.305 0.016
97559_at EEF2 1.285 0.033
97544_at YWHAZ 1.244 0.019
106647_at RCHY1 1.217 0.016
99169_at CARM1 1.212 0.016
102934_s_at CDC25C 1.199 0.001
106238_at TP53BP2 1.181 0.044
161636_r_at PLK3 1.024 0.038

↓, Down-regulated; —, no significant change; ↑, up-regulated. 

a

Changes in levels of mRNA for the genes are indicated relative to vehicle-treated controls. 

Figure 4.

Figure 4

Egr1 and Sfn expression in mammary tissues. A, Data from the microarray studies were plotted to show the fold changes in Egr1, Sfn, and Pgk1 in mammary tissues from mice with intact mammary glands treated with vehicle or both estrogen and P (designated Intact-vehicle and Intact-EP, respectively). The expression levels in mammary fat pads that have been cleared of epithelium and treated with both E and P (designated CFP-EP) are also shown. Both Egr1 and Sfn are induced by more than 2-fold by EP in intact tissues. In the absence of the epithelium (CFP-EP), levels of Egr1 were not different from the vehicle treated, indicating that the epithelium is required for induction of Egr1 by EP. The decreased level of Sfn mRNA in CFP-EP tissues indicates that this gene is predominantly expressed in the epithelium. Levels of mRNA for the housekeeping gene, Pgk1, were unchanged across treatments. B, Responses to treatment with vehicle or EP were confirmed by RT-qPCR using an independent set of wild-type mice (Trp53+/+) as well as with p53-deficient mice (Trp53−/−). Mice were ovariectomized and administered hormones for 4 d as described in Materials and Methods. Levels of both Egr1 and Sfn were significantly increased by EP treatment. Egr1 was similarly increased by EP in Trp53−/− tissues. Basal levels of Sfn were decreased in vehicle-treated Trp53−/− tissues by 70% relative to the vehicle-treated Trp53+/+ controls but was induced 3.3-fold by EP treatment (0.3 vs. 1.0 for Trp53−/−, vehicle and Trp53−/−, EP, respectively). Expression levels were normalized to expression of Pgk1 within each animal. Means for each treatment are expressed relative to the means for the Trp53+/+, vehicle-treated controls. All comparisons are relative to the Trp53+/+, vehicle, and significant differences are indicated by *, P < 0.05 or **, P < 0.01.

Although Egr1 and Sfn are potential activators of p53, it is possible that these are targets of p53 and reflect an increase in basal p53 activity. Therefore, it was important to determine whether Egr1 and Sfn may be targets of EP and upstream of p53 or downstream targets that are reporters of p53 activity, but not key regulators of p53. Responses to EP were examined in both wild-type and p53-deficient tissues (Trp53+/+ and Trp53−/−, respectively) to ascertain whether these genes are upstream of p53 or downstream targets. The mice were ovariectomized then treated with EP for 4 d as described in Materials and Methods. Pgk1 was used to normalize levels of Egr1 and Sfn because its expression was unaffected by the treatments. The levels of Egr1 mRNA were increased by EP treatment in both the Trp53+/+ and Trp53−/− mice (2.0- and 2.3-fold, respectively). Therefore, the induction of Egr1 is not p53 dependent. Similarly, levels of Sfn mRNA were increased 1.5-fold compared with the vehicle control in Trp53+/+ mammary tissues. Although the level of Sfn mRNA was reduced dramatically in Trp53−/− mammary tissues, levels of Sfn mRNA were increased 3-fold by EP in Trp53−/− tissues (0.3 vs. 1.0 in vehicle and EP, respectively). These results suggest that p53 activity is important for the maintenance of the basal level of Sfn but that E and P act upstream of p53 to enhance its expression in both the wild-type and p53-deficient mammary tissues. These results demonstrate that Egr1 and Sfn are among a group of genes that are induced within the mammary epithelium after treatment with EP.

Cross-regulation of p53 and NF-κB transcriptional activity by Egr1

The PPIs (Fig. 3) suggest that a subset of the proteins induced by EP treatment interacts with both NF-κB and p53 networks. Egr1 was selected for further study because it has both inhibited NF-κB activity and enhanced p53 activity in other cell systems (31,32). Luciferase reporter constructs were used to examine the effects of Egr1 expression on the transcriptional activity of NF-κB and p53 in MCF-7 breast cancer cells. Expression of Egr1 resulted in a significant reduction of NF-κB activity (P < 0.001) and a simultaneous increase in p53 activity (P < 0.05). These results confirm the ability of Egr1 to regulate reciprocally the transcriptional activities of both NF-κB and p53.

Discussion

The p53 pathway responds to a variety of cellular stresses, and its induction by DNA damage has been studied extensively. Although a critical pathway for the maintenance of genomic stability, p53-dependent responses to ionizing radiation vary dramatically among tissues. Ionizing radiation induces apoptosis in the lymph nodes and intestinal crypts, whereas arrest and repair is the prevalent response in liver and kidney, while brain and muscle are nearly devoid of a p53 response (33,34). In mammary epithelium the activity of p53 is regulated by hormonal exposures and varies across developmental stages (12,14,15). In particular, treatment with E and P leads to enhanced responsiveness of p53 to ionizing radiation (14). Neither steady-state levels of p53 mRNA (14) nor basal levels of p53 protein (12) are altered by hormonal stimulation. Therefore, it appears that E and P alter cellular targets that sensitize p53 by either enhancing posttranslational modifications of p53 or promoting interactions with factors that enhance the activity of p53. Direct analyses of p53 in the mammary epithelium are hampered by the fact that the epithelium comprises only a small fraction of the total nulliparous mammary gland (<20%). Attempts to isolate the epithelium by collagenase digestion led to induction of p53 (Jerry, D. J., unpublished data). In vitro approaches are hampered by the fact that the receptors for E and P are diminished when normal mammary epithelial cells are placed in culture.

Known breast cancer susceptibility genes provide candidates that may be subject to hormonal stimuli and regulate p53 activity. Among the 10 recognized breast cancer susceptibility genes (35), ATM and CHK2 are significant because they regulate p53 activity by inducing posttranslational modifications. Similarly, polymorphisms that increase MDM2 levels and promote degradation of p53 protein have been linked to increased risk of breast cancer (36,37). However, the expression microarray analysis failed to detect changes in these factors in a pattern consistent with the hormone-induced increase in p53 activity. Similarly, none of the recognized breast cancer susceptibility genes has been shown to be changed in parous mammary tissues (29,38,39,40). Therefore, it appears that treatment with E and P engages alternate pathways to sensitize p53 in the mammary epithelium.

The transcriptional responses in the mammary gland detected by the microarrays demonstrate the breadth of alterations induced by E and P. Although the magnitude of the effect was expected, comparisons of the transcriptional changes in mammary tissue with those detected in MCF-7 cells resulted in only 24 genes that were shared in both data sets (Table 3). This underscores the many distinctions between in vitro and in vivo conditions. Analysis of the biological relationships among EP-induced genes provided further insights into the responses within the tissue (Tables 4 and 5). Mapping the EP-responsive genes onto first-neighbor PPIs provided insights into the networks that are altered by the hormones. The overrepresentation of proteins that interact with p53 identified a broad complement of genes, suggesting that redundant mechanisms are present to ensure proper regulation of p53. The induction of Egr1 and Sfn expression after EP treatment was confirmed by RT-qPCR and was shown to be p53 independent (Fig. 4). Thus, both of these factors are responsive to EP and represent potent activators of p53 that can contribute to the sensitization of p53 to enhance responsiveness to DNA damage (41,42,43,44,45,46).

Among the genes up-regulated by EP, there was also a significant overrepresentation of transcriptional targets interacting with RelA, some of which can also influence the activities of p53. RelA contains a transactivation domain and is a subunit of the NF-κB transcription factor. NF-κB and p53 can cooperate to enhance apoptosis in some settings (47,48,49) but serve antagonistic roles in most instances. Cross-regulatory mechanisms of p53 and NF-κB subunits have been demonstrated on promoters of target genes, including p21, DR5, and PUMA (50). This cross-regulation appears to be via a competition for coregulatory factors such as p300 and CRE binding protein (50,51). NF-κB complexes increase during pregnancy (52). The induction of NF-κB during pregnancy appears to be important for expansion of the mammary epithelium because blocking the pathways by knockout of CHUK (conserved helix-loop-helix ubiquitous kinase, also known as IKKα) resulted in a failure of mammary gland development (53,54). In addition to promoting proliferation of the normal mammary epithelium, NF-κB appears to play a prominent role in inducing mammary tumors in mice (55,56) and is constitutively activated in a majority of human breast cancers, as well as promoting epithelial-mesenchymal transformation (57,58). NF-κB pathways are also induced during involution of the mammary gland, a period characterized by rapid apoptosis (59,60). However, NF-κB expression appears to be restricted to the cells that are surviving involution (61). Thus, the overrepresentation of RelA-associated protein appears to reflect the proliferative responses induced by EP treatment, rather than regulating p53 activity.

Although the activities of p53 and RelA appear to be antagonistic, the interaction network (Fig. 3) provides insights into how cross talk between p53 and NF-κB is integrated to balance the need for proliferation and genomic surveillance. Both p53 and RelA bind Egr1, heat shock 70-kDa protein 8, tumor protein p53 binding protein 2, and coactivator-associated arginine methyltransferase 1. Of these, Egr1 may be especially important in integrating the responses to EP and DNA damage. EP treatment promotes expression and activity of NF-κB (52) to allow proliferation of the mammary epithelium. However, Egr1 expression is also enhanced, which can bind and temper transcriptional activation mediated by RelA (31). Expression of Egr1 is further enhanced in response to DNA damage (62), leading to increased p53 activity. Egr1 can interact with multiple tumor suppressors and, thus, may have tumor suppressive activities that extend beyond sensitizing p53 (32). The reciprocal actions of Egr1 on transactivation by p53 and NF-κB were demonstrated directly in breast epithelial cells (Fig. 5). The increase in Sfn serves to reinforce and amplify the responsiveness of p53. Together, Egr1 and Sfn can collaborate to increase the activity of p53 that competes for the pool of transcriptional coactivators, and, thus, can limit the effects of NF-κB and sway the balance of responses toward p53-dependent apoptosis when cells encounter DNA damage.

Figure 5.

Figure 5

Effect of Egr1 expression on transcriptional activity of p53 and NF-κB. MCF-7 cells were transfected with the control plasmid (pcDNA vector) or an Egr1 expression plasmid (pcDNA-Egr1) together with luciferase reporter plasmids with responsive elements for either NF-κB (A) or p53 (B). The mean activity is shown ± sem for samples run in triplicate. The differences in reporter activity were significant for both NF-κB (P < 0.001) and p53 (P < 0.05) using a one-tailed t test.

Additional signaling pathways may collaborate with p53 and NF-κB to enhance the responsiveness of p53. Rbp1 was among the genes induced by EP treatments. Because retinoids have reduced the risk of breast cancer and are being examined in breast cancer chemoprevention trials (63), this would suggest a relevant pathway. Furthermore, retinoids enhanced p53 responses to ionizing radiation in mouse mammary tissues to an extent that was similar to that observed for EP (64). Retinoids enhance secretion of TGF-β in MCF-7 cells (65), and TGF-β influences p53 activity in mammary tissue (14). Expression of TGF-β is induced by ovarian hormones (66), and both Rbp1 and TGF-β3 are persistently elevated in mammary tissue from parous mice (29). Conversely, Igfals was identified as being overrepresented in the EP-treated tissues because of decreased expression of IGF-I, IGFBP-3, and IGFBP-5. Reductions in IGF-I are associated with a reduced risk of mammary tumors in women (67) as well as in rodents (29). IGF-I levels are persistently decreased in mammary tissue from parous rodents (68), and elevated IGF-I can overcome the protective effect of parity with respect to mammary tumors (69). Because IGF-I can antagonize p53 (70), the decrease in this pathway would likely also enhance p53 activity. Therefore, increases in Rbp1 and TGF-β signaling together with decreases in IGF-I appear to be targets of EP treatment in the mammary gland that can persistently enhance the activity of p53 in parous individuals.

The transcriptional profiles provide an integrated view of the responses to estrogen and P in the mammary gland, and establish a framework with which to understand the complex signaling underlying the balance between proliferation and the need for genomic surveillance. Combined treatment with E and P engage signaling pathways that enhance expression of a variety of factors that interact with p53 and RelA. The increased expression of Egr1 is significant because it can coordinate responses to cellular stresses and proliferative signals by enhancing p53 and diminishing NF-κB transcriptional activities. In addition, expression of Rbp1 was increased, and expression of IGF-I-related proteins was decreased. Together, these genes and pathways identify redundant mechanisms by which estrogen and P regulate p53 activity in the mammary epithelium and render it resistant to tumors. The results also provide targets for chemoprevention as well as surrogate endpoints with which to evaluate therapeutic agents.

Supplementary Material

[Supplemental Data]

Acknowledgments

We thank Brooke Bentley for technical assistance with immunohistochemical staining and Ellen Dickinson for providing the BALB/c-Trp53−/− mice used in these experiments. Initial analysis of the microarray data were also aided by Oscar Loureiro and Anyuan Guo.

Footnotes

This work was supported by grants from the National Institutes of Health (CA87531, CA095164, and CA105459 ES015739) (to D.J.J.), the Massachusetts Department of Public Health (43088PPP1017), and the Charlotte Geyer Foundation with additional support from the Cooperative State Research Extension, Education Service, United States Department of Agriculture, Massachusetts Agricultural Experiment Station, and Department of Veterinary and Animal Sciences under Project Nos. MAS00821 and NC-1010.

Present address for S.L. and K.A.B.: University of Massachusetts Medical School, 364 Plantation Street, Worcester, Massachusetts 01605.

Present address for K.J.M.: Celgene, 1700 Owens Street, San Francisco, California 94158.

Disclosure Summary: S.L., K.A.B., M.J.H., H.Y., A.L.R., L.A.M., S.S.S., H.T.S., J.L.B., and D.J.J. have nothing to disclose. K.J.M. was employed by Millennium Pharmaceuticals Corp. and is presently employed by Celgene. S.M.T. is employed presently by Millennium Pharmaceuticals Corp.

First Published Online June 12, 2008

Abbreviations: CFP, Cleared fat pad; E, 17β-estradiol; Egr1, early growth response 1; EP, 17β-estradiol and progesterone; ERE, estrogen-responsive element; GO, Gene Ontology; Gpx, glutathione peroxidase; ID, identification; IGFBP, IGF binding protein; KEGG, Kyoto Encyclopedia of Genes and Genomes; NF-κB, nuclear factor κB; P, progesterone; PPI, protein-protein interaction; qPCR, quantitative PCR; Rbp1, retinol binding protein 1; RT, reverse transcription.

References

  1. Russo J, Russo IH 1993 Development pattern of human breast and susceptibility to carcinogenesis. Eur J Cancer Prev 2(Suppl 3):85–100 [PubMed] [Google Scholar]
  2. Brisken C, Park S, Vass T, Lydon JP, O'Malley BW, Weinberg RA 1998 A paracrine role for the epithelial progesterone receptor in mammary gland development. Proc Natl Acad Sci USA 95:5076–5081 [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Bocchinfuso WP, Lindzey JK, Hewitt SC, Clark JA, Myers PH, Cooper R, Korach KS 2000 Induction of mammary gland development in estrogen receptor-α knockout mice. Endocrinology 141:2982–2994 [DOI] [PubMed] [Google Scholar]
  4. Ismail PM, Amato P, Soyal SM, DeMayo FJ, Conneely OM, O'Malley BW, Lydon JP 2003 Progesterone involvement in breast development and tumorigenesis–as revealed by progesterone receptor “knockout” and “knockin” mouse models. Steroids 68:779–787 [DOI] [PubMed] [Google Scholar]
  5. Feng Y, Manka D, Wagner KU, Khan SA 2007 Estrogen receptor-α expression in the mammary epithelium is required for ductal and alveolar morphogenesis in mice. Proc Natl Acad Sci USA 104:14718–14723 [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Grubbs CJ, Farnell DR, Hill DL, McDonough KC 1985 Chemoprevention of N-nitroso-N-methylurea-induced mammary cancers by pretreatment with 17 β-estradiol and progesterone. J Natl Cancer Inst 74:927–931 [PubMed] [Google Scholar]
  7. Sivaraman L, Stephens LC, Markaverich BM, Clark JA, Krnacik S, Conneely OM, O'Malley BW, Medina D 1998 Hormone-induced refractoriness to mammary carcinogenesis in Wistar- Furth rats. Carcinogenesis 19:1573–1581 [DOI] [PubMed] [Google Scholar]
  8. Rajkumar L, Guzman RC, Yang J, Thordarson G, Talamantes F, Nandi S 2001 Short-term exposure to pregnancy levels of estrogen prevents mammary carcinogenesis. Proc Natl Acad Sci USA 98:11755–11759 [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Ashcroft M, Kubbutat MH, Vousden KH 1999 Regulation of p53 function and stability by phosphorylation. Mol Cell Biol 19:1751–1758 [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Ashcroft M, Vousden KH 1999 Regulation of p53 stability. Oncogene 18:7637–7643 [DOI] [PubMed] [Google Scholar]
  11. Kuperwasser C, Pinkas J, Hurlbut GD, Naber SP, Jerry DJ 2000 Cytoplasmic sequestration and functional repression of p53 in the mammary epithelium is reversed by hormonal treatment. Cancer Res 60:2723–2729 [PubMed] [Google Scholar]
  12. Minter LM, Dickinson ES, Naber SP, Jerry DJ 2002 Epithelial cell cycling predicts p53 responsiveness to γ-irradiation during post-natal mammary gland development. Development 129:2997-3008 [DOI] [PubMed] [Google Scholar]
  13. Traurig HH 1967 A radioautographic study of cell proliferation in the mammary gland of the pregnant mouse. Anat Rec 159:239–248 [DOI] [PubMed] [Google Scholar]
  14. Becker KA, Lu S, Dickinson ES, Dunphy KA, Mathews L, Schneider SS, Jerry DJ 2005 Estrogen and progesterone regulate radiation-induced p53 activity in mammary epithelium through TGF-β-dependent pathways. Oncogene 24:6345–6353 [DOI] [PubMed] [Google Scholar]
  15. Sivaraman L, Conneely OM, Medina D, O'Malley BW 2001 p53 is a potential mediator of pregnancy and hormone-induced resistance to mammary carcinogenesis. Proc Natl Acad Sci USA 98:12379–12384 [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Jerry DJ, Kittrell FS, Kuperwasser C, Laucirica R, Dickinson ES, Bonilla PJ, Butel JS, Medina D 2000 A mammary-specific model demonstrates the role of the p53 tumor suppressor gene in tumor development. Oncogene 19:1052–1058 [DOI] [PubMed] [Google Scholar]
  17. Medina D, Kittrell FS 2003 p53 function is required for hormone-mediated protection of mouse mammary tumorigenesis. Cancer Res 63:6140–6143 [PubMed] [Google Scholar]
  18. Medina D 1973 Preneoplastic lesions in mouse mammary tumorigenesis. Meth Cancer Res 7:3–53 [Google Scholar]
  19. DeOme KB, Medina D 1969 A new approach to mammary tumorigenesis in rodents. Cancer 24:1255–1258 [DOI] [PubMed] [Google Scholar]
  20. Ihaka R, Gentleman R 1996 R: a language for data analysis and graphics. J Comput Graph Stat 3:299-314 [Google Scholar]
  21. Zhijin W, Irizarry RA, Gentleman R, Marinez-Murillo F, Spencer F 2004 A model-based background adjustment for oligonuceotide expression arrays. J Am Stat Assoc 99:909–917 [Google Scholar]
  22. Saeed AI, Sharov V, White J, Li J, Liang W, Bhagabati N, Braisted J, Klapa M, Currier T, Thiagarajan M, Sturn A, Snuffin M, Rezantsev A, Popov D, Ryltsov A, Kostukovich E, Borisovsky I, Liu Z, Vinsavich A, Trush V, Quackenbush J 2003 TM4: a free, open-source system for microarray data management and analysis. Biotechniques 34:374-378 [DOI] [PubMed] [Google Scholar]
  23. Gene Ontology Consortium 2006 The Gene Ontology (GO) project in 2006. Nucleic Acids Res 34:D322–D326 [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Kanehisa M, Goto S, Hattori M, Aoki-Kinoshita KF, Itoh M, Kawashima S, Katayama T, Araki M, Hirakawa M 2006 From genomics to chemical genomics: new developments in KEGG. Nucleic Acids Res 34:D354–D357 [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Castillo-Davis CI, Hartl DL 2003 GeneMerge—post-genomic analysis, data mining, and hypothesis testing. Bioinformatics 19:891–892 [DOI] [PubMed] [Google Scholar]
  26. Troester MA, Hoadley KA, Sorlie T, Herbert BS, Borresen-Dale AL, Lonning PE, Shay JW, Kaufmann WK, Perou CM 2004 Cell-type-specific responses to chemotherapeutics in breast cancer. Cancer Res 64:4218–4226 [DOI] [PubMed] [Google Scholar]
  27. McBryan J, Howlin J, Kenny PA, Shioda T, Martin F 2007 ERα-CITED1 co-regulated genes expressed during pubertal mammary gland development: implications for breast cancer prognosis. Oncogene 26:6406–6419 [DOI] [PubMed] [Google Scholar]
  28. Tan M, Li S, Swaroop M, Guan K, Oberley LW, Sun Y 1999 Transcriptional activation of the human glutathione peroxidase promoter by p53. J Biol Chem 274:12061–12066 [DOI] [PubMed] [Google Scholar]
  29. D'Cruz CM, Moody SE, Master SR, Hartman JL, Keiper EA, Imielinski MB, Cox JD, Wang JY, Ha SI, Keister BA, Chodosh LA 2002 Persistent parity-induced changes in growth factors, TGF-β3, and differentiation in the rodent mammary gland. Mol Endocrinol 16:2034–2051 [DOI] [PubMed] [Google Scholar]
  30. Carroll JS, Meyer CA, Song J, Li W, Geistlinger TR, Eeckhoute J, Brodsky AS, Keeton EK, Fertuck KC, Hall GF, Wang Q, Bekiranov S, Sementchenko V, Fox EA, Silver PA, Gingeras TR, Liu XS, Brown M 2006 Genome-wide analysis of estrogen receptor binding sites. Nat Genet 38:1289–1297 [DOI] [PubMed] [Google Scholar]
  31. Chapman NR, Perkins ND 2000 Inhibition of the RelA(p65) NF-κB subunit by Egr-1. J Biol Chem 275:4719–4725 [DOI] [PubMed] [Google Scholar]
  32. Baron V, Adamson ED, Calogero A, Ragona G, Mercola D 2006 The transcription factor Egr1 is a direct regulator of multiple tumor suppressors including TGFβ1, PTEN, p53, and fibronectin. Cancer Gene Ther 13:115–124 [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. MacCallum DE, Hupp TR, Midgley CA, Stuart D, Campbell SJ, Harper A, Walsh FS, Wright EG, Balmain A, Lane DP, Hall PA 1996 The p53 response to ionising radiation in adult and developing murine tissues. Oncogene 13:2575–2587 [PubMed] [Google Scholar]
  34. Midgley CA, Owens B, Briscoe CV, Thomas DB, Lane DP, Hall PA 1995 Coupling between γ irradiation, p53 induction and the apoptotic response depends upon cell type in vivo. J Cell Sci 108:1843–1848 [DOI] [PubMed] [Google Scholar]
  35. Walsh T, King MC 2007 Ten genes for inherited breast cancer. Cancer Cell 11:103–105 [DOI] [PubMed] [Google Scholar]
  36. Bond GL, Hu W, Bond EE, Robins H, Lutzker SG, Arva NC, Bargonetti J, Bartel F, Taubert H, Wuerl P, Onel K, Yip L, Hwang SJ, Strong LC, Lozano G, Levine AJ 2004 A single nucleotide polymorphism in the MDM2 promoter attenuates the p53 tumor suppressor pathway and accelerates tumor formation in humans. Cell 119:591–602 [DOI] [PubMed] [Google Scholar]
  37. Bond GL, Hirshfield KM, Kirchhoff T, Alexe G, Bond EE, Robins H, Bartel F, Taubert H, Wuerl P, Hait W, Toppmeyer D, Offit K, Levine AJ 2006 MDM2 SNP309 accelerates tumor formation in a gender-specific and hormone-dependent manner. Cancer Res 66:5104–5110 [DOI] [PubMed] [Google Scholar]
  38. Balogh GA, Heulings R, Mailo DA, Russo PA, Sheriff F, Russo IH, Moral R, Russo J 2006 Genomic signature induced by pregnancy in the human breast. Int J Oncol 28:399–410 [PubMed] [Google Scholar]
  39. Blakely CM, Stoddard AJ, Belka GK, Dugan KD, Notarfrancesco KL, Moody SE, D'Cruz CM, Chodosh LA 2006 Hormone-induced protection against mammary tumorigenesis is conserved in multiple rat strains and identifies a core gene expression signature induced by pregnancy. Cancer Res 66:6421–6431 [DOI] [PubMed] [Google Scholar]
  40. Uehara N, Unami A, Kiyozuka Y, Shikata N, Oishi Y, Tsubura A 2006 Parous mammary glands exhibit distinct alterations in gene expression and proliferation responsiveness to carcinogenic stimuli in Lewis rats. Oncol Rep 15:903–911 [PubMed] [Google Scholar]
  41. Krones-Herzig A, Adamson E, Mercola D 2003 Early growth response 1 protein, an upstream gatekeeper of the p53 tumor suppressor, controls replicative senescence. Proc Natl Acad Sci USA 100:3233-3238 [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Krones-Herzig A, Mittal S, Yule K, Liang H, English C, Urcis R, Soni T, Adamson ED, Mercola D 2005 Early growth response 1 acts as a tumor suppressor in vivo and in vitro via regulation of p53. Cancer Res 65: 5133–5143 [DOI] [PubMed] [Google Scholar]
  43. Nair P, Muthukkumar S, Sells SF, Han SS, Sukhatme VP, Rangnekar VM 1997 Early growth response-1-dependent apoptosis is mediated by p53. J Biol Chem 272:20131–20138 [DOI] [PubMed] [Google Scholar]
  44. Stavridi ES, Chehab NH, Malikzay A, Halazonetis TD 2001 Substitutions that compromise the ionizing radiation-induced association of p53 with 14-3-3 proteins also compromise the ability of p53 to induce cell cycle arrest. Cancer Res 61:7030–7033 [PubMed] [Google Scholar]
  45. Waterman MJ, Stavridi ES, Waterman JL, Halazonetis TD 1998 ATM-dependent activation of p53 involves dephosphorylation and association with 14-3-3 proteins. Nat Genet 19:175–178 [DOI] [PubMed] [Google Scholar]
  46. Yang HY, Wen YY, Chen CH, Lozano G, Lee MH 2003 14-3-3 σ positively regulates p53 and suppresses tumor growth. Mol Cell Biol 23:7096–7107 [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Fujioka S, Schmidt C, Sclabas GM, Li Z, Pelicano H, Peng B, Yao A, Niu J, Zhang W, Evans DB, Abbruzzese JL, Huang P, Chiao PJ 2004 Stabilization of p53 is a novel mechanism for proapoptotic function of NF-κB. J Biol Chem 279:27549–27559 [DOI] [PubMed] [Google Scholar]
  48. Benoit V, Hellin AC, Huygen S, Gielen J, Bours V, Merville MP 2000 Additive effect between NF-κB subunits and p53 protein for transcriptional activation of human p53 promoter. Oncogene 19:4787–4794 [DOI] [PubMed] [Google Scholar]
  49. Bohuslav J, Chen LF, Kwon H, Mu Y, Greene WC 2004 p53 induces NF-κB activation by an IκB kinase-independent mechanism involving phosphorylation of p65 by ribosomal S6 kinase 1. J Biol Chem 279:26115–26125 [DOI] [PubMed] [Google Scholar]
  50. Schumm K, Rocha S, Caamano J, Perkins ND 2006 Regulation of p53 tumour suppressor target gene expression by the p52 NF-κB subunit. EMBO J 25:4820–4832 [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Huang WC, Ju TK, Hung MC, Chen CC 2007 Phosphorylation of CBP by IKKα promotes cell growth by switching the binding preference of CBP from p53 to NF-κB. Mol Cell 26:75–87 [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Brantley DM, Yull FE, Muraoka RS, Hicks DJ, Cook CM, Kerr LD 2000 Dynamic expression and activity of NF-κB during post-natal mammary gland morphogenesis. Mech Dev 97:149–155 [DOI] [PubMed] [Google Scholar]
  53. Cao Y, Bonizzi G, Seagroves TN, Greten FR, Johnson R, Schmidt EV, Karin M 2001 IKKα provides an essential link between RANK signaling and cyclin D1 expression during mammary gland development. Cell 107:763–775 [DOI] [PubMed] [Google Scholar]
  54. Demicco EG, Kavanagh KT, Romieu-Mourez R, Wang X, Shin SR, Landesman-Bollag E, Seldin DC, Sonenshein GE 2005 RelB/p52 NF-κB complexes rescue an early delay in mammary gland development in transgenic mice with targeted superrepressor IκB-α expression and promote carcinogenesis of the mammary gland. Mol Cell Biol 25:10136–10147 [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Cao Y, Luo JL, Karin M 2007 IκB kinase α kinase activity is required for self-renewal of ErbB2/Her2-transformed mammary tumor-initiating cells. Proc Natl Acad Sci USA 104:15852–15857 [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Kim DW, Sovak MA, Zanieski G, Nonet G, Romieu-Mourez R, Lau AW, Hafer LJ, Yaswen P, Stampfer M, Rogers AE, Russo J, Sonenshein GE 2000 Activation of NF-κB/Rel occurs early during neoplastic transformation of mammary cells. Carcinogenesis 21:871–879 [DOI] [PubMed] [Google Scholar]
  57. Sovak MA, Bellas RE, Kim DW, Zanieski GJ, Rogers AE, Traish AM, Sonenshein GE 1997 Aberrant nuclear factor-κB/Rel expression and the pathogenesis of breast cancer. J Clin Invest 100:2952–2960 [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Hu Y, Sun H, Drake J, Kittrell F, Abba MC, Deng L, Gaddis S, Sahin A, Baggerly K, Medina D, Aldaz CM 2004 From mice to humans: identification of commonly deregulated genes in mammary cancer via comparative SAGE studies. Cancer Res 64:7748–7755 [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Clarkson RW, Wayland MT, Lee J, Freeman T, Watson CJ 2004 Gene expression profiling of mammary gland development reveals putative roles for death receptors and immune mediators in post-lactational regression. Breast Cancer Res 6:R92–R109 [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Stein T, Morris JS, Davies CR, Weber-Hall SJ, Duffy MA, Heath VJ, Bell AK, Ferrier RK, Sandilands GP, Gusterson BA 2004 Involution of the mouse mammary gland is associated with an immune cascade and an acute-phase response, involving LBP, CD14 and STAT3. Breast Cancer Res 6:R75–R91 [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Clarkson RW, Heeley JL, Chapman R, Aillet F, Hay RT, Wyllie A, Watson CJ 2000 NF-κB inhibits apoptosis in murine mammary epithelia. J Biol Chem 275:12737–12742 [DOI] [PubMed] [Google Scholar]
  62. Datta R, Rubin E, Sukhatme V, Qureshi S, Hallahan D, Weichselbaum RR, Kufe DW 1992 Ionizing radiation activates transcription of the EGR1 gene via CArG elements. Proc Natl Acad Sci USA 89:10149–10153 [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Zanardi S, Serrano D, Argusti A, Barile M, Puntoni M, Decensi A 2006 Clinical trials with retinoids for breast cancer chemoprevention. Endocr Relat Cancer 13:51–68 [DOI] [PubMed] [Google Scholar]
  64. Tu Y, Jerry DJ, Pazik B, Smith SS 2005 Sensitivity to DNA damage is a common component of hormone-based strategies for protection of the mammary gland. Mol Cancer Res 3:435–442 [DOI] [PubMed] [Google Scholar]
  65. Wang Y, He QY, Chen H, Chiu JF 2007 Synergistic effects of retinoic acid and tamoxifen on human breast cancer cells: proteomic characterization. Exp Cell Res 313:357-368 [DOI] [PubMed] [Google Scholar]
  66. Ewan KB, Shyamala G, Ravani SA, Tang Y, Akhurst R, Wakefield L, Barcellos-Hoff MH 2002 Latent transforming growth factor-β activation in mammary gland: regulation by ovarian hormones affects ductal and alveolar proliferation. Am J Pathol 160:2081–2093 [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Lukanova A, Toniolo P, Zeleniuch-Jacquotte A, Grankvist K, Wulff M, Arslan AA, Afanasyeva Y, Johansson R, Lenner P, Hallmans G, Wadell G, Lundin E 2006 Insulin-like growth factor I in pregnancy and maternal risk of breast cancer. Cancer Epidemiol Biomarkers Prev 15:2489–2493 [DOI] [PubMed] [Google Scholar]
  68. Wu Y, Cui K, Miyoshi K, Hennighausen L, Green JE, Setser J, LeRoith D, Yakar S 2003 Reduced circulating insulin-like growth factor I levels delay the onset of chemically and genetically induced mammary tumors. Cancer Res 63:4384–4388 [PubMed] [Google Scholar]
  69. Thordarson G, Slusher N, Leong H, Ochoa D, Rajkumar L, Guzman R, Nandi S, Talamantes F 2004 Insulin-like growth factor (IGF)-I obliterates the pregnancy-associated protection against mammary carcinogenesis in rats: evidence that IGF-I enhances cancer progression through estrogen receptor-α activation via the mitogen-activated protein kinase pathway. Breast Cancer Res 6:R423–R436 [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Mathews L, Schneider SS 2008 Insulin-like growth factor-I inhibits growth regulatory responses engaged by estrogen and progesterone in the mouse mammary gland. Eur J Cancer Prev 17:297-305 [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

[Supplemental Data]

Articles from Endocrinology are provided here courtesy of The Endocrine Society

RESOURCES