Skip to main content
Neoplasia (New York, N.Y.) logoLink to Neoplasia (New York, N.Y.)
. 2008 Jan;10(1):20–40. doi: 10.1593/neo.07889

Gene Expression Profiling Identifies Lobe-Specific and Common Disruptions of Multiple Gene Networks in Testosterone-Supported, 17β-Estradiol- or Diethylstilbestrol-Induced Prostate Dysplasia in Noble Rats12

Neville N C Tam *, Carol Ying-Ying Szeto *, Maureen A Sartor , Mario Medvedovic , Shuk-Mei Ho *
PMCID: PMC2216049  PMID: 18231636

Abstract

The xenoestrogen diethylstilbestrol (DES) is commonly believed to mimic the action of the natural estrogen 17β-estradiol (E2). To determine if these two estrogens exert similar actions in prostate carcinogenesis, we elevated circulating levels of estrogen in Noble (NBL) rats with E2/DES-filled implants, while maintaining physiological levels of testosterone (T) in the animals with T-filled implants. The two estrogens induced dysplasia in a lobe-specific manner, with E2 targeting only the lateral prostate (LP) and DES impacting only the ventral prostate (VP). Gene expression profiling identified distinct and common E2-disrupted versus DES-disrupted gene networks in each lobe. More importantly, hierarchical clustering analyses revealed that T + E2 treatment primarily affected the gene expression pattern in the LP, whereas T + DES treatment primarily affected the gene expression profile in the VP. Gene ontology analyses and pathway mapping suggest that the two hormone treatments disrupt unique and/or common cellular processes, including cell development, proliferation, motility, apoptosis, and estrogen signaling, which may be linked to dysplasia development in the rat prostate. These findings suggest that the effects of xenoestrogens and natural estrogens on the rat prostate are more divergent than previously suspected and that these differences may explain the lobe-specific carcinogenic actions of the hormones.

Introduction

Androgens are essential to the growth and functioning of the normal prostate and undoubtedly play a key role in prostate carcinogenesis [1]. Emerging evidence shows that estrogens also have a critical role in prostatic diseases, including prostate cancer (PCa) [2,3]. The incidence of PCa increases dramatically with age in human males, whose testosterone (T) levels in both the circulation and in prostate decline, whereas those of 17β-estradiol (E2) remain relatively stable [2–4]. Thus, the age-associated alterations in the sex hormone milieu toward an estrogen predominance have been proposed as an endogenous risk factor for prostate carcinogenesis. Dynamic changes in the expression of estrogen receptor-β during PCa progression in humans [5,6] also suggest the involvement of estrogen signaling in the malignant transformation of the prostate. Moreover, the findings of aberrant activation of aromatase expression in benign prostatic hyperplasia and PCa [3,7] further advance the concept that local production of estrogens is pivotal in the pathobiology of this gland.

Humans are exposed to xenoestrogens from various sources, including food, drugs, and other exogenous venues. Xenoestrogens are believed to cause endocrine disruptions that lead to pathogenesis in reproductive end organs [8,9]. One agent of public health concern is diethylstilbestrol (DES), a synthetic estrogen, which was widely used as a growth-promoting animal feed additive [10]. Epidemiological studies have established a strong association between maternal exposure to DES and increased risk of vaginal, cervical, and perhaps breast cancers in DES daughters [11,12]. It remains uncertain, however, whether prenatal exposure to DES elevates cancer risk in men [13]. Transient exposure of neonatal rodents to estrogen increases susceptibility of the prostate to inflammation and cancer development later in life [14]. Together, these findings strongly suggest that natural or synthetic estrogen-mediated endocrine disruption leads to the evolution of prostatic diseases, including PCa.

We have established the T plus estrogen-induced dysplasia/carcinoma model in Noble (NBL) rats as a robust study system for deciphering the contributions of estrogens, in adulthood, on the pathogenesis of dysplasia, a precancerous lesion, and adenocarcinoma of the prostate [15,16]. In this model, estrogens are administered through subcutaneous implants of hormone-filled Silastic capsules with the coimplantation of T-filled capsules to maintain physiological levels of this androgen in the circulation [17]. It is well known that increased exposure to estrogens leads to a decline in circulating levels of T through the hypothalamic-pituitary-gonadal axis. The topographical localization of premalignant/malignant lesions within the rat prostate gland has been found to be dependent on the type of estrogens used. Under the same exogenous androgen support, E2 induces epithelial dysplasia and adenocarcinoma in the lateral prostate (LP) but not in the ventral prostate (VP) [16–19], whereas synthetic DES specifically targets the VP but not the LP [19,20]. Why two different estrogens induce lobe-specific carcinogenic actions in the prostate remains unclear. To address this issue, we conducted gene expression profiling analyses on the LP and VP of NBL rats exposed to T + E2 or T + DES. The experimental design and the application of a bioinformatic tool of gene network mapping enabled us to identify lobe-specific and common estrogen-mediated disruptions in multiple biologic networks/pathways that may be linked to prostate carcinogenesis and explain the distinctive action of the two estrogens.

Materials and Methods

Animals and Hormonal Treatment

The animal usage protocol was approved by the Institutional Animal Care Committee at the University of Cincinnati. Male NBL rats (5–6 weeks old) were purchased from Charles River Laboratories (Kingston, NY), kept under standard conditions, and treated as previously reported [16,19,20]. Briefly, animals were randomized into three groups (n = 5 for each group). Rats in the T + E2 treatment group received subcutaneous implants of two pieces of 2-cm-long Silastic capsules containing T (Sigma, St Louis, MO) and one piece of 1-cm-long capsules packed with E2 (Sigma), whereas the T + DES treatment group received the same number of Silastic capsules of the same length filled with T and DES (Sigma). Age-matched untreated control rats were implanted with empty capsules. At the end of a 16-week treatment period, animals were sacrificed with an overdose of isoflurane, and VPs and LPs were excised. One half of each lobe was processed for histologic examination, and the other half was snap-frozen for RNA extraction.

Microarray Hybridization

The Atlas Glass Rat 3.8 I Microarray (Clontech, Palo Alto, CA) carrying 3800 named rat genes (spotted oligonucleotides) were used as the gene chip platform. The Atlas Glass Fluorescent Labeling Kit (Clontech) was used for synthesizing and purifying fluorescent-labeled cDNA probes for hybridization to glass microarrays. The labeling and hybridization procedures were performed in accordance with the manufacturer's instruction manual. In brief, amino-modified first-strand cDNA probes were synthesized with aminoallyl. 2′-deoxyuridine 5′-triphosphate incorporation. Then Cy3 fluorescent dye was coupled to the cDNAs derived from individual prostatic lobes, whereas Cy5 dye was conjugated to the universal rat reference RNA obtained from Stratagene (La Jolla, CA). Equal quantities of two labeled probes were mixed and hybridized in a Corning microarray hybridization chamber (Corning, Corning, NY) at 50°C overnight (≥16 hours). Spikes of positive control probes were also included as an internal control for the process of cDNA probe synthesis and the dye-coupled reaction. Finally, the signal was obtained using a microarray scanner (GenePix 4000B; Axon Instruments, Foster City, CA). A probe coverage of >90% was achieved for all arrays. Five animal replicates for each treatment/tissue group (T + E2-treated VP or LP, T + DES-treated VP and LP, and untreated VP and LP) were used to conduct a 30-microarray analysis to assess changes in the gene expression pattern due to treatment and lobe specificity.

Microarray Data Normalization and Analysis

The data were analyzed to identify differentially expressed genes in 1) the T + E2-treated LPs and VPs compared with untreated controls and 2) the T + DES-treated LPs and VPs compared with untreated controls. Five biologic replicate arrays for each experimental condition, all versus universal reference, were performed. R statistical software and the limma Bioconductor package [21] were used for analysis. Data normalization was performed in two steps separately for each microarray [22–24]. First, background-adjusted intensities were log-transformed, and the differences (M) and averages (A) of log-transformed values were calculated as M = log2(X1) - log2(X2) and A = [log2(X1) + log2(X2)]/2, where X1 and X2 denote the Cy5 and Cy3 intensities, respectively. Second, normalization was performed by fitting the array-specific local regression model of M as a function of A and obtaining residuals. The statistical analysis was performed for each gene separately by fitting a one-way analysis of variance (ANOVA) model with treatment. Estimated fold changes were calculated from the ANOVA model; an intensity-based empirical Bayes method was used to modify the resulting t-statistics from each comparison [25]. This method obtains more precise estimates of variance by pooling information across genes and by accounting for the dependency of variance on probe intensity level. Genes with a false discovery rate (FDR) <0.05 [26] were considered to be significantly differentially expressed. Clustering was performed using normalized, centered sample ratios. The gene list used for clustering consisted of all genes having an FDR <0.05 for at least one comparison (1063 genes). T + E2 and T + DES samples were each clustered using the Ward clustering method and Euclidean dissimilarity metric with 1063 genes.

Identification of Estrogen-Regulated Gene Expression

Genes differentially regulated in the different lobes with T + DES and T + E2 treatment were identified separately in each drug treatment. Gene lists were generated according to their expression signature in different gene expression clusters; each group was described in the Results section.

Pathway and Network Analysis

Biologic relationships between differentially expressed genes were mapped by Ingenuity Pathway Analysis (IPA) 3.1 software (www.ingenuity.com). Gene lists of different patterns of gene expression in response to T + E2 and T + DES treatments (as described in the Results section) were uploaded to the IPAWeb application in an Excel file format containing expression data and GenBank accession number as identifier. The biologic relationship of uploaded genes was mapped with IPA software into networks according to the published literature in the database. A score was assigned to each network in the data set to estimate the relevance of the network to the uploaded gene list. A higher score means that the network is more relevant to the gene list entered by the user [27]. The two highest score networks were selected in this study, and genes in these two networks were selected for further post hoc analysis.

Reverse Transcription and Real-Time Quantitative Polymerase Chain Reaction

The total RNA from each sample of prostate tissue was reverse-transcribed into cDNA using Superscript III (Invitrogen, Carlsbad, CA). Specific primers were designed using either Primer Express 3 (Applied Biosystems, Foster City, CA) or Primer3 software [28]; the sequence of the primer is listed in Table W1. Real-time quantitative polymerase chain reaction (q-PCR) was performed on the 7900HT Fast Real-Time PCR System (ABI Biosystems) using the Power SYBR Green PCR master mix (Applied Biosystems). Polymerase chain reaction was performed in a total volume of 10 µl containing 50 ng of total cDNA, 1x Power SYBR Green PCR master mix, and a final primer concentration of 800 nM. The relative expression level was analyzed by the ΔΔCt method [29] and one-way ANOVA followed by Tukey post hoc analysis, where P < .05 was considered statistically significant.

Results

T + E2 and T + DES Treatments Differentially Induced Dysplasia in LP and VP, Respectively

The expected physiological and histologic changes resulting from T + E2 and T + DES treatment were observed as reported previously [15,18–20]. Dysplasia was observed in the T + E2-treated LPs (100% incidence) and T + DES.treated VPs (100% incidence), whereas no preneoplastic lesions were observed in T + E2-treated VPs and T + DES LPs. In T + E2-treated LPs, the dysplastic lesions were often accompanied by inflammatory infiltrates [17].

Hierarchical Clustering Identified Differential Action of E2 and DES in the Two Prostate Lobes

Unsupervised hierarchical clustering was performed for each hormone treatment group (T + E2 or T + DES) to determine the relatedness of replicate LP and VP samples (n = 5 per group) according to similarity in gene expression patterns among the 1063 genes with a significant difference in expression across samples, without prior knowledge of gene and sample identity.

Hierarchical clustering of samples showed that all LPs treated with T + E2 formed one cluster distinct from the cluster containing all untreated LPs. The hormone treatment, however, did not partition the hormone-treated and -untreated VPs, which formed a single large cluster (Figure 1A). These results indicate that the T + E2 treatment induced changes in gene expression mainly in the LP and had little, if any, effect on the gene expression pattern in the VP.

Figure 1.

Figure 1

Hierarchical clustering analysis of T + E2 and T + DES gene expression data set. (A) Dendrogram of T + E2 expression data set in LP and VP. (B) Dendrogram of T + DES expression data set in LP and VP. (C) Venn diagram showing the number of genes differentially expressed in each treatment group compared with the respective untreated control. (D) Gene interaction network of a subset of differentially expressed genes that are common in both LP and VP dysplasia. Genes bordered with red were validated by quantitative real-time PCR.

In a similar manner, T + DES treatment segregated only the VPs into two distinct clusters (treated and untreated) and did not partition the LPs into distinctive groups (Figure 1B). These findings indicate that the hormone treatment altered primarily the gene expression pattern in the VP and had little effect on LP gene expression. It is interesting that the gene expression pattern in the VP after T + DES treatment appeared to more closely resemble that observed in the LPs, as the T + DES-treated VPs formed a cluster more closely linked to the LP cluster than the one comprising untreated VPs.

Identification of Estrogen-Induced Differentially Expressed Genes Related to Dysplasia in Rat LP or VP

We used the following criteria to identify two panels of dysplasia-related genes: 1) the T + E2-induced LP dysplasia panel contains genes whose expression changed following T + E2 treatment in the LPs harboring dysplasia but excludes those whose expression also changed in the VPs with no dysplastic changes (253 genes; Figure 1C, left; Table 1), and 2) the T + DES-induced VP dysplasia panel includes genes whose expression changed after T + DES treatment in the VPs harboring dysplasia but excludes those whose expression also changed in the LPs without dysplasia (198 genes; Figure 1C, right; Table 2).

Table 1.

T + E2-Induced LP Dysplasia Panel: Genes Whose Expression Selectively Changed following T + E2 Treatment in the LPs Harboring Dysplasia, but not in the VPs.

Gene Names Locus ID Symbol Fold Changes Functions
prohibitin* 25344 PHB 3.42 Cell death; cell signaling; cellular growth and proliferation; gene transcription
nuclease-sensitive element binding protein 1* 29206 NSEP1 3.08 Cell death; gene transcription
potassium channel, subfamily K, member 9* 84429 KCNK9 3.08 Cell death
vesicle-associated membrane protein 2* 24803 VAMP2 2.97 Cell death; cellular assembly and organization; cellular movement
cytochrome c oxidase subunit IV isoform 1* 29445 COX4I1 2.71 Others/unclassified
PR-Vbeta1* 498341 PR-Vbeta1 2.68 Others/unclassified
melanocortin 5 receptor* 25726 MC5R 2.47 Cell signaling
glutamate receptor, ionotropic, kainate 3* 298521 GRIK3 2.45 Cell signaling
6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 4* 54283 PFKFB4 2.41 Others/unclassified
potassium voltage-gated channel, Shaw-related subfamily, member 1* 25327 KCNC1 2.39 Molecular transport
actin alpha cardiac 1* 29275 ACTC1 2.34 Others/unclassified
eukaryotic translation elongation factor 1 alpha 1* 171361 EEF1A1 2.31 Cell death, protein synthesis
ATPase, Na+/K+ transporting, alpha 3 polypeptide* 24213 ATP1A3 2.29 Inflammation; molecular transport
gamma-aminobutyric acid A receptor, rho 1* 29694 GABRR1 2.25 Cell death; cell signaling
guanylate cyclase 1, soluble, alpha 3* 25201 GUCY1A3 2.25 Cellular movement
transforming growth factor alpha* 24827 TGFA 2.24 Biomolecule metabolism; cell cycle; cell death; cell morphology; cell signaling; cellular development; cellular growth and proliferation; cellular movement; gene transcription; molecular transport; protein synthesis
ribosomal protein S12* 65139 RPS12 2.20 Protein synthesis
tumor protein p53* 24842 TP53 2.16 Cell cycle; cell death; cell morphology; cell signaling; cellular assembly and organization; cellular development; cellular growth and proliferation; cellular movement; cellular response to therapeutics; DNA replication, recombination and repair; free radical scavenging; gene transcription; inflammation; molecular transport; post-translational modification; protein synthesis; RNA post-transcriptional modification
nuclear receptor subfamily 3, group C, member 2* 25672 NR3C2 2.13 Biomolecule metabolism; cell signaling; gene transcription; molecular transport; protein synthesis
secretory carrier membrane protein 1* 29521 SCAMP1 2.09 Cellular assembly and organization
CEA-related cell adhesion molecule 1* 81613 CEACAM1 2.08 Cell death; cell signaling; cellular growth and proliferation; cellular movement; inflammation
olfactory marker protein* 24612 OMP 2.08 Cell signaling
guanine nucleotide binding protein, alpha z subunit* 25740 GNAZ 2.05 Cell signaling; cellular development; cellular movement; molecular transport; biomolecule metabolism
phosphodiesterase 1C* 81742 PDE1C 2.04 Others/unclassified
ribosomal protein S3a* 29288 RPS3A 2.04 Cell death; cellular development; cellular growth and proliferation; protein synthesis
insulin-like 6* 50546 IL1RAP 1.95 Cell signaling
A kinase (PRKA) anchor protein 14* 60332 AKAP14 1.94 Others/unclassified
CD38 antigen* 25668 CD38 1.94 Cell cycle; cell death; cell signaling; cellular development; cellular growth and proliferation; cellular movement; biomolecule metabolism; molecular transport; post-translational modification
p21 (CDKN1A)-activated kinase 1* 29431 PAK1 1.93 Cell death; cell morphology; cell signaling; cellular assembly and organization; cellular development; cellular movement
regenerating islet-derived 3 alpha* 171162 REG3G 1.93 Others/unclassified
ubiquitin-conjugating enzyme E2I* 25573 UBE2I 1.93 Cell signaling; gene transcription; protein synthesis
ATPase, H+ transporting, V1 subunit F* 116664 ATP6V1F 1.91 Molecular transport
homeo box A1* 25607 HOXA1 1.89 Cell death; cellular development; cellular movement; gene transcription
regenerating islet-derived 3 gamma* 24620 REG3G 1.89 Others/unclassified
Fas apoptotic inhibitory molecule 2* 246274 FAIM2 1.86 Cell death
barrier to autointegration factor 1* 114087 BANF1 1.85 DNA replication, recombination, and repair
thyroid hormone receptor alpha* 81812 THRA 1.85 Biomolecule metabolism; cell death; cell morphology; cellular development; free radical scavenging; gene transcription; protein synthesis
neuromedin B receptor* 25264 NMBR 1.83 Others/unclassified
ATPase, Na+/K+ transporting, beta 2 polypeptide* 24214 ATP1B2 1.82 Others/unclassified
Chondroitin sulfate proteoglycan 5* 50568 CSPG5 1.81 Cell signaling
MAD homolog 7 (Drosophila)* 81516 SMAD7 1.78 Cell death; cell morphology; cell signaling; cellular development; cellular growth and proliferation; gene transcription
tachykinin receptor 2* 25007 TACR2 1.78 Others/unclassified
alanyl (membrane) aminopeptidase* 81641 ANPEP 1.76 Cell death; cell morphology; cellular development; cellular movement; protein synthesis
growth hormone-releasing hormone* 29446 GHRH 1.75 Cell morphology; cell signaling; cellular growth and proliferation; biomolecule metabolism; molecular transport
protein kinase N1* 29355 PKN1 1.75 Cell signaling; cellular growth and proliferation; cellular movement; gene transcription
slit homolog 3 (Drosophila)* 83467 SLIT3 1.74 Others/unclassified
dopamine receptor 2* 24318 DRD2 1.72 Cell death; cell signaling; cellular growth and proliferation; cellular movement; gene transcription; inflammation; biomolecule metabolism
glutathione peroxidase 3* 64317 GPX3 1.72 Cellular growth and Proliferation; Post-translational modification; protein synthesis
vasoactive intestinal peptide receptor 1* 24875 VIPR1 1.71 Cell death; cell signaling; cellular growth and proliferation; cellular movement; biomolecule metabolism; molecular transport
amphiphysin 1* 60668 AMPH 1.70 Cell signaling; cellular assembly and organization; cellular movement; gene transcription
G protein beta subunit-like* 64226 GBL 1.70 Others/unclassified
heat shock 70 kDa protein 5* 25617 HSPA5 1.69 Cell death; cellular growth and proliferation; inflammation
hydroxy-delta-5-steroid dehydrogenase, 3 beta- and steroid delta-isomerase 1* 29632 HSD3B2 1.68 Others/unclassified
POU domain, class 3, transcription factor 2* 29588 POU3F2 1.68 Cellular development; cellular growth and proliferation; cellular movement; gene transcription
ribosomal protein S9* 81772 RPS9 1.64 Protein synthesis
glucocorticoid modulatory element binding protein 2* 83635 GMEB2 1.63 Gene transcription
glutamate receptor, ionotropic, NMDA2B* 24410 GRIN2B 1.63 Cell signaling
solute carrier family 8 (sodium/calcium exchanger), member 3* 140448 SLC8A3 1.63 Cell death; cell signaling
ATPase, Ca2+ transporting, ubiquitous* 25391 ATP2A3 1.62 Others/unclassified
CTD-binding SR-like rA1* 56081 SR-A1 1.60 Others/unclassified
solute carrier family 2 (facilitated glucose transporter), member 2* 25351 SLC2A2 1.60 Cell death
calcineurin binding protein 1* 94165 CABIN1 1.59 Cell cycle; cell death; cell morphology; cell signaling; cellular development; cellular growth and proliferation; cellular movement; DNA replication, recombination, and repair; gene transcription; inflammation; biomolecule metabolism; molecular transport
heterogeneous nuclear ribonucleoprotein A1* 29578 LOC144983 1.59 Others/unclassified
casein kinase 1, gamma 1* 64086 CSNK1G1 1.57 Others/unclassified
potassium inwardly rectifying channel, subfamily J, member 10* 29718 KCNJ10 1.56 Cell morphology; cellular development; inflammation; molecular transport
potassium large conductance calcium-activated channel, subfamily M, beta member 1* 29747 KCNMB1 1.56 Cell signaling; molecular transport
potassium voltage-gated channel, Shaw-related subfamily, member 3* 117101 KCNC3 1.56 Molecular transport
CCAAT/enhancer binding protein (C/EBP), delta* 25695 CEBPD 1.55 Cell death; cellular development; cellular growth and proliferation; gene transcription; inflammation
guanine nucleotide binding protein, beta 3* 60449 GNB3 1.55 Cell signaling
ribosomal protein S19* 29287 RPS19 1.54 Cellular development; cellular growth and proliferation; cellular movement; protein synthesis
aquaporin 5* 25241 AQP5 1.53 Inflammation
chloride channel Kb* 79430 CLCNKB 1.53 Others/unclassified
fatty acid binding protein 3* 79131 FABP3 1.52 Cellular growth and proliferation; biomolecule metabolism; molecular transport
endothelial differentiation, lysophosphatidic acid G-protein-coupled receptor, 2* 116744 EDG2 1.51 Cell death; cell morphology; cell signaling; cellular development; cellular growth and proliferation; cellular movement; gene transcription
killer cell lectin-like receptor subfamily B member 1B* 25192 KLRB1 1.51 Cell death
suppression of tumorigenicity 18* 266680 ST18 1.50 Gene transcription
pregnancy upregulated nonubiquitously expressed CaM kinase* 29660 PNCK 1.49 Others/unclassified
Arg/Abl-interacting protein ArgBP2* 114901 SORBS2 1.48 Cell death; cell morphology; cell signaling
sodium channel, voltage-gated, type IV, alpha polypeptide* 25722 SCN4A 1.48 Molecular transport
calcitonin/calcitonin-related polypeptide, alpha* 24241 CALCA 1.47 Cell cycle; cell death; cell morphology; cell signaling; cellular development; cellular growth and proliferation; cellular movement; DNA replication, recombination, and repair; gene transcription; inflammation; biomolecule metabolism; molecular transport
RASD family, member 2* 171099 RASD2 1.47 Others/unclassified
spondin 1* 64456 SPON1 1.46 Others/unclassified
sulfotransferase family, cytosolic, 1C, member 2* 171072 SULT1C2 1.46 Others/unclassified
acyl-CoA synthetase long-chain family member 4* 113976 ACSL4 1.45 Cell death
aldehyde dehydrogenase family 1, subfamily A2* 116676 ALDH1A2 1.45 Cell death; cellular development; cellular growth and proliferation; biomolecule metabolism
CD3 antigen, zeta polypeptide* 25300 CD247 1.45 Cell death; cell morphology; cell signaling; cellular development; cellular growth and proliferation; gene transcription; inflammation; post-translational modification
discs, large homolog 4 (Drosophila)* 29495 DLG4 1.45 Cell signaling; cellular assembly and organization
calmodulin 3 24244 CALM3 1.43 Cell signaling; cellular growth and proliferation
carboxypeptidase E* 25669 CPE 1.43 Biomolecule metabolism; molecular transport
phosphofructokinase, muscle* 65152 PFKM 1.42 Others/unclassified
translocase of outer mitochondrial membrane 20 homology (yeast)* 266601 TOMM20 1.42 Others/unclassified
complement component 1, q subcomponent, beta polypeptide* 29687 C1QB 1.40 Others/unclassified
regulator of G-protein signaling 19* 59293 RGS19 1.40 Cell signaling; cellular development; biomolecule metabolism; protein synthesis
Carcinoembryonic antigen gene family (CGM3)* 24256 PSG18 1.39 Others/unclassified
gamma-glutamyl hydrolase* 25455 GGH 1.39 Others/unclassified
N-acetyltransferase 8 (camello like)* 64570 NAT8 1.37 Others/unclassified
pyrimidinergic receptor P2Y, G-protein-coupled, 6* 117264 P2RY6 1.37 Others/unclassified
protein tyrosine phosphatase, receptor type, K, extracellular region* 360302 PTPRK 1.36 Cellular growth and proliferation
cytochrome P450, 4a12* 266674 CYP4A22 1.35 Others/unclassified
gamma-aminobutyric acid A receptor, alpha 1* 29705 GABRA1 1.35 Cell morphology; cell signaling
ferritin, heavy polypeptide 1* 25319 FTH1 1.34 Cell death; cell morphology; cellular growth and proliferation; DNA replication, recombination, and repair; free radical scavenging; cell death; cell morphology; cellular growth and proliferation; free radical scavenging
guanylate cyclase 1, soluble, beta 2* 25206 GUCY1B2 1.34 Others/unclassified
proteasome (prosome, macropain) 28 subunit, beta* 29614 PSME2 1.31 Cell signaling; cellular growth and proliferation
small inducible cytokine A4* 116637 CCL4 1.31 Cell death; cell morphology; cell signaling; cellular development; cellular growth and proliferation; cellular movement; inflammation biomolecule metabolism
ADP-ribosylation factor 6* 79121 ARF6 1.30 Cell death; cell morphology; cell signaling; cellular assembly and organization; cellular movement; biomolecule metabolism; molecular transport; protein trafficking
mannan-binding lectin serine protease 2 64459 MASP2 1.30 Others/unclassified
dopamine receptor 4* 25432 DRD4 1.29 Cell signaling; biomolecule metabolism; molecular transport
ATP synthase, H+ transporting, mitochondrial F1 complex, delta subunit* 245965 ATP5D 1.26 Energy production; molecular transport; biomolecule metabolism
calponin 1 65204 CNN1 1.26 Cellular assembly and organization; cellular growth and proliferation; cellular movement
hydroxyacid oxidase 2 (long chain) 84029 HAO2 1.25 Others/unclassified
cytotoxic T-lymphocyte-associated protein 4 63835 CTLA4 1.23 Cell cycle; cell death; cell signaling; cellular development; cellular growth and proliferation; cellular movement; inflammation
potassium voltage-gated channel, subfamily H (eag-related), member 7* 170739 KCNH7 1.23 Others/unclassified
Ras association (RalGDS/AF-6) domain family 5 54355 RASSF5 1.23 Cell cycle; cell death; cellular growth and proliferation
thymopoietin 25359 TMPO 1.22 Cell cycle; cellular assembly and organization; DNA replication, recombination, and repair; gene transcription
polypyrimidine tract binding protein 1 29497 PTBP1 1.19 Protein synthesis
protein kinase C and casein kinase substrate in neurons 2 124461 PACSIN2 1.16 Cell morphology; cell signaling; cellular assembly and organization
leptin receptor overlapping transcript 56766 LEPROT 1.15 Others/unclassified
phospholipase C, delta 1 24655 PLCD1 1.15 Biomolecule metabolism; cellular growth and proliferation
synaptonemal complex protein SC65 59101 SC65 1.15 Others/unclassified
ADP-ribosylation factor 5 79117 ARF5 1.14 Molecular transport; protein trafficking
crystallin, beta B2 25422 CRYBB2 1.11 Others/unclassified
prolactin-like protein L 171556 PRLPL 1.10 Others/unclassified
protein phosphatase 3, catalytic subunit, beta isoform 24675 PPP3CB 1.09 Cell death; cellular development; inflammation
calpain 3 29155 CAPN3 1.07 Protein synthesis
cathepsin D 171293 CTSD 1.07 Cell death; cellular growth and proliferation; free radical scavenging; inflammation; molecular transport; protein synthesis
eukaryotic translation initiation factor 2B, subunit 2 beta 84005 EIF2B2 1.07 Cellular development; cellular growth and proliferation; protein synthesis
calcitonin/calcitonin-related polypeptide, alpha 24241 CALCA -1.13 Cell cycle; cell death; cell morphology; cell signaling; cellular development; cellular growth and proliferation; cellular movement; DNA replication, recombination, and repair; gene transcription; inflammation; biomolecule metabolism; molecular transport
forkhead box M1 58921 FOXM1 -1.13 Cell cycle; cell death; cell morphology; cellular growth and proliferation; gene transcription
adrenal secretory serine protease precursor 64565 TMPRSS11D -1.14 Others/unclassified
MAP-kinase activating death domain 94193 MADD -1.19 Cell death; cellular growth and proliferation
calmodulin 3 24244 CALM3 -1.22 Cell signaling; cellular growth and proliferation
neural visinin-like Ca2+-binding protein type 3* 50871 HPCAL1 -1.28 Others/unclassified
fertility protein SP22 117287 PARK7 -1.30 Cell death; cell signaling; cellular growth and proliferation
actin, beta* 81822 ACTB -1.31 Cellular growth and proliferation; cellular movement
G-protein-coupled receptor 37* 117549 GPR37 -1.33 Cell death; cell signaling
glycogen synthase kinase 3 alpha* 50686 GSK3A -1.34 Cellular movement
ribosomal protein L22 81768 RPL22 -1.34 Protein synthesis
actinin alpha 4* 63836 ACTN4 -1.35 Cell death; cellular growth and proliferation; cellular movement
midline 1* 54252 MID1 -1.35 Cellular assembly and organization
dipeptidase 1 (renal)* 94199 DPEP1 -1.36 Others/unclassified
polymeric immunoglobulin receptor* 25046 PIGR -1.36 Cell signaling; cellular growth and proliferation; cellular movement
adducin 2 (beta)* 24171 ADD2 -1.38 Cellular development; cellular growth and proliferation; inflammation; molecular transport
c-mer protooncogene tyrosine kinase* 65037 MERTK -1.38 Cell death; cell morphology; cell signaling; cellular development; cellular growth and proliferation
caspase 7* 64026 CASP7 -1.39 Cell death; protein synthesis
ddx5 gene 287765 DDX5 -1.40 Cell death; cellular growth and proliferation; gene transcription
vesicle docking protein* 56042 VDP -1.40 Cellular assembly and organization; molecular transport; protein trafficking
acetylcholinesterase* 83817 ACHE -1.41 Cell death; cell morphology; cell signaling; cellular development; cellular growth and proliferation; DNA replication, recombination, and repair; post-translational modification; protein synthesis
phenylalkylamine Ca2+ antagonist (emopamil) binding protein* 117278 EBP -1.41 Cellular development
phosphodiesterase 4C, cAMP-specific* 290646 PDE4C -1.41 Inflammation
transition protein 2* 24840 TNP2 -1.44 Others/unclassified
casein kinase 1, alpha 1* 113927 CSNK1A1 -1.45 Cell death
nuclear receptor subfamily 4, group A, member 3 58853 NR4A3 -1.46 Cellular development; cellular growth and proliferation; gene transcription
phospholipase C, beta 3* 29322 PLCB3 -1.46 Biomolecule metabolism; cellular movement; molecular transport
myelin-associated oligodendrocytic basic protein* 25037 MOBP -1.47 Others/unclassified
retinoic acid receptor, alpha* 24705 RARA -1.47 Cell death; cell signaling; cellular development; cellular growth and proliferation; cellular movement; biomolecule metabolism; gene transcription; inflammation
putative pheromone receptor Go-VN13C* 286986 EG665255 -1.48 Others/unclassified
prolyl 4-hydroxylase, beta polypeptide* 25506 P4HB -1.49 Cell death
protein kinase, lysine-deficient 1* 116477 WNK1 -1.49 Molecular transport
Pyruvate carboxylase* 25104 PC -1.49 Others/unclassified
ribosomal protein S15* 29285 RPS15 -1.49 Others/unclassified
ATP synthase, H+ transporting, mitochondrial F1 complex, O subunit* 192241 ATP5O -1.50 Others/unclassified
granzyme M (lymphocyte met-ase 1)* 29252 GZMM -1.50 Cell death
metastasis-associated 1* 64520 MTA1 -1.50 Cell morphology; cell signaling; cellular growth and proliferation; cellular movement; gene transcription
Rous sarcoma oncogene* 83805 SRC -1.50 Cell death; cell morphology; cell signaling; cellular development; cellular growth and proliferation; cellular movement; gene transcription; biomolecule metabolism; molecular transport; protein synthesis
neurexophilin 3* 59315 NXPH3 -1.51 Others/unclassified
phosphodiesterase 4A* 25638 PDE4A -1.51 Cell death; cell signaling; cellular development; inflammation; molecular transport; biomolecule metabolism
calcium channel, voltage-dependent, alpha 1I subunit* 56827 CACNA1I -1.52 Cell signaling
transient receptor potential cation channel, subfamily C, member 4* 84494 TRPC4 -1.53 Others/unclassified
tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein, epsilon polypeptide* 29753 YWHAE -1.53 Cell cycle; cell signaling; cellular movement
polymerase (RNA) II (DNA-directed) polypeptide G* 117017 POLR2G -1.54 Gene transcription
F-box only protein 2* 85273 FBXO2 -1.55 Cellular growth and proliferation; protein synthesis
apolipoprotein C–I* 25292 APOC1 -1.56 Biomolecule metabolism; molecular transport
proprotein convertase subtilisin/kexin type 3* 54281 FURIN -1.56 Cell signaling; cellular growth and proliferation; cellular movement; protein synthesis
chemokine orphan receptor 1* 84348 CXCR7 -1.57 Cellular growth and proliferation; cellular movement
heat shock protein, alpha-crystallin-related, B6* 192245 HSPB6 -1.57 Others/unclassified
interleukin 5* 24497 IL5 -1.57 Cell cycle; cell death; cell morphology; cell signaling; cellular development; cellular growth and proliferation; cellular movement; gene transcription; molecular transport
interleukin enhancer binding factor 3* 84472 ILF3 -1.57 Cellular growth and proliferation; gene transcription
protein tyrosine phosphatase, receptor type, F* 360406 PTPRF -1.57 Cell cycle; cell death; cell morphology; cell signaling; cellular assembly and organization; cellular growth and proliferation; cellular movement
par-3 (partitioning defective 3) homolog (C. elegans)* 81918 PARD3 -1.58 Cell morphology; cell signaling; cellular development; gene Transcription
ribosomal protein L27* 64306 RPL27 -1.58 Others/unclassified
linker for activation of T cells* 81511 LAT -1.59 Biomolecule metabolism; cell death; cell morphology; cell signaling; cellular development; gene transcription; molecular transport
nuclear factor I/C* 29228 NFIC -1.59 Gene transcription
potassium voltage-gated channel, shaker-related subfamily, beta member 1* 29737 KCNAB1 -1.59 Molecular transport
Bcl2-associated X protein* 24887 BAX -1.60 Cell cycle; cell death; cell morphology; cell signaling; cellular assembly and organization; cellular development; cellular growth and proliferation; cellular response to therapeutics; DNA replication, recombination, and repair; free radical scavenging; inflammation; biomolecule metabolism; molecular transport
carboxylesterase 1* 29225 ES22 -1.60 Others/unclassified
ribosomal protein L10A* 81729 RPL10A -1.60 Others/unclassified
upstream of NRAS* 117180 CSDE1 -1.60 Others/unclassified
nucleoporin 62* 65274 NUP62 -1.61 Cell death; cell signaling; cellular growth and proliferation; gene transcription
eukaryotic translation initiation factor 2B, subunit 4 delta* 117019 EIF2B4 -1.62 Cellular development; protein synthesis
apolipoprotein A–V* 140638 APOA5 -1.63 Biomolecule metabolism; molecular transport
myosin IE* 25484 MYO1E -1.64 Others/unclassified
paired box gene 8* 81819 PAX8 -1.64 Cellular development; gene transcription
prion protein* 24686 PRNP -1.64 Cell death; cellular development; cellular growth and proliferation; cellular movement
amelogenin X chromosome* 29160 AMELX -1.67 Others/unclassified
Unc4.1 homeobox (C. elegans)* 29375 UNCX4.1 -1.67 Cell death
cofilin 1* 29271 CFL1 -1.68 Cell morphology; cell signaling; cellular assembly and organization; cellular development; cellular growth and proliferation; cellular movement; molecular transport; protein trafficking
caudal type homeo box 1* 171042 CDX1 -1.69 Others/unclassified
phosphatidylinositol-4-phosphate 5-kinase, type II, alpha* 116723 PIP5K2A -1.70 Biomolecule metabolism; molecular transport
platelet-activating factor acetylhydrolase, isoform 1b, alpha1 subunit* 114113 PAFAH1B3 -1.70 Cell death; cellular development; biomolecule metabolism
protein tyrosine phosphatase, nonreceptor type substrate 1* 25528 SIRPA -1.70 Cell death; cell morphology; cell signaling; cellular assembly and organization; cellular development; cellular growth and proliferation; cellular movement; gene transcription; inflammation
acetylcholinesterase* 83817 ACHE -1.71 Cell death; cell morphology; cell signaling; cellular development; cellular growth and proliferation; DNA replication, recombination, and repair; post-translational modification; protein synthesis
ribosomal protein L36* 58927 RPL36 -1.71 Others/unclassified
G-protein-coupled receptor 24* 83567 MCHR1 -1.72 Cell signaling; molecular transport; biomolecule metabolism
epididymal retinoic acid-binding protein* 29552 LCN5 -1.73 Biomolecule metabolism
gap junction membrane channel protein beta 4* 117055 GJB4 -1.75 Others/unclassified
zinc finger protein 111* 170849 ZNF227 -1.75 Others/unclassified
cleavage and polyadenylation-specific factor 4* 252943 CPSF4 -1.76 Cellular growth and proliferation
ADP-ribosylation factor 1* 64310 ARF1 -1.77 Cellular assembly and organization; cellular growth and proliferation; biomolecule metabolism; molecular transport; protein trafficking
coagulation factor X* 29243 F10 -1.77 Cell signaling; cellular movement; inflammation; biomolecule metabolism; molecular transport
ephrin A1* 94268 EFNA1 -1.77 Cell death; cell morphology; cell signaling; cellular assembly and organization; cellular development; cellular growth and proliferation; cellular movement
rabaptin 5* 54190 RABEP1 -1.78 Others/unclassified
allograft inflammatory factor 1* 29427 AIF1 -1.79 Cell death; cell morphology; cellular development; cellular growth and proliferation
Cbp/p300-interacting transactivator with Glu/Asp-rich carboxy-terminal domain 1* 64466 CITED1 -1.79 Cellular development; cellular growth and proliferation; gene transcription
testis enhanced gene transcript* 24822 TEGT -1.79 Cell death
neurogenic differentiation 2* 54276 NEUROD2 -1.80 Cellular development; gene transcription
heat shock 10 kDa protein 1* 25462 HSPE1 -1.83 Cell death
hsp70-interacting protein* 246146 HSPBP1 -1.83 Others/unclassified
calreticulin* 64202 CALR -1.84 Cell death; cellular assembly and organization; cellular development; cellular growth and proliferation; cellular movement; gene transcription; molecular transport; protein trafficking
discoidin domain receptor family, member 1* 25678 DDR1 -1.84 Cell death; cellular development; cellular growth and proliferation; cellular movement
myosin ID* 25485 MYO1D -1.84 Others/unclassified
ribosomal protein L28* 64638 RPL28 -1.84 Others/unclassified
gap junction membrane channel protein alpha 3* 79217 GJA3 -1.85 Cell morphology; cell signaling; cellular development
hairy and enhancer of split 3 (Drosophila)* 64628 HES3 -1.85 Cellular development; gene transcription
ornithine decarboxylase antizyme 1* 25502 OAZ1 -1.87 Cell death; cellular growth and proliferation
a disintegrin and metallopeptidase domain 1a* 56777 ADAM1A -1.89 Cellular movement
mucosal vascular addressin cell adhesion molecule 1* 54266 MADCAM1 -1.89 Cell signaling; cellular development; cellular movement; inflammation
membrane protein, palmitoylated 3 (MAGUK p55 subfamily member 3)* 114202 MPP3 -1.90 Others/unclassified
sulfotransferase family 4A, member 1* 58953 SULT4A1 -1.90 Others/unclassified
CCAAT/enhancer binding protein (C/EBP), alpha* 24252 CEBPA -1.91 Cell cycle; cell death; cellular development; cellular growth and proliferation; cellular response to therapeutics; gene transcription; biomolecule metabolism; molecular transport
SNF-related kinase* 170837 SNRK -1.92 Cellular development
ADP-ribosylation factor 3* 140940 ARF3 -1.94 Molecular transport; protein trafficking
aspartyl-tRNA synthetase* 116483 DARS -1.94 Cell cycle; cell signaling; protein synthesis
glycine cleavage system protein H (aminomethyl carrier)* 171133 GCSH -1.94 Biomolecule metabolism; post-translational modification
3-hydroxyisobutyrate dehydrogenase* 63938 HIBADH -1.96 Others/unclassified
preoptic regulatory factor-2* 286903 KIAA1688 -1.96 Others/unclassified
amino-terminal enhancer of split* 29466 AES -1.97 Cell death; gene transcription
ribosomal protein L29* 29283 RPL29 -1.99 Others/unclassified
quinoid dihydropteridine reductase* 64192 QDPR -2.01 Others/unclassified
complexin 2* 116657 CPLX2 -2.02 Cellular assembly and organization; cellular movement
hairy and enhancer of split 2 (Drosophila)* 29567 HES2 -2.02 Gene transcription
transmembrane 4 superfamily member 11* 64364 PLLP -2.02 Molecular transport
ferritin light chain 1* 29292 FTL -2.03 Cellular growth and proliferation
growth hormone-releasing hormone receptor* 25321 GHRHR -2.04 Cell signaling; cellular growth and proliferation; molecular transport; biomolecule metabolism
membrane protein, palmitoylated 2 (MAGUK p55 subfamily member 2)* 85275 MPP2 -2.04 Cell signaling
ribosomal protein S27* 94266 RPS27 -2.05 Cell signaling; cellular growth and proliferation; protein synthesis
translocase of inner mitochondrial membrane 22 homolog (yeast)* 79463 TIMM22 -2.06 Others/unclassified
sec22 homolog* 117513 SEC22A -2.08 Molecular transport; protein trafficking
transmembrane protein with EGF-like and two follistatin-like domains 1* 63845 TMEFF1 -2.11 Cellular growth and proliferation
paired-like homeodomain transcription factor 3* 29609 PITX3 -2.12 Cellular development; gene transcription
fibrinogen, gamma polypeptide* 24367 FGG -2.25 Cell signaling
neurogenic differentiation 1* 29458 NEUROD1 -2.26 Cell death; cell morphology; cell signaling; cellular development; cellular growth and proliferation; cellular movement; gene transcription
steroid-sensitive gene 1* 64387 CCDC80 -2.31 Others/unclassified
ubiquitin A-52 residue ribosomal protein fusion product 1* 64156 UBA52 -2.33 Gene transcription; protein synthesis
synuclein, gamma* 64347 SNCG -2.42 Cell death; cell morphology; cellular growth and proliferation; cellular movement
*

The change in expression was significant compared with untreated control (P < .05). Gene names in italics are common in both panels of T + E2-treated LP and T + DES-treated VP.

Table 2.

T + DES-Induced VP Dysplasia Panel: Genes Whose Expression Selectively Changed following T + DES Treatment in the VPs Harboring Dysplasia, but not in the LPs.

Gene Names Locus ID Symbol Fold Changes Functions
glutathione peroxidase 3* 64317 GPX3 17.188 Cellular development; cellular growth and proliferation; post-translational modification; protein synthesis
ferritin, heavy polypeptide 1* 25319 FTH1 6.304 Cell death; cell morphology; cellular growth and proliferation; DNA replication, recombination, and repair; free radical scavenging; post-translational modification
protein phosphatase 2a, catalytic subunit, alpha isoform* 24672 PPP2CA 3.388 Biomolecule metabolism; cell death; cellular growth and proliferation; post-translational modification
monocarboxylate transporter* 80878 SLC16A3 2.263 Others/unclassified
nuclease-sensitive element binding protein 1* 29206 NSEP1 1.898 Cell death; gene transcription
thymopoietin* 25359 TMPO 1.814 Cell cycle; DNA replication, recombination, and repair; gene transcription
homeo box A1* 25607 HOXA1 1.586 Cell death; cellular development; cellular movement; gene transcription
cystatin C* 25307 CST3 1.585 Cell death; cellular development; cellular growth and proliferation
thyroid hormone receptor alpha* 81812 THRA 1.583 Cell death; cell morphology; cellular development; free radical scavenging; gene transcription
tubulin, alpha 1* 64158 TUBA1A 1.580 Inflammation
phospholipase C, delta 1* 24655 PLCD1 1.532 Cellular growth and proliferation
glutamate receptor, metabotropic 7* 81672 GRM7 1.481 Cell signaling
potassium voltage-gated channel, subfamily H (eag-related),member 7* 170739 KCNH7 1.481 Others/unclassified
plasminogen activator, urokinase* 25619 PLAU 1.461 Cell cycle; cell death; cell morphology; cell signaling; cellular assembly and organization; cellular development; cellular growth and proliferation; cellular movement; inflammation
vesicle-associated membrane protein 2 24803 VAMP2 1.452 Cell signaling; cellular assembly and organization; cellular movement; molecular transport
glyceraldehyde-3-phosphate dehydrogenase* 24383 GAPDH 1.401 Others/unclassified
protein phosphatase 1F (PP2C domain containing)* 287931 PPM1F 1.392 Biomolecule metabolism; cell death; post-translational modification
tumor protein p53* 24842 TP53 1.379 Cell cycle; cell death; cell morphology; cell signaling; cellular assembly and; organization; cellular development; cellular growth and proliferation; cellular movement; cellular response to therapeutics; DNA replication, recombination, and repair; free radical scavenging; gene transcription; inflammation; post-translational modification; protein synthesis
opioid receptor, mu 1* 25601 OPRM1 1.375 Cell death; cell signaling; cellular growth and proliferation; cellular movement; inflammation
calcitonin/calcitonin-related polypeptide, alpha* 24241 CALCA 1.361 Cell cycle; cell death; cell morphology; cell signaling; cellular development; cellular growth and proliferation; cellular movement; DNA replication, recombination, and repair; gene transcription; inflammation
testis-specific protein* 192229 C3ORF34 1.357 Others/unclassified
eukaryotic translation initiation factor 2B, subunit 2 beta* 84005 EIF2B2 1.334 Cellular development; cellular growth and proliferation
tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein, eta polypeptide* 25576 YWHAH 1.333 Cellular assembly and organization; gene transcription
glucagon-like peptide 1 receptor 25051 GLP1R 1.328 Cell death; gene transcription; inflammation
transient receptor potential cation channel, subfamily C, member 2* 64573 TRPC2 1.322 Others/unclassified
dopamine receptor 4* 25432 DRD4 1.311 Cell signaling
early growth response 3* 25148 EGR3 1.289 Cell death; cell signaling; cellular development; cellular growth and proliferation; gene transcription
glutamate receptor, ionotropic, NMDA2B* 24410 GRIN2B 1.248 Cell morphology; cell signaling
chemokine (C-X-C motif) ligand 5* 60665 CXCL6 1.236 Cell signaling; cellular movement
myosin IC 65261 MYO1C 1.190 Cell morphology
breast cancer anti-estrogen resistance 1 25414 BCAR1 1.190 Cell death; cell morphology; cell signaling; cellular assembly and organization; cellular development; cellular growth and proliferation; cellular movement
aryl hydrocarbon receptor nuclear translocator 2 25243 ARNT2 1.172 Gene transcription
CCAAT/enhancer binding protein (C/EBP), delta 25695 CEBPD 1.151 Cell cycle; cell death; cellular development; cellular growth and proliferation; gene transcription; inflammation; biomolecule metabolism; molecular transport
CTD-binding SR-like rA1 56081 SR-A1 1.148 Others/unclassified
ADP-ribosylation factor 6 79121 ARF6 1.142 Cell death; cell morphology; cellular assembly and organization; cellular movement
calmodulin 3 24244 CALM3 1.138 Cell death; cellular growth and proliferation; post-translational modification
synaptotagmin 3 25731 SYT3 1.115 Cell signaling; cellular assembly and organization; cellular movement; molecular transport
dynein, cytoplasmic, light chain 1 58945 DYNLL1 1.090 Cell death; cell morphology; cellular assembly and organization
MAD homolog 7 (Drosophila) 81516 SMAD7 1.062 Cell cycle; cell death; cell morphology; cellular development; cellular growth and proliferation; cellular movement; gene transcription
v-maf musculoaponeurotic fibrosarcom oncogene family, protein B (avian) 54264 MAFB 1.062 Cell death; cellular development; cellular movement; gene transcription
calmodulin 3 24244 CALM3 1.060 Cellular growth and proliferation; post-translational modification
thymosin, beta 10 50665 TMSB10 1.059 Cell death; cellular assembly and organization; cellular growth and proliferation
aldehyde dehydrogenase family 1, subfamily A2 116676 ALDH1A2 1.035 Cell death; cellular development; cellular growth and proliferation
syntaxin 1B2 24923 STX1B2 1.032 Others/unclassified
aldolase A 24189 ALDOA -1.013 Others/unclassified
prosaposin 25524 PSAP -1.031 Cell cycle; cell death; cellular growth and proliferation; biomolecule metabolism; molecular transport; post-translational modification
vesicle-associated membrane protein 2 24803 VAMP2 -1.031 Cell death; cell signaling; cellular assembly and organization; cellular movement; molecular transport
Rous sarcoma oncogene 83805 SRC -1.070 Biomolecule metabolism; cell cycle; cell death; cell morphology; cell signaling; cellular assembly and organization; cellular development; cellular growth and proliferation; cellular movement; DNA replication, recombination, and repair; gene transcription; molecular transport; post-translational modification
acetylcholinesterase 83817 ACHE -1.072 Cell death; cell morphology; cell signaling; cellular assembly and organization; cellular development; cellular growth and proliferation; DNA replication, recombination, and repair; post-translational modification; protein synthesis
Max interacting protein 1 25701 MXI1 -1.120 Cell morphology; cellular growth and proliferation; gene transcription
betacellulin 64022 BTC -1.137 Cell cycle; cell death; cell signaling; cellular development; cellular growth and proliferation; cellular movement; DNA replication, recombination, and repair
argininosuccinate synthetase 25698 ASS1 -1.165 Others/unclassified
nuclear receptor subfamily 1, group D, member 2 259241 NR1D2 -1.170 Gene transcription
stannin 29140 SNN -1.242 Cell death
phosphatase and tensin homolog* 50557 PTEN -1.272 Cell cycle; cell death; cell morphology; cell signaling; cellular assembly and organization; cellular development; cellular growth and proliferation; cellular movement; gene transcription inflammation; biomolecule metabolism; molecular transport; post-translational modification
solute carrier family 17 (sodium-dependent inorganic phosphate cotransporter), member 7* 116638 SLC17A7 -1.287 Cell signaling
carnitine palmitoyltransferase 1, liver* 25757 CPT1A -1.291 Biomolecule metabolism
isopentenyl-diphosphate delta isomerase* 89784 IDI1 -1.294 Others/unclassified
LIM homeobox protein 5 124451 LHX5 -1.317 Others/unclassified
calpain, small subunit 1* 29156 CAPNS1 -1.318 Cell death; cellular assembly and organization; cellular growth and proliferation; cellular movement
filaggrin 24641 FLG -1.325 Cellular assembly and organization
microsomal glutathione S-transferase 1 171341 MGST1 -1.329 Biomolecule metabolism
protein tyrosine phosphatase, nonreceptor type substrate 1* 25528 SIRPA -1.330 Cell death; cell morphology; cell signaling; cellular assembly and organization; cellular development; cellular growth and proliferation; cellular movement; gene transcription; inflammation
neurofibromatosis 2 25744 NF2 -1.333 Cell death; cellular growth and proliferation; cellular movement
Notch gene homolog 3 (Drosophila)* 56761 NOTCH3 -1.335 Cellular development; gene transcription
killer cell lectin-like receptor subfamily B member 1B* 25192 KLRB1 -1.338 Cell death
basic transcription element binding protein 1* 117560 KLF9 -1.339 Gene transcription
regulating synaptic membrane exocytosis 1* 84556 RIMS1 -1.353 Cell morphology; cell signaling; cellular assembly and organization; cellular movement; molecular transport
transglutaminase 1* 60335 TGM1 -1.376 Biomolecule metabolism; cell death; cellular development; post-translational modification
translocase of inner mitochondrial membrane 23 homolog (yeast)* 54312 IMM23 -1.393 Others/unclassified
lysozyme 25211 LYZ -1.399 Others/unclassified
solute carrier family 25 (mitochondrial carrier; oxoglutarate carrier), member 11* 64201 SLC25A11 -1.407 Others/unclassified
hydroxysteroid 11-beta dehydrogenase 1* 25116 HSD11B1 -1.407 Cellular development; cellular growth and proliferation
potassium intermediate/small conductance calcium-activated channel, subfamily N, member 3* 54263 KCNN3 -1.412 Cell signaling
B-cell leukemia/lymphoma 2* 24224 BCL2 -1.413 Cell cycle; cell death; cell morphology; cell signaling; cellular assembly and organization; cellular development; cellular growth and proliferation; cellular movement; cellular response to therapeutics; DNA replication, recombination, and repair; biomolecule metabolism; gene transcription; inflammation; molecular transport; post-translational modification
hyperpolarization-activated, cyclic nucleotide-gated K+ 4* 59266 HCN4 -1.415 Cell signaling
perforin 1 (pore-forming protein)* 50669 PRF1 -1.417 Cell death; cell morphology; cell signaling; DNA replication, recombination, and repair; inflammation
calcitonin/calcitonin-related polypeptide, alpha* 24241 CALCA -1.433 Cell morphology; cell signaling; cellular development; cellular growth and proliferation; cellular movement; DNA replication, recombination, and repair; gene transcription; inflammation
nuclear RNA export factor 1* 59087 NXF1 -1.436 Protein synthesis
fibroblast growth factor 14* 63851 FGF14 -1.439 Others/unclassified
solute carrier family 7 (cationic amino acid transporter,y+ system), member 3* 29485 SLC7A3 -1.455 Others/unclassified
ATP synthase, H+ transporting, mitochondrial F0 complex, subunit b, isoform 1* 171375 ATP5F1 -1.456 Cellular growth and proliferation
septin 3* 56003 SEPT3 -1.468 Others/unclassified
cholinergic receptor, nicotinic, beta polypeptide 2 (neuronal)* 54239 CHRNB2 -1.469 Cell signaling
wild-type p53-induced gene 1* 64394 ZMAT3 -1.480 Cell death; cellular growth and proliferation; DNA replication, recombination, and repair
glutamate receptor, ionotropic, kainate 4* 24406 GRIK4 -1.482 Cell signaling
Kruppel-like factor 15* 85497 KLF15 -1.488 Gene transcription
aquaporin 6* 29170 AQP6 -1.496 Others/unclassified
unc-5 homolog A (C. elegans)* 60629 UNC5A -1.498 Cell death
integrin beta 4* 25724 ITGB4 -1.502 Cell death; cell morphology; cellular assembly and organization; cellular development; cellular growth and proliferation; cellular movement; gene transcription
peptidylprolyl isomerase A* 25518 PPIA -1.526 Cell death; cellular development; cellular growth and proliferation; cellular movement; DNA replication, recombination, and repair; inflammation; post-translational modification
FXYD domain-containing ion transport regulator 6* 63847 FXYD6 -1.562 Others/unclassified
ribosomal protein L37* 81770 RPL37 -1.579 Others/unclassified
RT1 class II, locus Da* 294269 HLA-DRA -1.580 Cell signaling; inflammation
FK506 binding protein 12-rapamycin-associated protein 1* 56718 FRAP1 -1.596 Biomolecule metabolism; cell cycle; cell death; cell morphology; cell signaling; cellular development; cellular growth and proliferation; cellular movement; gene transcription; inflammation; post-translational modification; protein synthesis
eukaryotic translation elongation factor 1 alpha 1* 171361 EEF1A1 -1.618 Cell death; protein synthesis
glutathione S-transferase theta 1* 25260 GSTT1 -1.622 Biomolecule metabolism
solute carrier family 4, member 1* 24779 SLC4A1 -1.630 Cell death; cellular growth and proliferation
fibroblast growth factor 17* 29368 FGF17 -1.642 Cellular growth and proliferation
forkhead box M1* 58921 FOXM1 -1.646 Cell cycle; cell death; cell morphology; cellular growth and proliferation; gene transcription
ATPase, class II, type 9A* 84011 ATP9A -1.649 Others/unclassified
phosphorylase kinase, gamma 2 (testis)* 140671 PHKG2 -1.679 Biomolecule metabolism; post-translational modification
arrestin, beta 1* 25387 ARRB1 -1.683 Cellular movement
rhoB gene* 64373 RHOB -1.684 Cell death; cell morphology; cell signaling; cellular assembly and organization; cellular development; cellular growth and proliferation; cellular movement; gene transcription
par-3 (partitioning defective 3) homolog (C. elegans)* 81918 PARD3 -1.686 Cell morphology; cell signaling; cellular assembly and organization; cellular development; gene transcription
ribosomal protein L21* 79449 RPL21 -1.687 Others/unclassified
RAS protein-specific guanine nucleotide-releasing factor 1* 192213 RASGRF1 -1.715 Cell morphology; cell signaling; cellular growth and proliferation; gene transcription
acetylcholinesterase* 83817 ACHE -1.741 Cell death; cell morphology; cell signaling; cellular assembly and organization; cellular development; cellular growth and proliferation; DNA replication, recombination, and repair; post-translational modification; protein synthesis
ribosomal protein L18* 81766 RPL18 -1.741 Others/unclassified
granzyme M (lymphocyte met-ase 1)* 29252 GZMM -1.745 Cell death
Bcl2-associated X protein* 24887 BAX -1.746 Cell cycle; cell death; cell morphology; cellular assembly and organization; cellular development; cellular growth and proliferation; cellular response to therapeutics; DNA replication, recombination, and repair; free radical scavenging; inflammation; biomolecule metabolism; molecular transport; protein synthesis
legumain* 63865 LGMN -1.749 Cellular movement
RAB6A, member RAS oncogene family* 84379 RAB6A -1.752 Cellular assembly and organization
matrix metalloproteinase 16* 65205 MMP16 -1.763 Cellular movement; inflammation
olfactory receptor 226* 65140 OR6A2 -1.770 Others/unclassified
mitogen-activated protein kinase 14* 81649 MAPK14 -1.775 Biomolecule metabolism; cell cycle; cell death; cell morphology; cellular development; cellular growth and proliferation; cellular movement; gene transcription; inflammation; post-translational modification
c-mer protooncogene tyrosine kinase* 65037 MERTK -1.780 Biomolecule metabolism; cell death; cell morphology; cell signaling; cellular development; cellular growth and proliferation; post-translational modification
clathrin, heavy polypeptide (Hc)* 54241 CLTC -1.786 Cell morphology; cellular growth and proliferation
transient receptor potential cation channel, subfamily C, member 4* 84494 TRPC4 -1.793 Others/unclassified
carnitine palmitoyltransferase 1b* 25756 CPT1B -1.795 Biomolecule metabolism
PDZ and LIM domain 1 (elfin)* 54133 PDLIM1 -1.796 Gene transcription
fatty acid amide hydrolase* 29347 FAAH -1.798 Cell death; inflammation
fatty acid binding protein 4, adipocyte* 79451 FABP4 -1.820 Gene transcription
protein tyrosine phosphatase, nonreceptor type 12* 117255 PTPN12 -1.826 Biomolecule metabolism; cell morphology; cell signaling; cellular assembly and organization; cellular development; cellular movement; post-translational modification
potassium voltage-gated channel, Shab-related subfamily, member 2* 117105 KCNB2 -1.834 Others/unclassified
glutathione S-transferase, pi 2* 29438 GSTP1 -1.856 Cell death; cellular growth and proliferation; biomolecule metabolism; gene transcription
Fc receptor, IgG, alpha chain transporter* 29558 FCGRT -1.870 Cell signaling; molecular transport
voltage-gated channel like 1* 266760 VGCNL1 -1.871 Others/unclassified
prostaglandin D2 synthase* 25526 PTGDS -1.888 Cell death; cell morphology
MAP-kinase activating death domain* 94193 MADD -1.934 Cell death; cellular growth and proliferation
neogenin* 81735 NEO1 -1.935 Cellular growth and proliferation; cellular movement; gene transcription
calbindin 1* 83839 CALB1 -1.950 Cell death; cell morphology; cell signaling
guanylate cyclase 2e* 79222 GUCY2D -1.957 Others/unclassified
adrenal secretory serine protease precursor* 64565 TMPRSS11D -1.971 Others/unclassified
ubc2e ubiquitin-conjugating enzyme* 641452 Ube2d2 -1.986 Others/unclassified
neuroblastoma, suppression of tumorigenicity 1* 50594 NBL1 -2.003 Cellular movement
collagen, type 1, alpha 1* 29393 COL1A1 -2.023 Cell morphology; cellular growth and proliferation; cellular movement
homeodomain interacting protein kinase 3* 83617 HIPK3 -2.034 Biomolecule metabolism; cell signaling; post-translational modification
intercellular adhesion molecule 1* 25464 ICAM1 -2.052 Cell death; cell morphology; cell signaling; cellular development; cellular growth and proliferation; cellular movement; inflammation
cyclin-dependent kinase 5* 140908 CDK5 -2.134 Biomolecule metabolism; cell death; cell morphology; cell signaling; cellular assembly and organization; cellular development; cellular growth and proliferation; cellular movement; molecular transport; post-translational modification
ATPase, Cu2+ transporting, beta polypeptide* 24218 ATP7B -2.137 Cell death
Jun D protooncogene* 24518 JUND -2.159 Cell death; cell morphology; cellular growth and proliferation; gene transcription
neurexophilin 3* 59315 NXPH3 -2.168 Others/unclassified
heterogeneous nuclear ribonucleoprotein methyltransferase-like 3 (S. cerevisiae)* 89820 PRMT3 -2.171 Biomolecule metabolism; post-translational modification
midline 1* 54252 MID1 -2.180 Cellular assembly and; organization
ATP-binding cassette, subfamily C (CFTR/MRP), member 1* 24565 ABCC1 -2.191 Cell death; cellular movement; inflammation; biomolecule metabolism; molecular transport
prothymosin alpha* 29222 PTMA -2.313 Cell cycle; cell death; cellular development; cellular growth and proliferation; gene transcription
ubiquilin 1* 114590 UBQLN1 -2.322 Others/unclassified
similar to Leydig cell tumor 10 kDa protein* 288913 C19ORF53 -2.346 Others/unclassified
Inhibitor of DNA binding 2, dominant negative helix-loop-helix protein* 25587 ID2 -2.357 Cell cycle; cell death; cell morphology; cellular development; cellular growth and proliferation; cellular movement; gene transcription
lectin, galactose binding, soluble 1* 56646 LGALS1 -2.385 Cell cycle; cell death; cell morphology; cellular development; cellular growth and proliferation; cellular movement gene transcription; inflammation; post-translational modification
calreticulin* 64202 CALR -2.401 Cell cycle; cell death; cellular assembly and organization; cellular development; cellular growth and proliferation; cellular movement; gene transcription; post-translational modification
carbonic anhydrase 5* 54233 CA5A -2.427 Inflammation
mitogen-activated protein kinase 13* 29513 MAPK13 -2.438 Cell death; cell morphology; gene transcription; inflammation
cytochrome P450, family 19, subfamily a, polypeptide 1* 25147 CYP19A1 -2.440 Cell death; cell morphology; cellular development; cellular growth and proliferation; cellular movement; gene transcription; biomolecule metabolism
nucleobindin 1* 84595 NUCB1 -2.513 Others/unclassified
tropomodulin 2* 58814 TMOD2 -2.563 Cell signaling; cellular assembly and organization
apolipoprotein B editing complex 1* 25383 APOBEC1 -2.655 Others/unclassified
solute carrier family 7 (cationic amino acid transporter, y+ system), member 7* 83509 SLC7A7 -2.720 Cell signaling; cellular growth and proliferation
hsp70-interacting protein* 246146 HSPBP1 -2.772 Post-translational modification
Rsec5 protein* 171455 EXOC2 -2.779 Cell signaling; molecular transport
MAD homolog 9 (Drosophila)* 85435 SMAD9 -2.815 Others/unclassified
ERM-binding phosphoprotein* 59114 SLC9A3R1 -2.851 Cell signaling; cellular growth and proliferation
low-density lipoprotein receptor-related protein 3* 89787 LRP3 -2.869 Others/unclassified
Rab geranylgeranyl transferase, a subunit* 58983 RABGGTA -2.890 Biomolecule metabolism; post-translational modification
cadherin EGF LAG seven-pass G-type receptor 2* 83465 CELSR2 -2.923 Others/unclassified
tropomyosin 4* 24852 TPM4 -3.019 Cellular movement
cytochrome P450, family 2, subfamily e, polypeptide 1* 25086 CYP2E1 -3.161 Cell death; biomolecule metabolism
CEA-related cell adhesion molecule 9* 116711 CEACAM9 -3.207 Others/unclassified
discoidin domain receptor family, member 2* 83573 DDR2 -3.253 Others/unclassified
phosphoglucomutase 1* 24645 PGM1 -3.308 Others/unclassified
gamma-glutamyl carboxylase* 81716 GGCX -3.455 Post-translational modification
Cplx1 complexin 1* 64832 CPLX1 -3.464 Cell signaling; cellular assembly; and organization; cellular movement; molecular transport
septin 9* 83788 SEPT9 -3.781 Cell cycle; protein synthesis
matrix metallopeptidase 8* 63849 MMP8 -3.785 Cell death, cellular movement
thyroid hormone receptor interactor 10* 116717 TRIP10 -4.232 Cell death
carboxylesterase 3* 113902 CES1 -4.325 Others/unclassified
cyclin-dependent kinase 2* 362817 CDK2 -4.458 Biomolecule metabolism; cell cycle; cell death; cell morphology; cellular development; cellular growth and proliferation; DNA replication, recombination, and repair; gene transcription; post-translational modification
monoamine oxidase B* 25750 MAOB -4.873 Cell death
solute carrier family 6 (neurotransmitter transporter, betaine/GABA), member 12* 50676 SLC6A12 -5.287 Others/unclassified
peroxiredoxin 1* 117254 PRDX1 -5.360 Cell death; cell morphology; cellular growth and proliferation; gene transcription; post-translational modification
ADP-ribosylation factor 4* 79120 ARF4 -5.374 Others/unclassified
glypican 3* 25236 GPC3 -5.638 Cell death; cellular growth and proliferation
protein kinase C, delta binding protein* 85332 PRKCDBP -6.059 Others/unclassified
cd86 antigen* 56822 CD86 -6.118 Cell signaling; cellular development; cellular growth and proliferation; cellular movement; gene transcription; inflammation
Sjogren syndrome antigen B* 81783 SSB -6.131 Gene transcription; protein synthesis
secretoglobin, family 2A, member 1* 25010 PSBP1 -6.227 Others/unclassified
procollagen, type I, alpha 2* 84352 COL1A2 -7.076 Cell morphology
A kinase (PRKA) anchor protein 1* 114124 AKAP1 -8.502 Cell death
methionine adenosyltransferase II, alpha* 171347 MAT2A -9.474 Others/unclassified
FXYD domain-containing ion transport regulator 1* 58971 FXYD1 -9.644 Cellular growth and proliferation; gene transcription
CD24 antigen* 25145 CD24 -11.299 Cell death; cell morphology; cell signaling; cellular assembly and organization; cellular development; cellular growth and proliferation; cellular movement
nuclear receptor subfamily 1, group I, member 2* 84385 NR1I2 -11.958 Biomolecule metabolism; gene transcription; inflammation; molecular transport; post-translational modification
glial cell line derived neurotrophic factor family receptor alpha 1* 25454 GFRA1 -17.708 Cell death; cell morphology; cellular development; cellular; growth and proliferation; cellular movement
cell division cycle 25 homolog A (S. cerevisiae)* 171102 CDC25A -19.859 Cell cycle; cell death; cell morphology; cellular growth and proliferation; dna replication, recombination, and repair
5-hydroxytryptamine (serotonin) receptor 5B* 79247 HTR5B -22.285 Others/unclassified
NSFL1 (p97) cofactor (p47)* 83809 NSFL1C -22.400 Others/unclassified
membrane and microfilament-associated protein p58* 207121 RGD: 727794 -49.533 Cellular assembly and organization
*

The change in expression was significant compared with untreated control (P < .05). Gene names in italics are common in both panels of T + E2-treated LP and T + DES-treated VP.

Image plots (Figure 2, A, and B1 and B2) showed the up- and downregulated genes in the T + E2-induced LP dysplasia panel (253 genes), respectively. Although genes in this panel were distributed among 27 IPA networks, two major networks with the highest relevancy scores were identified: one related to cell morphology, cellular growth, proliferation, and movement (Figure 2C), and the other related to apoptosis and cell signaling (Figure 2D). From these two networks, six genes were selected for post hoc confirmation by real-time q-PCR (Figure 3). All six genes showed the predicted expression patterns. Significant differences (P < .01/.05) were observed between the transcript levels in the T + E2-treated and -untreated LPs, whereas no significant differences (P > .05) in the transcript levels were observed between the treated and untreated VP groups. These genes are the CCAAT/enhancer binding protein-delta (Cebpd), the tumor protein Tp53, MAD homolog 7 (Smad7), homeobox A1 (Hoxa1), killer cell lectin-like receptor subfamily B member 1 (Klrb1), and neurexophilin 3 (Nxph3). Notably, the q-PCR data correlated well with those obtained by microarray analyses (Figure W1A). T + E2-response genes found in VP, in contrast to those found in the LP, were not mapped to any particular IPA networks (data not shown).

Figure 2.

Figure 2

Heat maps and gene interaction networks of differentially expressed genes found exclusively in the LP dysplasia following T + E2 treatment. Red and green denote upregulated and downregulated expression, respectively, as compared with the overall gene's mean value normalized to the universal rat reference RNA. Columns represent data from a single prostate sample, and rows correspond to a single gene probe. (A) A single cluster of upregulated genes identified in the T + E2 LP dysplasia, marked with pink (left panel); a selected region (blue box) of this cluster is enlarged (right panel). (B1 and B2) Two separate downregulated gene clusters observed only in T + E2 LP dysplasia, marked with pink (left panels); selected clusters are magnified (right panels). (C and D) Two representative gene interaction networks (with the highest relevancy scores) generated by IPA analysis from the differentially expressed genes in the T + E2 LP dysplasia panel. Green indicates downregulated; red, upregulated. Genes bordered in red were validated by real-time q-PCR. See Figure 1 for key to IPA network.

Figure 3.

Figure 3

Post hoc real-time q-PCR analyses of selected genes in the T + E2 LP dysplasia panel. Data were normalized to the levels of Rpl19. Bars indicate standard deviations (SD) of three to five animals in each treatment group. *P < .05, **P < .01 by one-way ANOVA with Tukey post hoc analysis.

Image plots illustrated distinctive upregulated (Figure 4A) and downregulated (Figure 4, B1 and B2) genes in the T + DES-induced VP dysplasia panel. In addition, a unique set of genes was exclusively repressed by T + DES in the VP harboring dysplasia (Figure 4B2). These genes were also underexpressed in LPs compared with the untreated VPs. Genes in the T + DES-treated VP panel (198 genes) mapped primarily to two major networks related to 1) apoptosis, cellular development, growth, and proliferation and 2) estrogen signaling, as they exhibited the highest relevancy scores (Figure 4, C and D, respectively). From the two networks, nine genes were selected for post hoc confirmation by q-PCR analyses that correlated well with those obtained by microarrays (Figure W1B). All nine genes showed significant increases in transcript levels (P < .01/.05) in the VP with T + DES treatment and exhibited little or no change in expression (P > .05) in the LPs of the treated group compared with untreated controls (Figure 5). They include Cebpd, Tp53, v-maf musculoaponeurotic fibrosarcoma oncogene homolog B (Mafb), Hoxa1, breast cancer anti-estrogen resistance 1 (Bcar1), plasminogen activator urokinase (Plau), thyroid hormone receptor alpha (Thra), glutathione peroxidase 3 (Gpx3), and v-src sarcoma viral oncogene homolog (Src).

Figure 4.

Figure 4

Heat maps and gene interaction networks of differentially expressed genes found uniquely in the VP dysplasia following T + DES treatment. (A) A single cluster of upregulated genes identified only in the T + DES VP dysplasia, marked with pink (left panel), and the selected cluster enlarged (right panel). (B1 and B2) Clusters of downregulated genes in the VP but not in the LP following T + DES exposure. Interestingly, a single cluster (B2, left panel) corresponds to the downregulated genes in the T + DES-treated VP and is also underexpressed in both untreated and treated LPs. A selected region (blue box) of this cluster is enlarged (right panel). (C and D) Two representative gene interaction networks (with the highest relevancy scores) generated by IPA analysis from the differentially expressed genes in the T + DES VP dysplasia panel. Green indicates downregulated; red, upregulated. Genes bordered with red were validated by real-time q-PCR. See Figure 1 for key to IPA network.

Figure 5.

Figure 5

Post hoc validation of selected genes in the T + DES VP dysplasia panel by real-time q-PCR. The data were normalized to the levels of Rpl19. Bars indicated standard deviations (SD) of three to five animals in each treatment group. *P < .05, **P < .01 by one-way ANOVA with Tukey post hoc analysis.

Only 32 genes were found to be common in both dysplasia-related panels (Figure 1C), and 23 (of 32) genes formed a unique gene network when mapped by IPA (Figure 1D). We postulate that this gene network is central to E2- or DES-induced dysplasia in the LP or VP.

Identification of Estrogen-induced Differentially Expressed Genes That Are Not Related to Dysplasia in Rat LP or VP

We also identified two sets of hormone-induced genes that are not related to dysplasia. These genes exhibited differential expression in the VP following T + E2 treatment (14 genes; Figure 1C, left bottom) and in the LP following T + DES treatment (190 genes; Figure 1C, right bottom). They are published in Tables W2 and W3.

Genes Insensitive to the T + E2 and T + DES Treatments

Two sets of 156 and 80 genes were identified as insensitive in the LP and/or VP to T + E2 or T + DES treatment, respectively (Figure 6, A–C; Tables W4 and W5). Forty-one genes were found in both hormone-insensitive panels and mapped to an IPA network (Figure 6D) that includes the androgen receptor (Ar), a key regulator of prostate function.

Figure 6.

Figure 6

Heat maps of genes insensitive to (A) T + E2 or (B) T + DES. (C) Venn diagram showing the number of genes insensitive to T + E2 and/or T + DES treatment. Note that 41 genes are found in common in both hormone-insensitive panels and mapped to an IPA network (D) that includes the Ar, a key regulator of prostate function. Gray indicates no change in gene expression levels compared with those in untreated counterparts. See Figure 1 for key to IPA network.

Discussion

Exposure of the human prostate to estrogen may increase as men age [2–4] or with increased exposure to dietary estrogens such as DES or zeranol residues in meat, bisphenol A from food containers, or phytoestrogens [9]. The NBL rat provides a relevant model for elucidating mechanisms underlying carcinogenesis caused by such exposures, such as dysplasia and PCa, that consistently developed in the LP or the VP following chronic T-supported treatment with E2 or DES, respectively [15,18–20]. In the present study, we assessed transcriptional profiles elicited by the xenoestrogen DES in comparison with those of the endogenous hormone E2 and observed prostatic lobe-specific differential responses to the two estrogens in NBL rats. Hierarchical clustering revealed that the T + E2 treatment principally affects the LP gene expression profile whereas the T + DES targets that of the VP. Two distinct panels of dysplasia-related genes were identified: the T + E2 LP panel (253 genes) and the T + DES VP panel (198 genes), with only 32 overlapping genes. These findings indicate that the two estrogens alter gene expression profiles only in the prostatic lobe that develops dysplasia following each of the hormonal treatments. These findings imply a functional divergence of the two estrogens with respect to their oncogenic actions in the rat prostate. Thus, endocrine disruption of gene expression and dysplasia induction of the two estrogens are categorically different [9], although they may share some biologic convergence [30].

The IPA analysis identified distinct gene interaction networks based on differential expression profiles. It classified genes into functional categories and suggested a possible mechanistic linkage among the differentially expressed genes. We also used this tool to help prioritize genes for confirmation. The T + E2 treatment altered the expression of LP genes primarily in two major networks: one related to cell morphology, cellular growth, proliferation, and movement, and the other related to apoptosis and cell signaling. Exposure to T + DES also affected expression of genes in a similar network associated with apoptosis, cellular development, growth, and proliferation. Intriguingly, it also altered the expression of a set of genes linked to E2, a finding that may explain why, after T + DES treatment, gene expression profiles in the VP shifted closer to those in the treated and untreated LPs, which are under the influence of natural estrogen. By first mapping genes to the IPA network before selecting genes for confirmation, we were able to validate most of the expression changes in the microarrays with real-time q-PCR. In this regard, we confirmed three genes (Cebpd, Hoxa1, and Tp53) common in the two dysplasia panels. Importantly, these three genes were upregulated only in the prostatic lobes that developed dysplasia, i.e., the T + E2-treated LP and the T + DES-treated VP. Other lobe-specific genes that were confirmed were Smad7, Klb1, and Nxph3 in the T + E2-treated LP and Mafb, Src, Bcar1, Gpx3, Plau, and Thra in T + DES-treated VP. Our findings suggest that these gene expression changes induced by T + E2/DES treatment could be causative factors of dysplasia development. However, it remains possible that these gene expression changes are simply part of the neoplastic transformation process. Future studies employing laser capture-microdissected samples to establish the temporal relationship between change in gene expression pattern and evolution of dysplasia may shed new light on the cause-effect link between the two.

Among all these validated genes, Cebpd, Hoxa1, and Tp53 are the ones upregulated in common in the two dysplasia panels. CCAAT/enhancer binding proteins (C/EBP) are a highly conserved family of basic leucine zipper transcription factors; six members (C/EBP-alpha to -zeta) have been identified to date. This protein family plays a critical role in cell proliferation, apoptosis, and inflammation, depending on the cell type and specific physiological stress [31]. The precise functional role of C/EBP-delta, in particular, in normal prostate biology and PCa remains poorly understood. Cebpd was shown to be an androgen-repressed gene in the normal rat prostate [32,33], supporting its role as an apoptotic mediator and/or a negative regulator of cell proliferation. In a human androgen-dependent PCa xenograft, androgen withdrawal, however, resulted in a decline in the expression of C/EBP-delta [32]. Most published studies focus on the androgenic regulation of C/EBP-delta; this is, therefore, the first report of the association of estrogen-mediated upregulation of C/EBP-delta gene expression with the early phase of prostate carcinogenesis. A recent study showed overexpression of C/EBP-beta, another member of the family, in proliferative inflammatory atrophy in human prostate specimens, suggesting its role in the inflammation-associated carcinogenesis in the prostate [34]. In microarray data, we observed downregulation of C/EBP-alpha (Cebpa) only in T + E2-treated dysplastic LPs, corroborating its putative suppressive function in epithelial tumorigenesis [35]. Together, these findings raise the possibility that an orchestrated deregulation of C/EBP-family transcripts is involved in the pathogenesis of PCa in humans and rodents.

Homeobox gene (HOX) is a family of transcription factors related to growth and development. In rodents and humans, 39 members of the HOX gene family have been assigned to four gene clusters (A–D) and are numbered according to their expression along the anterior- posterior axis. Hox13 was shown to be involved in cell differentiation during normal prostate development [36], whereas upregulations of HOXC have been observed in PCa cells [37,38]. Knockdown of HOXC6 gene expression led to apoptosis of PCa cells [39], and overexpression of the HOXC8 gene correlated with the loss of tumor differentiation in human PCa [37]. Increased expression of the HOXA1 transcript was detected in human clinical cervical cancer samples and in several cervical cell lines compared with expression in normal tissues [40,41]. Our data suggest that ectopic expression of Hoxa1 is related to the evolution of dysplasia in both LP and VP and that the detailed roles of Hoxa1 in PCa require further study.

Tp53 is a well-known tumor-suppressive transcription factor and acts as a gatekeeper in signaling pathways involved in monitoring cellular stress such as DNA damage and in the determination of congruous responses (DNA repair, cell growth arrest, or apoptosis) to specific physiological stress. Previous studies demonstrated that mutation of Tp53 is not necessarily as late an event in prostate carcinogenesis as previously reported; it is also frequently found in high-grade prostate intraepithelial neoplasia, a precursor lesion of PCa [42,43]. In this regard, we observed upregulation of Tp53 gene expression in dysplastic rat prostate glands treated with sex hormone, implying a role in early prostate tumorigenesis, but whether these glands express wild-type or mutated Tp53 is not known. Alternatively, the induction of Tp53 expression may be a compensatory response to cellular stress imposed by hormonal changes. We recently demonstrated that T + E2 treatment induced oxidative and nitrosative stress, accompanied by DNA and protein damages, specifically in the LP, which is susceptible to dysplasia/cancer induction [17].

Smad7, an inhibitory Smad, is rapidly induced by TGF-β, thereby creating a negative feedback loop to a variety of TGF-β signaling responses, including proliferation, differentiation, apoptosis, inflammation, tissue remodeling, angiogenesis, and cell adhesion [44]. Disruption of the TGF-β signaling cascade by aberrant overexpression of this negative modulator leads to increased tumorigenicity in colon cancer cells by blocking TGF-β-mediated growth inhibition and apoptosis [45]. In contrast, Smad7 has been shown to mediate apoptosis induced by TGF-β in PCa cells [46], perhaps through crosstalking with other cellular signaling cascades such as the p38 mitogen-activated protein kinase [47] and β-catenin/Wnt [48] pathways. In our study, upregulation of Smad7 in the dysplastic LP (also in the T + DES VP microarray data) suggests perturbations of TGF-β feedback regulation and thus the loss of balance between cell proliferation and apoptosis in the early phase of prostate carcinogenesis.

A particularly interesting gene network found in the T + DES-treated VP (Figure 4, B1 and B2) is related to E2 signaling. Members include Src, Bcar1, and Plau. Src, a nonreceptor protein tyrosine kinase, triggers the nongenomic estrogen signaling leading to activation of ERK1/2 and cell proliferation in prostate epithelial cells [49,50]. It remains to be determined if the downregulation of Src by DES represents a classic negative feedback loop. The Bcar1 interacts and modulates Src activity [51]. An increase in Bcar1 gene expression, as observed in the rat dysplastic VP, has also been reported in human PCa [52]. The coordinated dysregulation of both Src (downregulation) and Bcar1 (upregulation) may reflect a disruption of the Src-dependent signaling pathway during the early development of PCa, a notion further supported by the altered expression of Plau, a downstream target of Src [53]. Interestingly, both Bcar1 and Plau are involved in cell survival and migration [54,55].

Our group recently demonstrated significant oxidative stress and inflammation in the T + E2-treated NBL rat LP [17,56]. The microarray data show a relatively small fraction of genes related to redox homeostasis and immune response, possibly due to inherent limitations of the type and coverage size (about 3800 genes) of the array chips we selected. However, T + E2 treatment was found to upregulate Klrb1 in the LP. This gene, also known as NKR-P1A/CD161 in humans, encodes a natural killer (NK) cell C-type lectin-like receptor that regulates NK cell functions. KLRB1 is expressed not only in NK cells but also in immune cell types, such as T cells, monocytes, and dendritic cells [57]. Thus, it is likely that increased expression of Klrb1 in LP with T + E2 treatment is due to the infiltration of immune cells [17].

The observation of a dramatic induction of Gpx3 in the VP dysplasia panel, as shown by microarray (>17-fold) and q-PCR (>70-fold), is consistent with our previous report of a marked elevation of GPX enzyme activity and lipid peroxidation in this lobe after T + DES treatment [56]. Hence, the activation in this glutathione-associated detoxification enzyme might be a response to hormone-induced oxidative stress in the gland or represent a cytoprotective mechanism, adapted by dysplastic cells, against oxidative insults. An elevation of GPX3 expression, but not of GPX1 and GPX2, was found in precancerous lesions of NKX3.1 mutant mouse prostate [58]. Taken together, these findings suggest that GPX3 is specifically involved in early prostatic transformation that has a mechanistic link to oxidative stress.

In summary, we showed that genes such as Cebpd, Tp53, and Hoxa1 are at the core of disrupted biologic networks related to hormoneinduced dysplasia. Unbiased gene profiling clearly demonstrated differential susceptibility of the LP and VP to natural and xenoestrogen with regard to alterations in gene expression and induction of dysplasia. Our data suggest more functional divergence between DES and E2 in the disruption of prostatic functions than that previously suspected. Methodologically, the combined utility of expression profiling and gene network mapping provides an instrumental platform for transcriptome studies.

Supplementary Material

Supplementary Figures and Tables
neo1001_0020SD1.pdf (161.1KB, pdf)

Abbreviations

C/EBP

CCAAT/enhancer binding protein

DES

diethylstilbestrol

E2

17β-estradiol

LP

lateral prostate

NBL

Noble

PCa

prostate cancer

PCR

polymerase chain reaction

VP

ventral prostate

Footnotes

2

This article refers to supplementary materials, which are designated by Tables W1, W2, W3, W4, and W5 and Figure W1 and are available online at www.neoplasia.com.

References

  • 1.Taplin ME, Ho SM. Clinical review 134: the endocrinology of prostate cancer. J Clin Endocrinol Metab. 2001;86:3467–3477. doi: 10.1210/jcem.86.8.7782. [DOI] [PubMed] [Google Scholar]
  • 2.Ho SM, Leung YK, Chung I. Estrogens and antiestrogens as etiological factors and therapeutics for prostate cancer. Ann NY Acad Sci. 2006;1089:177–193. doi: 10.1196/annals.1386.005. [DOI] [PubMed] [Google Scholar]
  • 3.Carruba G. Estrogens and mechanisms of prostate cancer progression. Ann NY Acad Sci. 2006;1089:201–217. doi: 10.1196/annals.1386.027. [DOI] [PubMed] [Google Scholar]
  • 4.Bosland MC. Sex steroids and prostate carcinogenesis: integrated, multifactorial working hypothesis. Ann NY Acad Sci. 2006;1089:168–176. doi: 10.1196/annals.1386.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Leav I, Lau KM, Adams JY, McNeal JE, Taplin ME, Wang J, Singh H, Ho SM. Comparative studies of the estrogen receptors β and α and the androgen receptor in normal human prostate glands, dysplasia, and in primary and metastatic carcinoma. Am J Pathol. 2001;159:79–92. doi: 10.1016/s0002-9440(10)61676-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Zhu X, Leav I, Leung YK, Wu M, Liu Q, Gao Y, McNeal JE, Ho SM. Dynamic regulation of estrogen receptor-β expression by DNA methylation during prostate cancer development and metastasis. Am J Pathol. 2004;164:2003–2012. doi: 10.1016/s0002-9440(10)63760-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Ellem SJ, Risbridger GP. Aromatase and prostate cancer. Minerva Endocrinol. 2006;31:1–12. [PubMed] [Google Scholar]
  • 8.Singleton DW, Khan SA. Xenoestrogen exposure and mechanisms of endocrine disruption. Front Biosci. 2003;8:s110–s118. doi: 10.2741/1010. [DOI] [PubMed] [Google Scholar]
  • 9.Leffers H, Naesby M, Vendelbo B, Skakkebaek NE, Jorgensen M. Oestrogenic potencies of zeranol, oestradiol, diethylstilboestrol, bisphenol-A and genistein: implications for exposure assessment of potential endocrine disrupters. Hum Reprod. 2001;16:1037–1045. doi: 10.1093/humrep/16.5.1037. [DOI] [PubMed] [Google Scholar]
  • 10.Epstein SS. The chemical jungle: today's beef industry. Int J Health Serv. 1990;20:277–280. doi: 10.2190/3MFD-569U-K87C-P8CM. [DOI] [PubMed] [Google Scholar]
  • 11.Herbst AL, Ulfelder H, Poskanzer DC. Adenocarcinoma of the vagina. Association of maternal stilbestrol therapy with tumor appearance in young women. N Engl J Med. 1971;284:878–881. doi: 10.1056/NEJM197104222841604. [DOI] [PubMed] [Google Scholar]
  • 12.Troisi R, Hatch EE, Titus-Ernstoff L, Hyer M, Palmer JR, Robboy SJ, Strohsnitter WC, Kaufman R, Herbst AL, Hoover RN. Cancer risk in women prenatally exposed to diethylstilbestrol. Int J Cancer. 2007;121:356–360. doi: 10.1002/ijc.22631. [DOI] [PubMed] [Google Scholar]
  • 13.Strohsnitter WC, Noller KL, Hoover RN, Robboy SJ, Palmer JR, Titus-Ernstoff L, Kaufman RH, Adam E, Herbst AL, Hatch EE. Cancer risk in men exposed in utero to diethylstilbestrol. J Natl Cancer Inst. 2001;93:545–551. doi: 10.1093/jnci/93.7.545. [DOI] [PubMed] [Google Scholar]
  • 14.Ho SM, Tang WY, Belmonte de Frausto J, Prins GS. Developmental exposure to estradiol and bisphenol A increases susceptibility to prostate carcinogenesis and epigenetically regulates phosphodiesterase type 4 variant 4. Cancer Res. 2006;66:5624–5632. doi: 10.1158/0008-5472.CAN-06-0516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Leav I, Ho SM, Ofner P, Merk FB, Kwan PW, Damassa D. Biochemical alterations in sex hormone-induced hyperplasia and dysplasia of the dorsolateral prostates of Noble rats. J Natl Cancer Inst. 1988;80:1045–1053. doi: 10.1093/jnci/80.13.1045. [DOI] [PubMed] [Google Scholar]
  • 16.Leav I, Merk FB, Kwan PW, Ho SM. Androgen-supported estrogenenhanced epithelial proliferation in the prostates of intact Noble rats. Prostate. 1989;15:23–40. doi: 10.1002/pros.2990150104. [DOI] [PubMed] [Google Scholar]
  • 17.Tam NN, Leav I, Ho SM. Sex hormones induce direct epithelial and inflammation-mediated oxidative/nitrosative stress that favors prostatic carcinogenesis in the Noble rat. Am J Pathol. 2007;171:1334–1341. doi: 10.2353/ajpath.2007.070199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Thompson CJ, Tam NN, Joyce JM, Leav I, Ho SM. Gene expression profiling of testosterone and estradiol-17β-induced prostatic dysplasia in Noble rats and response to the antiestrogen ICI 182,780. Endocrinology. 2002;143:2093–2105. doi: 10.1210/endo.143.6.8846. [DOI] [PubMed] [Google Scholar]
  • 19.Bosland MC, Ford H, Horton L. Induction at high incidence of ductal prostate adenocarcinomas in NBL/Cr and Sprague-Dawley Hsd:SD rats treated with a combination of testosterone and estradiol-17β or diethylstilbestrol. Carcinogenesis. 1995;16:1311–1317. doi: 10.1093/carcin/16.6.1311. [DOI] [PubMed] [Google Scholar]
  • 20.Ofner P, Bosland MC, Vena RL. Differential effects of diethylstilbestrol and estradiol-17β in combination with testosterone on rat prostate lobes. Toxicol Appl Pharmacol. 1992;112:300–309. doi: 10.1016/0041-008x(92)90200-c. [DOI] [PubMed] [Google Scholar]
  • 21.Smyth GK. Linear models and empirical Bayes methods for assessing differential expression in microarray experiments. Stat Appl Genet Mol Biol. 2004;3 doi: 10.2202/1544-6115.1027. Article 3. [DOI] [PubMed] [Google Scholar]
  • 22.Guo J, Sartor M, Karyala S, Medvedovic M, Kann S, Puga A, Ryan P, Tomlinson CR. Expression of genes in the TGF-β signaling pathway is significantly deregulated in smooth muscle cells from aorta of aryl hydrocarbon receptor knockout mice. Toxicol Appl Pharmacol. 2004;194:79–89. doi: 10.1016/j.taap.2003.09.002. [DOI] [PubMed] [Google Scholar]
  • 23.Karyala S, Guo J, Sartor M, Medvedovic M, Kann S, Puga A, Ryan P, Tomlinson CR. Different global gene expression profiles in benzo[a] pyrene- and dioxin-treated vascular smooth muscle cells of AHR-knockout and wild-type mice. Cardiovasc Toxicol. 2004;4:47–73. doi: 10.1385/ct:4:1:47. [DOI] [PubMed] [Google Scholar]
  • 24.Sartor M, Schwanekamp J, Halbleib D, Mohamed I, Karyala S, Medvedovic M, Tomlinson CR. Microarray results improve significantly as hybridization approaches equilibrium. Biotechniques. 2004;36:790–796. doi: 10.2144/04365ST02. [DOI] [PubMed] [Google Scholar]
  • 25.Sartor MA, Tomlinson CR, Wesselkamper SC, Sivaganesan S, Leikauf GD, Medvedovic M. Intensity-based hierarchical Bayes method improves testing for differentially expressed genes in microarray experiments. BMC Bioinformatics. 2006;7:538. doi: 10.1186/1471-2105-7-538. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Benjamini YHY. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Stat Soc. 1995;57:289–300. [Google Scholar]
  • 27.Daly-Burns B, Alam TN, Mackay A, Clark J, Shepherd CJ, Rizzo S, Tatoud R, O'Hare MJ, Masters JR, Hudson DL. A conditionally immortalized cell line model for the study of human prostatic epithelial cell differentiation. Differentiation. 2007;75:35–48. doi: 10.1111/j.1432-0436.2006.00113.x. [DOI] [PubMed] [Google Scholar]
  • 28.Rozen S, Skaletsky HJ. Primer3. 1998 Available at: http://www-genome.wi.mit.edu/genome_software/other/primer3.html.
  • 29.Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 2001;25:402–408. doi: 10.1006/meth.2001.1262. [DOI] [PubMed] [Google Scholar]
  • 30.Buterin T, Koch C, Naegeli H. Convergent transcriptional profiles induced by endogenous estrogen and distinct xenoestrogens in breast cancer cells. Carcinogenesis. 2006;27:1567–1578. doi: 10.1093/carcin/bgi339. [DOI] [PubMed] [Google Scholar]
  • 31.Ramji DP, Foka P. CCAAT/enhancer-binding proteins: structure, function and regulation. Biochem J. 2002;365:561–575. doi: 10.1042/BJ20020508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Yang G, Gregory CW, Shang Q, O'Brien DA, Zhang YL. Differential expression of CCAAT/enhancer-binding protein-delta (C/EBPdelta) in rat androgen-dependent tissues and human prostate cancer. J Androl. 2001;22:471–480. [PubMed] [Google Scholar]
  • 33.Desai KV, Michalowska AM, Kondaiah P, Ward JM, Shih JH, Green JE. Gene expression profiling identifies a unique androgen-mediated inflammatory/immune signature and a PTEN (phosphatase and tensin homolog deleted on chromosome 10)-mediated apoptotic response specific to the rat ventral prostate. Mol Endocrinol. 2004;18:2895–2907. doi: 10.1210/me.2004-0033. [DOI] [PubMed] [Google Scholar]
  • 34.Wang GL, Shi X, Salisbury E, Sun Y, Albrecht JH, Smith RG, Timchenko NA. Growth hormone corrects proliferation and transcription of phosphoenolpyruvate carboxykinase in livers of old mice via elimination of CCAAT/enhancer-binding protein α-Brm complex. J Biol Chem. 2007;282:1468–1478. doi: 10.1074/jbc.M608226200. [DOI] [PubMed] [Google Scholar]
  • 35.Loomis KD, Zhu S, Yoon K, Johnson PF, Smart RC. Genetic ablation of CCAAT/enhancer binding protein α in epidermis reveals its role in suppression of epithelial tumorigenesis. Cancer Res. 2007;67:6768–6776. doi: 10.1158/0008-5472.CAN-07-0139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Huang L, Pu Y, Hepps D, Danielpour D, Prins GS. Posterior Hox gene expression and differential androgen regulation in the developing and adult rat prostate lobes. Endocrinology. 2007;148:1235–1245. doi: 10.1210/en.2006-1250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Waltregny D, Alami Y, Clausse N, de Leval J, Castronovo V. Overexpression of the homeobox gene HOXC8 in human prostate cancer correlates with loss of tumor differentiation. Prostate. 2002;50:162–169. doi: 10.1002/pros.10045. [DOI] [PubMed] [Google Scholar]
  • 38.Miller GJ, Miller HL, van Bokhoven A, Lambert JR, Werahera PN, Schirripa O, Lucia MS, Nordeen SK. Aberrant HOXC expression accompanies the malignant phenotype in human prostate. Cancer Res. 2003;63:5879–5888. [PubMed] [Google Scholar]
  • 39.Ramachandran S, Liu P, Young AN, Yin-Goen Q, Lim SD, Laycock N, Amin MB, Carney JK, Marshall FF, Petros JA, et al. Loss of HOXC6 expression induces apoptosis in prostate cancer cells. Oncogene. 2005;24:188–198. doi: 10.1038/sj.onc.1207906. [DOI] [PubMed] [Google Scholar]
  • 40.Shim C, Zhang W, Rhee CH, Lee JH. Profiling of differentially expressed genes in human primary cervical cancer by complementary DNA expression array. Clin Cancer Res. 1998;4:3045–3050. [PubMed] [Google Scholar]
  • 41.Hung YC, Ueda M, Terai Y, Kumagai K, Ueki K, Kanda K, Yamaguchi H, Akise D, Ueki M. Homeobox gene expression and mutation in cervical carcinoma cells. Cancer Sci. 2003;94:437–441. doi: 10.1111/j.1349-7006.2003.tb01461.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Al-Maghrabi J, Vorobyova L, Chapman W, Jewett M, Zielenska M, Squire JA. p53 Alteration and chromosomal instability in prostatic high-grade intraepithelial neoplasia and concurrent carcinoma: analysis by immunohistochemistry, interphase in situ hybridization, and sequencing of laser-captured microdissected specimens. Mod Pathol. 2001;14:1252–1262. doi: 10.1038/modpathol.3880471. [DOI] [PubMed] [Google Scholar]
  • 43.Downing SR, Russell PJ, Jackson P. Alterations of p53 are common in early stage prostate cancer. Can J Urol. 2003;10:1924–1933. [PubMed] [Google Scholar]
  • 44.Hayashi H, Abdollah S, Qiu Y, Cai J, Xu YY, Grinnell BW, Richardson MA, Topper JN, Gimbrone MA, Jr, Wrana JL, et al. The MAD-related protein Smad7 associates with the TGFβ receptor and functions as an antagonist of TGFβ signaling. Cell. 1997;89:1165–1173. doi: 10.1016/s0092-8674(00)80303-7. [DOI] [PubMed] [Google Scholar]
  • 45.Halder SK, Beauchamp RD, Datta PK. Smad7 induces tumorigenicity by blocking TGF-β-induced growth inhibition and apoptosis. Exp Cell Res. 2005;307:231–246. doi: 10.1016/j.yexcr.2005.03.009. [DOI] [PubMed] [Google Scholar]
  • 46.Landstrom M, Heldin NE, Bu S, Hermansson A, Itoh S, ten Dijke P, Heldin CH. Smad7 mediates apoptosis induced by transforming growth factor β in prostatic carcinoma cells. Curr Biol. 2000;10:535–538. doi: 10.1016/s0960-9822(00)00470-x. [DOI] [PubMed] [Google Scholar]
  • 47.Edlund S, Bu S, Schuster N, Aspenstrom P, Heuchel R, Heldin NE, ten Dijke P, Heldin CH, Landstrom M. Transforming growth factor-β1 (TGF- β)-induced apoptosis of prostate cancer cells involves Smad7-dependent activation of p38 by TGF-β-activated kinase 1 and mitogen-activated protein kinase kinase 3. Mol Biol Cell. 2003;14:529–544. doi: 10.1091/mbc.02-03-0037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Edlund S, Lee SY, Grimsby S, Zhang S, Aspenstrom P, Heldin CH, Landstrom M. Interaction between Smad7 and β-catenin: importance for transforming growth factor β-induced apoptosis. Mol Cell Biol. 2005;25:1475–1488. doi: 10.1128/MCB.25.4.1475-1488.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Migliaccio A, Castoria G, Di Domenico M, de Falco A, Bilancio A, Lombardi M, Barone MV, Ametrano D, Zannini MS, Abbondanza C, et al. Steroid-induced androgen receptor-oestradiol receptor β-Src complex triggers prostate cancer cell proliferation. EMBO J. 2000;19:5406–5417. doi: 10.1093/emboj/19.20.5406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Chieffi P, Kisslinger A, Sinisi AA, Abbondanza C, Tramontano D. 17β-Estradiol-induced activation of ERK1/2 through endogenous androgen receptor-estradiol receptor α-Src complex in human prostate cells. Int J Oncol. 2003;23:797–801. [PubMed] [Google Scholar]
  • 51.Burnham MR, Bruce-Staskal PJ, Harte MT, Weidow CL, Ma A, Weed SA, Bouton AH. Regulation of c-SRC activity and function by the adapter protein CAS. Mol Cell Biol. 2000;20:5865–5878. doi: 10.1128/mcb.20.16.5865-5878.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Fromont G, Vallancien G, Validire P, Levillain P, Cussenot O. BCAR1 expression in prostate cancer: association with 16q23 LOH status, tumor progression and EGFR/KAI1 staining. Prostate. 2007;67:268–273. doi: 10.1002/pros.20516. [DOI] [PubMed] [Google Scholar]
  • 53.Kleiner S, Faisal A, Nagamine Y. Induction of uPA gene expression by the blockage of E-cadherin via Src- and Shc-dependent Erk signaling. FEBS J. 2007;274:227–240. doi: 10.1111/j.1742-4658.2006.05578.x. [DOI] [PubMed] [Google Scholar]
  • 54.Gaylis FD, Keer HN, Wilson MJ, Kwaan HC, Sinha AA, Kozlowski JM. Plasminogen activators in human prostate cancer cell lines and tumors: correlation with the aggressive phenotype. J Urol. 1989;142:193–198. doi: 10.1016/s0022-5347(17)38709-8. [DOI] [PubMed] [Google Scholar]
  • 55.Zhang X, Fei Z, Bu X, Zhen H, Zhang Z, Gu J, Chen Y. Expression and significance of urokinase type plasminogen activator gene in human brain gliomas. J Surg Oncol. 2000;74:90–94. doi: 10.1002/1096-9098(200006)74:2<90::AID-JSO2>3.0.CO;2-7. [DOI] [PubMed] [Google Scholar]
  • 56.Tam NN, Ghatak S, Ho SM. Sex hormone-induced alterations in the activities of antioxidant enzymes and lipid peroxidation status in the prostate of Noble rats. Prostate. 2003;55:1–8. doi: 10.1002/pros.10169. [DOI] [PubMed] [Google Scholar]
  • 57.Poggi A, Costa P, Zocchi MR, Moretta L. Phenotypic and functional analysis of CD4+ NKRP1A+ human T lymphocytes. Direct evidence that the NKRP1A molecule is involved in transendothelial migration. Eur J Immunol. 1997;27:2345–2350. doi: 10.1002/eji.1830270932. [DOI] [PubMed] [Google Scholar]
  • 58.Ouyang X, DeWeese TL, Nelson WG, Abate-Shen C. Loss-of-function of Nkx3.1 promotes increased oxidative damage in prostate carcinogenesis. Cancer Res. 2005;65:6773–6779. doi: 10.1158/0008-5472.CAN-05-1948. [DOI] [PubMed] [Google Scholar]

Associated Data

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

Supplementary Materials

Supplementary Figures and Tables
neo1001_0020SD1.pdf (161.1KB, pdf)

Articles from Neoplasia (New York, N.Y.) are provided here courtesy of Neoplasia Press

RESOURCES