Skip to main content
Erschienen in: Breast Cancer Research 1/2019

Open Access 01.12.2019 | Research article

Genomic signature of parity in the breast of premenopausal women

verfasst von: Julia Santucci-Pereira, Anne Zeleniuch-Jacquotte, Yelena Afanasyeva, Hua Zhong, Michael Slifker, Suraj Peri, Eric A. Ross, Ricardo López de Cicco, Yubo Zhai, Theresa Nguyen, Fathima Sheriff, Irma H. Russo, Yanrong Su, Alan A. Arslan, Pal Bordas, Per Lenner, Janet Åhman, Anna Stina Landström Eriksson, Robert Johansson, Göran Hallmans, Paolo Toniolo, Jose Russo

Erschienen in: Breast Cancer Research | Ausgabe 1/2019

Abstract

Background

Full-term pregnancy (FTP) at an early age confers long-term protection against breast cancer. Previously, we reported that a FTP imprints a specific gene expression profile in the breast of postmenopausal women. Herein, we evaluated gene expression changes induced by parity in the breast of premenopausal women.

Methods

Gene expression profiling of normal breast tissue from 30 nulliparous (NP) and 79 parous (P) premenopausal volunteers was performed using Affymetrix microarrays. In addition to a discovery/validation analysis, we conducted an analysis of gene expression differences in P vs. NP women as a function of time since last FTP. Finally, a laser capture microdissection substudy was performed to compare the gene expression profile in the whole breast biopsy with that in the epithelial and stromal tissues.

Results

Discovery/validation analysis identified 43 differentially expressed genes in P vs. NP breast. Analysis of expression as a function of time since FTP revealed 286 differentially expressed genes (238 up- and 48 downregulated) comparing all P vs. all NP, and/or P women whose last FTP was less than 5 years before biopsy vs. all NP women. The upregulated genes showed three expression patterns: (1) transient: genes upregulated after FTP but whose expression levels returned to NP levels. These genes were mainly related to immune response, specifically activation of T cells. (2) Long-term changing: genes upregulated following FTP, whose expression levels decreased with increasing time since FTP but did not return to NP levels. These were related to immune response and development. (3) Long-term constant: genes that remained upregulated in parous compared to nulliparous breast, independently of time since FTP. These were mainly involved in development/cell differentiation processes, and also chromatin remodeling. Lastly, we found that the gene expression in whole tissue was a weighted average of the expression in epithelial and stromal tissues.

Conclusions

Genes transiently activated by FTP may have a role in protecting the mammary gland against neoplastically transformed cells through activation of T cells. Furthermore, chromatin remodeling and cell differentiation, represented by the genes that are maintained upregulated long after the FTP, may be responsible for the lasting preventive effect against breast cancer.
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s13058-019-1128-x) contains supplementary material, which is available to authorized users.
Julia Santucci-Pereira, Anne Zeleniuch-Jacquotte, Yelena Afanasyeva and Hua Zhong contributed equally to this work.
Abkürzungen
BMI
Body mass index
CV
Coefficient of variation
FDR
False discovery rate
FTP
Full-term pregnancy
GO
Gene Ontology
IPA
Ingenuity® Pathways Analysis
LCM
Laser capture microdissection
NP
Nulliparous
NUSE
Normalized Unscaled Standard Error
P
Parous
PLM
Probe-level models
QC
Quality control
RMA
Robust Multi-Array Average
RT-PCR
Reverse transcription polymerase chain reaction
TSLP
Time since last pregnancy
WT
Whole tissue

Background

The association of parity with breast cancer risk is well documented by both epidemiological and experimental data [14]. While the relationship is complex, with a transient increase in risk after each full-term pregnancy (FTP), the long-term effect for women who have their first FTP at an early age is a marked reduction in risk [5]. A better understanding of the molecular mechanisms underlying the effects of parity on the breast may help develop strategies to prevent breast cancer.
We previously reported the results of a study that assessed gene expression differences in the breast of 67 parous (P) and 40 nulliparous (NP) postmenopausal women who were free of any pathology and had volunteered to undergo a tissue biopsy [68]. We reported that in the postmenopausal breast, parity-induced gene expression changes were related to differentiation of this organ [6]. More specifically, we found that genes upregulated in the P breast, as compared to the NP breast, represented biological processes involved in differentiation and development, cell junction, RNA metabolic processes, and splicing machinery. The downregulated genes represented biological processes involved in cell proliferation, regulation of IGF-like growth factor receptor signaling, somatic stem cell maintenance, muscle cell differentiation, and apoptosis [6, 7].
We here report on a study with a similar design and conducted in the same general population, but focusing on premenopausal women. The main objective of this study was to assess the parity-associated gene expression differences in the breast of premenopausal women. Because the breast undergoes involution after pregnancy and there is a short-term increase in breast cancer risk following each FTP, we examined the gene expression differences in P vs. NP women as a function of time since last FTP. Additionally, we conducted a substudy, in which laser capture microdissection (LCM) was used to isolate breast epithelial cells from the stroma to evaluate how the gene expression observed in RNA extracted from whole breast tissue relates to gene expression in RNA extracted from breast epithelial cells and from stroma separately.

Methods

Study population and eligibility criteria

Study subjects were volunteers recruited among healthy women between the ages of 29 and 47 years and residing in Norrbotten County, Sweden. Exclusion criteria included a history of any cancer, complete bilateral oophorectomy, breast biopsy or breast implants, and hormonal treatment for infertility. Women who had completed a FTP or breastfed in the 12 months prior to enrollment, used oral contraceptives in the 6 months prior to enrollment, or used thyroid or steroid hormones, anti-coagulants, or diabetes medications in the 3 months prior to enrollment were also ineligible. The study was approved by the Regional Ethical Review Board for Northern Sweden at the University of Umeå, Sweden.
Volunteers who signed informed consent were scheduled for a biopsy. Women who had not had a mammogram within the year preceding enrollment received one prior to the biopsy to exclude breast cancer. Parous (P) women were defined as women who had had one or more full-term pregnancies, defined as a pregnancy lasting at least 37 weeks. The nulliparous (NP) group included women who had never been pregnant or who had no history of pregnancies lasting more than 8 weeks.

Data and breast tissue collection

Eligible volunteers completed a questionnaire that collected data on reproductive history, medical history, height and weight, first-degree family history of breast cancer, history of tobacco use, and current medications. Breast core needle biopsies were performed by two experienced radiologists (P. Bordas and A. Eriksson) at the Mammography Department at Sunderby Hospital, Luleå, Sweden. A 12-Gauge BARD® MONOPTY® core biopsy needle was used, and four to eight biopsies were taken from the upper outer quadrant of one breast. One biopsy specimen was placed in 70% ethanol for histopathological analysis, and the remaining in RNALater® (Ambion) solution. Tissue samples were stripped of all personal identifiers and sent to Fox Chase Cancer Center for analysis. All samples were first reviewed by the study pathologist (J. Russo) to confirm the absence of atypia or cancer. During all the experiments, the researchers at Fox Chase Cancer Center were blinded to the parity status of the samples.

Gene expression microarrays

Total RNA from the biopsies was isolated using the Allprep RNA/DNA Mini Kit (Qiagen, Alameda, CA, USA). Quantity and quality of total RNA were assessed using NanoDrop v3.3.0 (NanoDrop Technologies, Wilmington, DE, USA) and Agilent 2100 Bioanalyzer (Agilent Technologies, CA, USA), respectively.
GeneChip Expression 3′-Amplification Two-Cycle cDNA Synthesis Kit (Affymetrix, Santa Clara, CA) was used for sample preparation and hybridization to Affymetrix Human Genome Gene Chip U133 Plus 2.0 arrays. For quality control purposes, we included in each batch (9 to 12 samples) one blinded duplicate sample from another batch.
After scanning, all microarrays were subjected to quality control (QC) analysis to ensure that they were in the acceptable ranges for standard Affymetrix quality measures (Scale Factor, Percent Present, and Average Background). In addition, quality was assessed using graphical tools based on Affymetrix probe-level models (PLM) [9]. The Normalized Unscaled Standard Error (NUSE) plot, in particular, was used to disqualify lower quality arrays. Ten arrays (8%—9 P and 1 NP) did not fulfill quality criteria and were not included in the statistical analysis. The concordance correlation coefficients for the QC replicates were greater than 98%.

Data preprocessing and batch adjustment

The Affymetrix data were analyzed using R language for statistical computing (R version 2.14.1) [10] and Bioconductor [11]. Preprocessing methods and filtering criteria were similar to those used in our previous study [6, 7]. Probesets for which both the proportion of present calls was < 75% and the difference in proportion of present calls between P and NP samples was < 25% were filtered out. Probesets with coefficient of variation (CV) across samples falling in the first quartile were also excluded. After filtering, 14,920 probesets (27%) remained in the analysis. To account for inter-batch variability, the data were adjusted for batch using the ComBat method [12] implemented in the Bioconductor package sva [13].

Statistical analysis of gene expression differences between parous and nulliparous women

Linear regression models were used to identify probesets differentially expressed in P vs. NP samples. For each probeset, an unadjusted p value measuring the significance of parity (yes/no) as an independent predictor of the log-transformed normalized gene expression value was calculated using single regression. We also used multiple regression analysis to identify differentially expressed probesets while controlling for potential confounders. The associations of subject characteristics with parity status and with gene expression were examined to identify potential confounders. Multivariate models were adjusted for age, body mass index (BMI) (which was associated with gene expression), and smoking duration (which was associated with both gene expression and parity status). We also adjusted for phase of cycle/use of a hormonal IUD, which could affect gene expression. False discovery rate (FDR) was used to control for multiple comparisons, using QVALUE in the R package version 1.28.0 [14].
In order to identify the most robust parity-associated differences in gene expression, we first analyzed the data using a discovery/validation resampling approach. A discovery dataset was generated by selecting at random 2/3 of the P women and 2/3 of the NP women from the complete dataset. The remaining women formed a corresponding validation dataset. This step was repeated 12 times, leading to 12 discovery/validation dataset pairs. Probesets with FDR < 20% in any discovery dataset and p value < 0.05 in the corresponding validation dataset were considered validated for this dataset pair. We report the probesets (and corresponding genes) that were validated in at least 2 of the 12 dataset pairs.
The breast undergoes involution after pregnancy, which is likely to be associated with transient changes in gene expression. Further, although the long-term effect of early parity (before 35 years of age) is a reduction in breast cancer risk for pregnancy [5], it is well documented that there is a short-term increase in risk after each FTP. This suggests that the gene expression pattern in the breast may be different in the first few years after pregnancy than in later years. Therefore, we examined the parity-associated gene expression differences according to time between last FTP and biopsy (time since last pregnancy, TSLP). To optimize our chances of detecting TSLP-related differences in gene expression, we included in these analyses probesets that were differentially expressed (FDR < 10% and at least 1.2-fold change) in the subgroup of P women whose last FTP was ≤ 5 years before biopsy as compared to NP women, in addition to the probesets identified in the overall P-NP comparison. All women were included in these analyses and the patterns of expression were examined using clustering analysis in women classified according to TSLP (< 5, 5–10, or > 10 years). Considering upregulated and downregulated genes separately, K-means cluster analyses were performed using Multiexperiment Viewer software (MeV- v.4.8.1) [15], with Pearson uncentered as distance metric. We examined the clusters formed after randomly setting the number of clusters (K) to 2, 3, 4, or 5.

Mining for functional categories and pathways

Data mining methods were applied to the differentially expressed genes to detect biological processes and pathways affected by parity. Ingenuity® Pathways Analysis (IPA) software version 24390178 (QIAGEN) was used to investigate canonical pathways. Gene Ontology (GO) enrichment analysis was performed using conditional hypergeometric tests in the Bioconductor GOstats package [16]. We carried out analyses separately for each cluster of upregulated genes. GO (gene ontology) terms with p value < 0.01 were considered enriched. To evaluate the GO terms enriched by each cluster of genes, the terms were grouped into broader terms (developmental process, immune response, or others) following the GO hierarchical tree graph view from GO consortium [17]. Few genes were identified as downregulated; therefore, we did not conduct GO analyses for these genes but rather examined the literature to identify their roles.
In addition, genes that were validated by real-time RT-PCR were used for analysis into cBioPortal for Cancer Genomics (http://​www.​cbioportal.​org/​) [18, 19]. We evaluated whether these genes have been described to be modified in breast cancer cases available in the cBioPortal databank; in addition, an overall survival Kaplan-Meier curve was generated. In total, 11 genes were evaluated among 5796 breast cancer patients.

Validation through real-time RT-PCR

Eleven genes were selected for real-time RT-PCR analyses based on their biological roles in cell differentiation, proliferation, and chromatin remodeling. The assays were performed in the subset of 17 NP and 20 P samples (10 with TSLP < 5, and 10 with TSLP > 5) from whom sufficient RNA remained available, using TaqMan® Gene Expression Master Mix and TaqMan® Gene Expression Assays (Life Technologies). The end point used in the RT-PCR quantification, Ct, was defined as the PCR cycle number at which each assay target passes the threshold. Each assay was normalized to 18S, a housekeeping gene used as endogenous control (ΔCt). The difference between parous and nulliparous were estimated as the difference in mean ΔCt values (−ΔΔCt). To assess the statistical significance of the differences between P and NP, batch-adjusted p values were calculated using linear regression and comparisons with p value < 0.10 and fold change of at least 1.2 were considered statistically significant. Gene expression measured using RT-PCR and Affymetrix arrays were compared in the 37 subjects for whom both assays were used. Fold changes were estimated from multiple regressions using batch-adjusted, RMA (Robust Multi-Array Average)-normalized gene expression intensities, and intraclass and Spearman correlation coefficients were calculated.

Immunohistochemical (IHC) staining

Paraffin sections at 4 μm were deparaffinized and placed in the antigen unmasking solution (Vector Laboratories, Burlingame, CA) and microwaved for 10 min at 100 °C. After cooling for 20 min, slides were quenched with Peroxide Block (BioGenex, Fremont, CA; #HK111) for 10 min, followed by blocking with Power Block (BioGenex, #HK085) for 20 min at room temperature. The sections were then stained with primary antibodies using a i6000 BioGenex Autostainer following standard protocol. The antibodies used were as follows: Cytokeratin 5 (BioGenex, #AN484-10 M, pre-diluted), CD123 (BD Biosciences, #554527; 1:400 dilution), LAMP3 (Abcam, #ab111090), Desmocollin 3 (Abcam, #ab190118; 1:150 dilution), CD2 (Abcam, #ab131276; 1:200 dilution), and CD3D (Abcam, #ab109531; 1:150 dilution). A Super Sensitive TM Polymer-HRP Detection System (BioGenex; #QD430-XAKE) was used to detect the staining. The images were acquired at × 400 magnification using an Aperio Digital Pathology Slide Scanner (Leica Biosystems, Buffalo Grove, IL) and analyzed by Aperio ImageScope Software (Leica Biosystems).

Laser capture microdissection (LCM)

In additional samples, we conducted a substudy to assess how gene expression in RNA extracted from whole breast tissue relates to gene expression in RNA extracted from epithelial and stromal tissues. Breast biopsy tissue fixed in RNA later was frozen and cryostat was used for histological sectioning. The frozen sections were then stained with hematoxylin and eosin specially prepared utilizing RNAse-free water to avoid RNA degradation [20]. LCM was performed using the VERITAS Microdissection Instrument (Arcturus, CA, USA) to select and capture all the epithelial tissue present in each section. The tissue left on the slide was then scrapped into a different tube and classified as stroma. The RNA extraction from the collected cells was performed using the Arcturus® PicoPure®RNA Isolation Kit (Life Technologies). RNA was also extracted from a second core biopsy, in which no LCM was performed (called hereafter whole tissue, WT).
For each woman included in this substudy, three microarrays were performed in the same batch, using RNA extracted from (1) the epithelial cells of the mammary gland, (2) the stroma, and (3) WT. RNA amplification and labelling was performed using MessageAmpTM Premier RNA Amplification Kit (Life Technologies), and the arrays were hybridized to Affymetrix Human Genome Gene Chip U133 Plus 2.0 arrays. All arrays were subjected to QC analysis as described earlier. The arrays were RMA pre-processed, and probesets with < 75% of present calls and/or low CV (i.e., CV in the first quartile) were filtered out, resulting in 10,252 probesets for analysis. All values were log-transformed and normalized prior to analysis.
Nine subjects (5 NP and 4 P) had successful arrays for whole tissue, epithelial tissue, and stromal tissue. For each subject, a linear regression model was fitted across all genes. The gene expression values in the whole tissue were modeled as a linear function of the gene expression in epithelial and stromal tissues.
Gene expression comparison between the breast tissue types was performed for ten subjects (5 P and 5 NP) with successful epithelium and stroma arrays. For each probe, the fold changes were calculated as the median of within subject fold changes [expression in epithelium] / [expression in stroma] for each subject. A paired two-sample t-test was performed for each probe set, and p values were adjusted for multiple comparisons using the FDR approach. Probes with FDR < 10% and fold changes of at least 20% were considered statistically different between epithelium and stroma. GO analysis was performed using the same methodology described above. The small number of samples limited our gene expression analyses between tissue types in function of parity.

Results

Volunteers included in the analysis

A total of 307 women between ages 29 and 47 volunteered between March 2011 and June 2012 (Fig. 1). After exclusions related to eligibility, lack of epithelial structures, or QC failures, samples from 109 women (30 NP and 79 P) were included in the main study comparing P vs. NP, and samples from 10  women were included in the LCM substudy comparing the tissue types (Fig. 1).
P and NP women were similar with respect to most characteristics, such as age of menarche, breast density, and body mass index (BMI) (Table 1). However, the median age and median smoking duration were lower in NP than P women, and a larger proportion of P had a hormonal intra-uterine device (IUD).
Table 1
Descriptive characteristics of the study subjects
Characteristics
Parous (n = 79)
Nulliparous (n = 30)
p value
Age at visit, years
39.9 (30.1, 47.3)
35.8 (30.1, 46.2)
0.03
Number of FTP
 1
13 (16%)
  
 2
44 (46%)
  
 3+
22 (28%)
  
Time since last FTP, years
 ≤ 5
30 (39%)
  
 6–10
29 (37%)
  
 > 10
20 (25%)
  
Age at first FTP, years
26 (18, 27)
  
Phase of cycle/hormonal IUD1
  
0.05
 Luteal
21 (28%)
14 (52%)
 
 Ovulatory
10 (13%)
4 (15%)
 
 Follicular
21 (28%)
7 (26%)
 
 Hormonal IUD
23 (31%)
2 (7%)
 
 Missing
4
3
 
OC ever use
74 (94%)
27 (90%)
0.68
Breast density, BIRADS
 1
10 (13%)
4 (13%)
0.38
 2
16 (20%)
2 (7%)
 
 3
10 (13%)
4 (13%)
 
 4
43 (54%)
20 (67%)
 
Age at menarche, years
13.0 (11.0, 16.0)
13.0 (11.0, 15.0)
0.60
Family history of breast cancer
7 (9%)
2 (7%)
0.99
Height, cm
165.0 (148.0, 184.0)
166.5 (157.0, 174.0)
0.77
Weight, kg
68.0 (41.0, 115.0)
64.0 (46.0, 97.0)
0.38
BMI
24.2 (18.7, 38.0)
23.4 (18.7, 37.1)
0.24
Smoking
 Never
51 (64%)
20 (67%)
0.15
 Past
23 (29%)
5 (17%)
 
 Current
5 (6%)
5 (17%)
 
 Smoking duration, years
8.0 (0.5, 20.0)
17.0 (7.0, 30.0)
0.006
 Years since quitting (past smokers)
13.0 (0.1, 26.0)
6.0 (1.0, 12.0)
0.09
1p value for IUD/ no IUD = 0.01; excluding women having an IUD, there was no statistically significant difference in phase of menstrual cycle between P and NP

Differential gene expression

Using the discovery and validation approach described in “Methods”, 54 probesets, representing 43 genes (Table 2), were differentially expressed between P and NP women. Of the 43 genes, 40 were upregulated and 3 downregulated in the parous premenopausal breast (Table 2). Upregulated genes in the P breast included APOBEC3G, DSC3, FZD8, and EAF2, while FOXQ1 was among the downregulated genes.
Table 2
Genes differentially expressed between parous and nulliparous premenopausal women (discovery/validation approach)
ProbeID
EntrezID
Symbol
GeneName
FDR
Fold change
Genes upregulated in parous women
 206641_at
608
TNFRSF17
Tumor necrosis factor receptor superfamily, member 17
0.013
2.41
 228504_at
6332
SCN7A
Sodium channel, voltage-gated, type VII, alpha subunit
0.006
2.15
 237625_s_at
3514
IGKC
Immunoglobulin kappa constant
0.006
1.94
 222838_at
57823
SLAMF7
SLAM family member 7
0.017
1.93
 224342_x_at
96610
LOC96610
BMS1 homolog, ribosome assembly protein (yeast) pseudogene
0.032
1.80
 206121_at
270
AMPD1
Adenosine monophosphate deaminase 1
0.021
1.76
 206033_s_at
1825
DSC3
Desmocollin 3
0.006
1.68
 1555759_a_at
6352
CCL5
Chemokine (C-C motif) ligand 5
0.018
1.67
 206478_at
9834
KIAA0125
KIAA0125
0.006
1.65
 213193_x_at
28639
TRBC1
T cell receptor beta constant 1
0.006
1.60
 207651_at
29909
GPR171
G protein-coupled receptor 171
0.014
1.60
 206310_at
6691
SPINK2
Serine peptidase inhibitor, Kazal type 2 (acrosin-trypsin inhibitor)
0.006
1.59
 231647_s_at
83416
FCRL5
Fc receptor-like 5
0.012
1.59
 203130_s_at
3800
KIF5C
Kinesin family member 5C
0.023
1.58
 205831_at
914
CD2
CD2 molecule
0.012
1.54
 216430_x_at
28823
IGLV1-44
Immunoglobulin lambda variable 1-44
0.034
1.52
 211339_s_at
3702
ITK
IL2-inducible T cell kinase
0.016
1.50
 206666_at
3003
GZMK
Granzyme K (granzyme 3; tryptase II)
0.030
1.49
 204562_at
3662
IRF4
Interferon regulatory factor 4
0.017
1.47
 213539_at
915
CD3D
CD3d molecule, delta (CD3-TCR complex)
0.011
1.45
 206181_at
6504
SLAMF1
Signaling lymphocytic activation molecule family member 1
0.014
1.45
 206150_at
939
CD27
CD27 molecule
0.019
1.44
 206991_s_at
1234
CCR5
Chemokine (C-C motif) receptor 5 (gene/pseudogene)
0.010
1.41
 235153_at
138065
RNF183
Ring finger protein 183
0.026
1.41
 214470_at
3820
KLRB1
Killer cell lectin-like receptor subfamily B, member 1
0.038
1.40
 230011_at
150365
MEI1
Meiosis inhibitor 1
0.010
1.39
 211902_x_at
10730
YME1L1
YME1-like 1 (S. cerevisiae)
0.015
1.37
 221584_s_at
3778
KCNMA1
Potassium large conductance calcium-activated channel, subfamily M, alpha member 1
0.034
1.36
 219551_at
55840
EAF2
ELL associated factor 2
0.015
1.35
 220402_at
63970
TP53AIP1
Tumor protein p53 regulated apoptosis inducing protein 1
0.015
1.35
 206761_at
10225
CD96
CD96 molecule
0.036
1.33
 212314_at
23231
SEL1L3
Sel-1 suppressor of lin-12-like 3 (C. elegans)
0.014
1.31
 204682_at
4053
LTBP2
Latent transforming growth factor beta binding protein 2
0.006
1.31
 207655_s_at
29760
BLNK
B cell linker
0.015
1.31
 204205_at
60489
APOBEC3G
Apolipoprotein B mRNA-editing enzyme, catalytic polypeptide-like 3G
0.014
1.26
 223322_at
83593
RASSF5
Ras association (RalGDS/AF-6) domain family member 5
0.024
1.25
 227405_s_at
8325
FZD8
Frizzled family receptor 8
0.024
1.24
 211469_s_at
10663
CXCR6
Chemokine (C-X-C motif) receptor 6
0.019
1.22
 233555_s_at
55959
SULF2
Sulfatase 2
0.032
1.20
 209574_s_at
753
LDLRAD4
Low-density lipoprotein receptor class A domain containing 4
0.045
1.15
Genes downregulated in parous women
 227475_at
94234
FOXQ1
Forkhead box Q1
0.026
0.64
 244680_at
2743
GLRB
Glycine receptor, beta
0.046
0.79
 236399_at
54103
PION
Pigeon homolog (Drosophila)
0.041
0.86
Analyses of gene expression according to TSLP identified 286 genes (416 probesets) differentially expressed between P and NP samples (Fig. 2). Among these, 238 genes (352 probesets) were upregulated in P women, while 48 genes (64 probesets) were downregulated (Additional file 1).
For probesets upregulated in the P women, gene expression differences clustered in three expression patterns as a function of TSLP (Fig. 3). The first cluster consisted of 83 genes (107 probesets) which were upregulated following the last FTP, but whose expression progressively returned to the level of expression observed in NP women (Fig. 3a). These 83 genes were named “transient” genes. The second cluster consisted of probesets for which the P-NP differences were the highest for women with < 5 years since last FTP, decreased with increasing TSLP but remained elevated as compared to NP women. The 95 genes (154 probesets) in this cluster were called “long-term changing” (Fig. 3b). In the last cluster, which included 60 genes (91 probesets), the fold changes between P an NP appeared constant, regardless of the TSLP. Therefore, we called these “long-term constant” genes (Fig. 3c).
Gene Ontology (GO) enrichment analyses showed an abundance of GO terms associated to developmental processes or immune response (Fig. 4), other less-abundant enriched GO terms were related to proliferation, intracellular transport, and cell death. Among the genes whose expression was transiently affected by parity 55%, including CD8A, XCL1, and GZMA (Table 3), enriched GO terms related to immune response (Additional file 2). Among the genes classified as long-term changing, 32% were related to immune response (e.g., CD2) and 24% were involved in developmental processes (e.g., EAF2) (Table 3, Additional file 3). Notably, of the long-term constant genes, 56% were related to developmental/differentiation processes (Additional file 4), including EGR3, DSC3, KRT5, and FZD8 (Table 3). These data indicate that the proportion of genes involved in immune response decreases among the genes that are upregulated irrespectively of TSLP, while the proportion of genes related to developmental processes increases (chi-squared p value = p < 0.001).
Table 3
Genes that enriched developmental processes and immune response gene ontologies
Transient genes
 Developmental process
  ANXA1
CAMK4
CD8A
CD27
EOMES
ITK
  JAK3
LCK
PTPN22
PTPRC
SASH3
THEMIS
 Immune response
  ANXA1
CAMK4
CCL5
CD8A
CD27
CD48
  CD96
CD247
CORO1A
CST7
CXCL14
CXCL9
  EOMES
FASLG
FYB
GZMA
HCST
IGLC1
  IKZF1
IL16
IL7R
ITGAL
ITK
JAK3
  KLHL6
LAT
LAX1
LCK
LCP2
LY9
  PRKCB
PTPN22
PTPRC
SASH3
SELPLG
SEMA4D
  sSH2D1A
SLAMF1
THEMIS
TRAC
XCL1
 
Long-term changing genes
 Developmental process
  C3
CCR2
CD2
CXCL10
DACT1
DKK3
  EAF2
EPHA7
FGF1
FGFR2
HCLS1
HLA-DOA
  IL12RB1
IRF4
LAMA2
OSR2
PDGFRA
SIPA1L1
  SPHK1
     
 Immune response
  APOBEC3G
BLNK
C1S
C3
CCR2
CD2
  CD3D
CD38
CRTAM
CXCL10
GPR183
HLA-DOA
  HLA-DPB1
IGKC
IL12RB1
IRF4
LPXN
MZB1
  NFATC2
PAWR
POU2AF1
PRKCQ
SAMSN1
SLAMF7
  TRBC1
     
Long-term constant genes
 Developmental process
  BHLHE22
CCL19
CCL2
DCN
DSC3
EGR3
  EPHB1
FHL2
FZD8
GLI3
GPR65
KCNMA1
  KIF5C
KRT5
MYLK
NFASC
PRKCA
SALL1
  SDC1
SULF1
SULF2
TAGLN
TREM2
TYMS
  VCAM1
WIPF3
XDH
ZIC1
  
 Immune response
  CCL2
CCL19
GLI3
HLA-DPA1
HLA-DRA
VCAM1
Italicized genes enriched both developmental and immune response processes
Evaluation of the canonical pathways represented by the differentially expressed genes also showed that pathways involved in signaling and activation of T cells were enriched by both transient and long-term genes (Fig. 5).
The 48 genes (64 probesets) downregulated in the P breast (Table 4) fell into two patterns. Twenty-two genes, including WIF1, EDN1, CXCL1, and FOXQ1, were downregulated in the P breast and remained with lower expression levels compared to NP breast irrespective of the number of years since last FTP. The remaining 26 genes were downregulated in women with TSLP < 5 years; however, the expression increased reaching similar or higher levels than those in NP women when TSLP increased. These genes included DLG5, KDM4B, and TOX3. We did not conduct a GO enrichment analysis for these genes because of their limited number.
Table 4
Genes downregulated within parous breast
EntrezID
ProbeID
Symbol
GeneName
Genes downregulated constantly
 653
205431_s_at
BMP5
Bone morphogenetic protein 5
 694
1559975_at
BTG1
B cell translocation gene 1, anti-proliferative
 285382
242447_at
C3orf70
Chromosome 3 open reading frame 70
 1277
202310_s_at
COL1A1
Collagen, type I, alpha 1
 131873
242641_at
COL6A6
Collagen, type VI, alpha 6
 2919
204470_at
CXCL1
Chemokine (C-X-C motif) ligand 1 (melanoma growth-stimulating activity, alpha)
 55184
219951_s_at
DZANK1
Double zinc ribbon and ankyrin repeat domains 1
 1906
222802_at
EDN1
Endothelin 1
 133121
229916_at
ENPP6
Ectonucleotide pyrophosphatase/phosphodiesterase 6
 94234
227475_at
FOXQ1
Forkhead box Q1
 2743
205280_at
GLRB
Glycine receptor, beta
 253012
242601_at
HEPACAM2
HEPACAM family member 2
 3736
230849_at
KCNA1
Potassium voltage-gated channel, shaker-related subfamily, member 1 (episodic ataxia with myokymia)
 79442
219949_at
LRRC2
Leucine-rich repeat containing 2
 9848
205442_at
MFAP3L
Microfibrillar-associated protein 3-like
 4300
204918_s_at
MLLT3
Myeloid/lymphoid or mixed-lineage leukemia (trithorax homolog, Drosophila); translocated to, 3
 3340
1554010_at
NDST1
N-Deacetylase/N-sulfotransferase (heparan glucosaminyl) 1
 11069
205651_x_at
RAPGEF4
Rap guanine nucleotide exchange factor (GEF) 4
 950
201647_s_at
SCARB2
Scavenger receptor class B, member 2
 1317
236217_at
SLC31A1
Solute carrier family 31 (copper transporters), member 1
 116441
228489_at
TM4SF18
Transmembrane 4 L six family member 18
 11197
204712_at
WIF1
WNT inhibitory factor 1
Genes downregulated transiently
 170692
230040_at
ADAMTS18
ADAM metallopeptidase with thrombospondin type 1 motif, 18
 267
202203_s_at
AMFR
Autocrine motility factor receptor, E3 ubiquitin protein ligase
 56892
218541_s_at
C8orf4
Chromosome 8 open reading frame 4
 401546
229964_at
C9orf152
Chromosome 9 open reading frame 152
 760
209301_at
CA2
Carbonic anhydrase II
 54102
227742_at
CLIC6
Chloride intracellular channel 6
 1281
211161_s_at
COL3A1
Collagen, type III, alpha 1
 9231
229689_s_at
DLG5
Discs, large homolog 5 (Drosophila)
 54898
213712_at
ELOVL2
ELOVL fatty acid elongase 2
 2069
205767_at
EREG
Epiregulin
 2330
205776_at
FMO5
Flavin containing monooxygenase 5
 2717
214430_at
GLA
Galactosidase, alpha
 26585
218469_at
GREM1
Gremlin 1, DAN family BMP antagonist
 2892
230144_at
GRIA3
Glutamate receptor, ionotropic, AMPA 3
 23704
222379_at
KCNE4
Potassium voltage-gated channel, Isk-related family, member 4
 23030
212492_s_at
KDM4B
Lysine (K)-specific demethylase 4B
 5366
204286_s_at
PMAIP1
Phorbol-12-myristate-13-acetate-induced protein 1
 11098
202458_at
PRSS23
Protease, serine, 23
 5744
211756_at
PTHLH
Parathyroid hormone-like hormone
 157869
214725_at
SBSPON
Somatomedin B and thrombospondin, type 1 domain containing
 1811
206143_at
SLC26A3
Solute carrier family 26, member 3
 51012
229835_s_at
SLMO2
Slowmo homolog 2 (Drosophila)
 6431
206108_s_at
SRSF6
Serine/arginine-rich splicing factor 6
 27324
216623_x_at
TOX3
TOX high mobility group box family member 3
 7366
207392_x_at
UGT2B15
UDP glucuronosyltransferase 2 family, polypeptide B15
 151126
1555801_s_at
ZNF385B
Zinc finger protein 385B
Finally, we examined genes reported to be related to chromatin remodeling. Among those upregulated, we observed APOBEC3G, TOX, UHRF1, and NAP1L2, while KDM4B and TOX3 were downregulated.

Validation of microarray results

P/NP differences in gene expression were confirmed for eight out of the 11 genes analyzed by real-time PCR in a subset of 37 samples (20 P and 17 NP); Ct values, ∆Cts, and −∆∆Cts are shown in Additional file 5. WIF1 was downregulated, while EAF2, BHLHE22, APOBEC3G, DSC3, KRT5, EGR3, and RASGRP1 were upregulated in the P breast (Table 5). The intraclass correlation coefficient (ICC) comparing the real-time to microarray measurements varied from 0.35 (EAF2) to 0.92 (WIF1), and Spearman correlation coefficients varied from 0.41 (EAF2) to 0.94 (WIF1) (Table 5).
Table 5
Correlation of real-time RT-PCR and microarray results
Genes
Affymetrix probeset
TaqMan Assay ID
Microarray
Real time
ICC
Spearman correlation
FC
P
FC
P
WIF1
204712_at
Hs00183662_m1
0.39
0.055
0.32
0.034
0.92
0.94
FOXQ1
227475_at
Hs00536425_s1
0.64
0.020
0.70
0.218
0.64
0.69
FZD8
227405_s_at
Hs00259040_s1
1.3
0.003
1.17
0.188
0.43
0.42
SDC1
201286_at
Hs00896423_m1
1.48
0.014
1.19
0.456
0.73
0.62
EAF2
219551_at
Hs00218407_m1
1.36
0.008
1.24
0.059
0.35
0.41
BHLHE22
228636_at
Hs01084964_s1
1.24
0.078
1.35
0.022
0.41
0.53
APOBEC3G
204205_at
Hs00222415_m1
1.44
0.000
1.44
0.001
0.39
0.48
DSC3
206033_s_at
Hs00170032_m1
1.70
0.002
1.45
0.063
0.64
0.51
KRT5
201820_at
Hs00361185_m1
1.51
0.012
1.54
0.062
0.64
0.71
EGR3
206115_at
Hs00231780_m1
1.71
0.026
1.57
0.072
0.76
0.70
RASGRP1
205590_at
Hs00996727_m1
1.61
0.002
1.71
0.002
0.66
0.74
Correlation of real-time RT-PCR and microarray results were calculated using the 20 parous vs. 17 nulliparous samples, for whom enough RNA material was still available. FC mean fold change, P batch-adjusted p value, ICC intraclass correlation coefficient
We also analyzed the 11 selected genes in 5796 patients of breast cancer available in cBioPortal. One or more of these genes were altered in 770 (13%) breast cancer patients. BHLHE22 was the gene that appears altered in the largest percentage of patients (6.9%); the alterations included amplification, deep deletion, and missense mutations. The other ten genes showed to be altered in 1.5% or less patients (Table 6). An overall Kaplan-Meier survival curve has been generated comparing the group of patients with these 11 genes altered versus patients without the alterations. The group of patients that contain alteration in these genes has shorter overall survival, median of 143.13 versus 168.3 months, logrank test p value = 9.09e−5 (Fig. 6).
Table 6
Gene alteration in 5796 breast cancer subjects
Genes
Number of samples altered
Percent of samples altered
BHLHE22
400
6.9
EGR3
87
1.5
WIF1
73
1.3
FOXQ1
73
1.3
FZD8
64
1.1
DSC3
57
1.0
KRT5
55
1.0
SDC1
35
0.6
EAF2
30
0.5
RASGRP1
30
0.5
APOBEC3G
28
0.5
We performed IHC analyses to evaluate whether the changes in gene expression reflected into the protein expression levels. We have evaluated two proteins related to differentiation Desmocollin 3 and Cytokeratin 5. These two markers were presented in all tested samples, and although we have observed slight upregulation of their genes by microarray and RT-PCR, we did not detect protein differences between P and NP breast tissues. Related to immune response, we evaluated markers for dendritic cells because of their role in antigen presentation for T cell activation. Using two markers CD123 and LAMP3, we detected few positive cells with no differences between P and NP samples. We also evaluated CD2 and CD3D, markers of T cell activation (Fig. 7). The percentage of positive cells for CD2 was low in both groups, we observed slight increase in the percentage of CD2-positive cells in parous women (1.6 times; P median = 0.59%, NP median = 0.31%); however, it was not statistically significant (p value = 0.156). The overall percentage of CD3D-positive cells was higher ranging from 0.6 to 9.4%. We confirmed a statistically significant twofold increase in the percentage of CD3D-positive cells in the P breast (P median = 3.28%, NP median = 1.62%, p value = 0.006) (Fig. 7). We also compared whether the percentage of positive cells between P women with TSLP ≤ 5 and > 5 differed. For both CD2 and CD3D, we see a slight larger number of positive cells in TSLP ≤ 5, but were not statistically significant (CD2 p value = 0.16 and CD3D p value = 0.09).

Laser capture microdissection (LCM) results

We found that a linear function of gene expression levels in RNA from epithelial tissue and RNA from stromal tissue predicted extremely well the level of expression in RNA extracted from the whole tissue (0.91 < R2 < 0.95), i.e., the gene expression in whole tissue was a weighted average of the gene expression in epithelial tissue and in stromal tissue for all nine individuals included in this substudy (Table 7). This was observed regardless of the proportion of each tissue type found on the paraffin sections, which varied across individuals (regression coefficient for epithelial tissue ranged from 0.09 to 0.68).
Table 7
Linear regression coefficients of gene expression levels in breast tissue
Subject
β coefficients
R 2
Epithelial tissue
Stromal tissue
1
0.091
0.910
0.943
2
0.461
0.503
0.945
3
0.495
0.425
0.905
4
0.516
0.406
0.922
5
0.287
0.624
0.929
6
0.354
0.622
0.953
7
0.681
0.308
0.920
8
0.342
0.654
0.939
9
0.122
0.877
0.926
Analysis of gene expression differences between epithelium and stroma showed 730 genes (956 probesets) with higher expression levels in the epithelium, while 663 genes (1020 probesets) were more expressed in the stroma (Additional file 6). GO analyses of these genes demonstrated a broader range of biological processes enriched by the genes with higher expression levels in the stroma (306 GO terms), which included cell motility, cell signaling, angiogenesis, development, and lipid process among other processes. Conversely, the genes with higher expression in the epithelium, enriched 12 GO terms, consisting a biological process involved in epithelial development and tight junction among other GOs (Additional file 7).

Discussion

This study evaluated gene expression differences in parous and non-parous breast using biopsies from healthy premenopausal volunteers. Using a discovery/validation approach, we found 43 differentially expressed genes (Table 2). Evaluation of expression differences between NP and P as a function of TSLP identified 238 genes up- and 48 genes downregulated in the P breast. The downregulated genes fell into two patterns of expression (transient and long-term), while the upregulated genes fell into three patterns (transient, long-term changing, and long-term constant) (Fig. 3). Through GO enrichment analyses, we found that genes whose expression was transiently increased after pregnancy were mainly related to immune response. Long-term changing genes included immune- and development-related genes, while genes categorized as long-term constant were mainly involved in cell differentiation and developmental processes (Fig. 4). LCM performed in a small set of samples indicated that the gene expression observed on whole-tissue arrays corresponded to the weighted average of the gene expression observed in the epithelial and stromal tissues.
Among the 43 differentially expressed genes (Table 2) found in our discovery/validation analysis were DSC3 and KRT5, whose differential expression was confirmed by RT-PCR. These genes were also found in our previous study conducted in postmenopausal women [6, 7], indicating that the expression of these genes is durably modified by pregnancy. Consistent with this observation, DSC3 and KRT5 fell into the “long-term constant” category in our analysis by TSLP. These two genes are involved in cell communication and epithelial differentiation [21, 22]. Additionally, DSC3 has been suggested to act as a tumor suppressor [2326], and its silencing is a common event in breast tumors [26].
While a discovery/validation approach is valuable to reduce the chance of false-positive results, our sample size was fairly small, which can lead to unstable results and lack of detection of some associations [27, 28]. This was a concern particularly because the breast undergoes major remodeling after a pregnancy/breastfeeding. Thus, genes may go through successive changes in pattern of expression after pregnancy, and analyses ignoring time since last pregnancy could miss transient expression modifications. We therefore also examined gene expression differences according to time since last pregnancy. To the best our knowledge, this is the first study that used a whole-transcriptome approach to demonstrate a cluster of biological functions following distinct expression patterns in the human breast following pregnancy.
The observation that the “long-term constant” genes were mainly involved in cell differentiation and developmental processes (Fig. 4) is consistent with the transcriptomic profile we previously described in the parous postmenopausal breast, in which upregulated genes showed enrichment of similar processes [68]. These findings indicate that the parity signature set after pregnancy remains until the postmenopausal years. Other development-related genes upregulated in the P breast and confirmed by RT-PCR were RASGRP1 (RAS guanyl-releasing protein 1—calcium and DAG-regulated), EGR3 (early growth response 3), and BHLHE22 (basic helix-loop-helix family member e22). These genes, in addition to differentiation, are known to regulate proliferation and cell growth [2932].
Among the genes classified as developmental, we also found components of the WNT pathway. WNT canonical and non-canonical pathways participate in cell fate determination, cell polarity, adhesion, and motility [33, 34], all important functions in the differentiation of the breast. Differentiation induced by parity has been demonstrated to alter WNT/Notch signaling in mice [35], and we have described alterations in the methylation profile of genes belonging to this pathway and its regulation in the postmenopausal breast [36]. In the current study, we observed two genes of this pathway upregulated in the P breast, FZD8 (frizzled family receptor 8) which is a transmembrane receptor transducing WNT signals, and EAF2 (ELL-associated factor 2), which is important for early embryonic development and critical for adult tissue homeostasis and prevention against tumor initiation [37, 38]. WNT inhibitory factor 1, WIF1, was downregulated in P women, and although methylation of WIF1 has been observed in several tumors [39], including breast cancer [40], this can be an indication that the WNT pathway has an important role in the shifting of the stem cells to a more differentiated status in the P breast, as demonstrated earlier [8, 41]. Yet related to WNT pathway, we found FOXQ1 or forkhead box Q1, constantly downregulated in the P breast. This gene is a direct target of the canonical WNT pathway and its overexpression has been associated with different tumors and cancer cell lines [42]. Suppression of FOXQ1 inhibits cell proliferation, motility/invasion, and epithelial-mesenchymal transition phenotypes in cancer cells [4345]; a similar effect could be predicted in the breast of P women.
Also consistent with our previous study in postmenopausal women, we observed several genes with roles in chromatin remodeling [8]. In the current study, there were four long-term upregulated genes involved in chromatin remodeling: APOBEC3G, TOX, UHRF1, and NAP1L2. These genes interact with chromatin, either by binding with histones or recruiting histone modifiers, influencing cell proliferation and differentiation among other biological processes [4652]. In addition, APOBEC3G (apolipoprotein B mRNA-editing enzyme, catalytic polypeptide-like 3G) is involved in RNA editing [53], and deletion in APOBEC3 gene has been correlated to breast cancer risk [54]. The upregulation of these genes in the breast of P women years after delivery indicates chromatin remodeling is enabling a permanent differentiation of the breast epithelial cells.
We also evaluated whether selected genes from this study are modified in breast cancer cases. Of the genes evaluated, BHLHE22 was the most commonly modified in breast cancer (6.9% of the cases). The other genes were altered in a small percentage of breast cancer cases (Table 6). Of interest is that patients who have some alteration (amplification, deep deletion, or missense mutation) in the tested genes have a lower overall survival (Fig. 6). Although this analysis can indicate that these genes are associated with breast cancer, our interpretation is limited because the parity status of these breast cancer cases is not available.
In addition to genes involved in development and chromatin remodeling, we observed a large number of genes known to participate in immune response. The immune system has several roles in the mammary gland, being important not only for protection against pathogens, but also it is secreted into the milk and participates at the different stages of the gland development, including involution [5558]. Of great interest, most of these immune-related genes were only upregulated in the biopsies collected within 5 years since pregnancy. This observation is consistent with our previous work in postmenopausal women, in which we did not observe enrichment of immune response in an older population [6, 7]. It is also in agreement with previous studies on molecular profiling of pregnancy performed in younger populations that reported changes in immune-response genes [59, 60].
Previous studies have showed differences in expression patterns of immune-related genes at distinct mammary developmental stages, before and/or after pregnancy in humans [59] and rodents [61, 62]. Rodent studies demonstrated that in the first days of mammary gland involution there is activation of genes related to acute-phase response and inflammation [61, 62], followed by activation of monocyte and lymphoid chemokines and immunoglobulin genes [61]. The inflammatory-like environment observed during the breast involution has been proposed as one of the mechanisms that could explain the transient increase in breast cancer risk observed after each pregnancy [57, 63]. In this study, we observed that most of the genes that underwent transient changes in expression enriched biological processes related to activation and development of lymphocytes, mainly T cells (Additional file 2). Only one GO term related to inflammation (GO:0006925: inflammatory cell apoptotic process) was enriched among the transient genes (Additional file 2). This may be due to the fact that biopsies were collected at least 1 year after FTP and/or breastfeeding; thus, we may have missed an early inflammation stage. Both human and murine postpartum mammary glands have an organized influx of immune cells; however, these cells are not observed after 1 year postpartum in human, or 12 weeks in murine [57]. We have performed immunohistochemistry reactions for dendritic cells and T cell activation markers on a subset of our samples. Using antibodies anti-LAMP3 and anti-CD123 for dendritic cells, antigen presenters, we detected few positive cells and no differences between P and NP samples (data not shown). When using anti-CD2 and anti-CD3D, markers of T cell activation, we observed an increase in cells positive for CD3D in P breast. This data suggest that there are differences in the number of activated T cells between P and NP breast (Fig. 7).
Normal pregnancy is characterized by an early expansion of regulatory T cells [64], which modulate immune tolerance during pregnancy [64]. In addition, microchimeric cells of fetal origin that persist in the maternal circulation after delivery are postulated to provide immune surveillance with the contribution of T cells, protecting against breast cancer [6567]. Evidence shows that microchimeric cells are more frequent in healthy women than in breast cancer-affected women, and breast cancers without circulating microchimeric cells are more aggressive [6570]. In this work, we found several upregulated genes related to activation of T cells after FTP (Additional files 2, 3, and 4). This suggests that the activation of T cells in the breast tissue of P women could trigger an early response against transformed cells, destroying them and protecting the mammary gland from neoplastic transformation. Tumor surveillance by the immune system and its impact on disease outcomes in cancer patients, and in breast cancer patients in particular, have been documented [7175]. Finak et al. studied the stromal gene expression and clinical outcomes in breast cancer and observed that the gene set expressed predominantly in the good-outcome cluster was enriched by elements of the T helper type 1 [71]. Eight (CD2, CD8A, XCL1, CD3D, GZMA, CD247, CD48, CD52) of the genes present in this cluster [71] were also upregulated in the parous women. Poor-outcome cluster [71] showed downregulation of CXCL14, which was upregulated in the parous breast, and upregulation of CXCL1 and EDN1, which were downregulated in the parous breast. This overlap between Finak study and ours indicates that the activation of the T cell response is a beneficial mechanism against transformed cells. Winslow et al. described gene sets with better prognostics for triple-negative breast cancers [72]. These gene sets involved cytotoxic immune response, including the genes mentioned above, and HLA encoding genes [72]. HLA-DRA (major histocompatibility complex, class II, DR alpha) and HLA-DPA1 (major histocompatibility complex, class II, DP alpha 1) were constantly activated in our study. The upregulation of HLA-DPA1 has been associated with a benign behavior of certain neurological tumors and related to the immune defense-associated genes [76]. Therefore, a similar role could be attributed to the upregulation of these two genes in the cancer preventative effect of pregnancy in the premenopausal women.
Using tissue from nine women whom arrays could be conducted in epithelial tissue, stromal tissue, and whole tissue, we observed that a linear regression of gene expression in epithelial and stromal tissues predicted gene expression in the whole tissue extremely well (R2 ≥ 0.90 for all subjects). We also observed that the proportion of each tissue in the whole-tissue biopsy sample (as estimated by the linear regression coefficient) varied substantially across women (range for epithelial tissue, 0.09–0.68) (Table 7). This suggests that the P/NP gene expression differences we observed either are present in each of the two tissues (epithelial and stromal) or are present in only one of the two tissues but are of such magnitude that they are observed in whole tissue biopsies, despite the fact that some of the individual biopsies may contain only a small proportion of this tissue type.
When comparing gene expression in stroma versus epithelia, we observed a large number of genes differentially expressed among these two tissues. The epithelia, being a more differentiated tissue, enriched fewer GO terms, and these were mainly involved in epithelial development and tight junction (Additional file 7). The stroma showed enrichment of a broader range of functions, lipid homeostasis, lipid storage, metabolic process, vascularization, and migration to cite a few (Additional file 7). This extensive list of biological process is expected because the stroma is constituted of different types of cells (e.g., adipocytes, fibroblasts, endothelial cells). Among the genes found in the stroma, we did not observe enrichment of many immune-related GO terms, which is consistent with the fact we did not observe a large number of immune cells in the histopathology of these samples. Furthermore, parity status was not included in the comparison of the epithelia vs. stroma. Among the samples used in the LCM analyses (5 NP and 5 P), only one had the FTP less than 5 years before the biopsy, the group in which we found significant enrichment of immune response genes in the P/NP main study. The limited number of successful LCM microarrays did not allow us to perform analyses to understand whether parity induced more changes in expression of genes present in the stromal or epithelial cells of the breast.
This study has several strengths. A major strength is that all women were volunteers from the general population who were free of any breast abnormality. Also, all biopsies were histologically examined to confirm normality of tissue and presence of breast epithelium and stroma. Histological evaluations and gene expression experiments were performed on unidentifiable samples to prevent bias. Our study also had limitations. Because our main analysis was based on whole breast tissue microarrays, we were not able to characterize parity-associated differences in gene expression separately for epithelial and stromal tissues. While this would be of interest, it would require large biopsies in order to obtain a sufficient amount of each tissue type, which is difficult to justify in studies of healthy volunteers. The variability in composition of breast tissue among women adds to the challenge of understanding the mechanisms responsible for the preventive effect of parity and comparison of different studies. Because of the limited sample size, we could not conduct our analysis by time since last pregnancy partitioning our data in discovery/validation. Lastly, these results were generated based on a relatively homogenous population; therefore, confirmation of these results including a more ethnically heterogeneous population is needed.

Conclusions

Altogether, parous premenopausal breast shows a specific transcriptome profile, in which genes controlling chromatin remodeling and cell differentiation are activated after FTP and stay upregulated for many years, supporting the data from postmenopausal parous women previously published [68].
Of great novelty is that this work shows that the genes involved in immune response were in majority related to T cell activation and these were activated soon after the FTP; however, their expression return to levels similar to those observed in the nulliparous breast five or more years after FTP. These transcripts may work in protecting the mammary gland against neoplastically transformed cells through T cells. However, because this immune surveillance appears transitory, we infer that cell differentiation, activated by the genes whose expression was permanently affected by parity, may be the main molecular mechanism responsible for the preventive effect of parity against breast cancer.

Acknowledgements

The authors thank the women of Norrbotten County, Sweden, for their willing contribution to the project, the staff of the Mammography Department, Sunderby Hospital, Luleå, Sweden, and Fox Chase Cancer Center Genomics Facility.

Funding

This work was supported by grant 02-2010-117 from the Avon Foundation for Women Breast Cancer Research Program and by NCI P30-CA006927 to Fox Chase Cancer Center.

Availability of data and materials

The datasets generated and analyzed during the current study are available in the Gene Expression Omnibus (GEO) repository. The data regarding the analysis of P vs. NP has accession number GSE112825 [https://​www.​ncbi.​nlm.​nih.​gov/​geo/​query/​acc.​cgi?​acc=​GSE112825]. The data generated from the LCM substudy has accession number GSE111662 [https://​www.​ncbi.​nlm.​nih.​gov/​geo/​query/​acc.​cgi?​acc=​GSE111662].
This study was reviewed and approved by the Regional Ethical Review Board for Northern Sweden at the University of Umeå, Sweden (Dnr07-156 M with amendments 08-020 and 2010/397-32) and by Internal Review Board of Fox Chase Cancer Center, USA (FCCC-IRB 02-829). All volunteers that were involved in this work signed informed consent to participate in this study.
Not applicable

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
2.
Zurück zum Zitat Cancer CGoHFiB. Breast cancer and breastfeeding: collaborative reanalysis of individual data from 47 epidemiological studies in 30 countries, including 50302 women with breast cancer and 96973 women without the disease. Lancet. 2002;360:187–95.CrossRef Cancer CGoHFiB. Breast cancer and breastfeeding: collaborative reanalysis of individual data from 47 epidemiological studies in 30 countries, including 50302 women with breast cancer and 96973 women without the disease. Lancet. 2002;360:187–95.CrossRef
3.
Zurück zum Zitat Russo J, Tay LK, Russo IH. Differentiation of the mammary gland and susceptibility to carcinogenesis. Breast Cancer Res Treat. 1982;2:5–73.PubMedCrossRef Russo J, Tay LK, Russo IH. Differentiation of the mammary gland and susceptibility to carcinogenesis. Breast Cancer Res Treat. 1982;2:5–73.PubMedCrossRef
4.
Zurück zum Zitat Russo IH, Koszalka M, Russo J. Comparative study of the influence of pregnancy and hormonal treatment on mammary carcinogenesis. Br J Cancer. 1991;64:481–4.PubMedPubMedCentralCrossRef Russo IH, Koszalka M, Russo J. Comparative study of the influence of pregnancy and hormonal treatment on mammary carcinogenesis. Br J Cancer. 1991;64:481–4.PubMedPubMedCentralCrossRef
5.
Zurück zum Zitat Albrektsen G, Heuch I, Hansen S, Kvale G. Breast cancer risk by age at birth, time since birth and time intervals between births: exploring interaction effects. Br J Cancer. 2005;92:167–75.PubMedCrossRef Albrektsen G, Heuch I, Hansen S, Kvale G. Breast cancer risk by age at birth, time since birth and time intervals between births: exploring interaction effects. Br J Cancer. 2005;92:167–75.PubMedCrossRef
6.
Zurück zum Zitat Belitskaya-Levy I, Zeleniuch-Jacquotte A, Russo J, Russo IH, Bordas P, Ahman J, et al. Characterization of a genomic signature of pregnancy identified in the breast. Cancer Prev Res (Phila). 2011;4:1457–64.CrossRef Belitskaya-Levy I, Zeleniuch-Jacquotte A, Russo J, Russo IH, Bordas P, Ahman J, et al. Characterization of a genomic signature of pregnancy identified in the breast. Cancer Prev Res (Phila). 2011;4:1457–64.CrossRef
7.
Zurück zum Zitat Peri S, de Cicco RL, Santucci-Pereira J, Slifker M, Ross EA, Russo IH, et al. Defining the genomic signature of the parous breast. BMC Med Genet. 2012;5:46. Peri S, de Cicco RL, Santucci-Pereira J, Slifker M, Ross EA, Russo IH, et al. Defining the genomic signature of the parous breast. BMC Med Genet. 2012;5:46.
8.
Zurück zum Zitat Russo J, Santucci-Pereira J, de Cicco RL, Sheriff F, Russo PA, Peri S, et al. Pregnancy-induced chromatin remodeling in the breast of postmenopausal women. Int J Cancer. 2012;131:1059–70.PubMedPubMedCentralCrossRef Russo J, Santucci-Pereira J, de Cicco RL, Sheriff F, Russo PA, Peri S, et al. Pregnancy-induced chromatin remodeling in the breast of postmenopausal women. Int J Cancer. 2012;131:1059–70.PubMedPubMedCentralCrossRef
9.
Zurück zum Zitat Bolstad BM, Collin F, Brettschneider J, Simpson K, Cope L, Irizarry RA, et al. Quality assessment of Affymetrix GeneChip Data. In: Gentleman R, Carey V, Huber W, Irizarry R, Dudoit S, editors. Bioinformatics and computational biology solutions using R and bioconductor. New York: Springer; 2005. p. 33–47.CrossRef Bolstad BM, Collin F, Brettschneider J, Simpson K, Cope L, Irizarry RA, et al. Quality assessment of Affymetrix GeneChip Data. In: Gentleman R, Carey V, Huber W, Irizarry R, Dudoit S, editors. Bioinformatics and computational biology solutions using R and bioconductor. New York: Springer; 2005. p. 33–47.CrossRef
11.
Zurück zum Zitat Gentleman RC, Carey VJ, Bates DM, Bolstad B, Dettling M, Dudoit S, et al. Bioconductor: open software development for computational biology and bioinformatics. Genome Biol. 2004;5:R80.PubMedPubMedCentralCrossRef Gentleman RC, Carey VJ, Bates DM, Bolstad B, Dettling M, Dudoit S, et al. Bioconductor: open software development for computational biology and bioinformatics. Genome Biol. 2004;5:R80.PubMedPubMedCentralCrossRef
12.
Zurück zum Zitat Johnson WE, Li C, Rabinovic A. Adjusting batch effects in microarray expression data using empirical Bayes methods. Biostatistics. 2007;8:118–27.PubMedCrossRef Johnson WE, Li C, Rabinovic A. Adjusting batch effects in microarray expression data using empirical Bayes methods. Biostatistics. 2007;8:118–27.PubMedCrossRef
13.
Zurück zum Zitat Leek JT, Johnson WE, Parker HS, Jaffe AE, Storey JD. The sva package for removing batch effects and other unwanted variation in high-throughput experiments. Bioinformatics. 2012;28:882–3.PubMedPubMedCentralCrossRef Leek JT, Johnson WE, Parker HS, Jaffe AE, Storey JD. The sva package for removing batch effects and other unwanted variation in high-throughput experiments. Bioinformatics. 2012;28:882–3.PubMedPubMedCentralCrossRef
15.
Zurück zum Zitat Saeed AI, Sharov V, White J, Li J, Liang W, Bhagabati N, et al. TM4: a free, open-source system for microarray data management and analysis. Biotechniques. 2003;34:374–8.PubMedCrossRef Saeed AI, Sharov V, White J, Li J, Liang W, Bhagabati N, et al. TM4: a free, open-source system for microarray data management and analysis. Biotechniques. 2003;34:374–8.PubMedCrossRef
16.
Zurück zum Zitat Falcon S, Gentleman R. Using GOstats to test gene lists for GO term association. Bioinformatics. 2007;23:257–8.PubMedCrossRef Falcon S, Gentleman R. Using GOstats to test gene lists for GO term association. Bioinformatics. 2007;23:257–8.PubMedCrossRef
17.
Zurück zum Zitat Ashburner M, Ball CA, Blake JA, Botstein D, Butler H, Cherry JM, et al. Gene ontology: tool for the unification of biology. The Gene Ontology Consortium. Nat Genet. 2000;25:25–9.PubMedPubMedCentralCrossRef Ashburner M, Ball CA, Blake JA, Botstein D, Butler H, Cherry JM, et al. Gene ontology: tool for the unification of biology. The Gene Ontology Consortium. Nat Genet. 2000;25:25–9.PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal. 2013;6:pl1.PubMedPubMedCentralCrossRef Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal. 2013;6:pl1.PubMedPubMedCentralCrossRef
19.
Zurück zum Zitat Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2012;2:401–4.CrossRefPubMed Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2012;2:401–4.CrossRefPubMed
20.
Zurück zum Zitat Coudry RA, Meireles SI, Stoyanova R, Cooper HS, Carpino A, Wang X, et al. Successful application of microarray technology to microdissected formalin-fixed, paraffin-embedded tissue. J Mol Diagn. 2007;9:70–9.PubMedPubMedCentralCrossRef Coudry RA, Meireles SI, Stoyanova R, Cooper HS, Carpino A, Wang X, et al. Successful application of microarray technology to microdissected formalin-fixed, paraffin-embedded tissue. J Mol Diagn. 2007;9:70–9.PubMedPubMedCentralCrossRef
21.
Zurück zum Zitat North AJ, Chidgey MA, Clarke JP, Bardsley WG, Garrod DR. Distinct desmocollin isoforms occur in the same desmosomes and show reciprocally graded distributions in bovine nasal epidermis. Proc Natl Acad Sci U S A. 1996;93:7701–5.PubMedPubMedCentralCrossRef North AJ, Chidgey MA, Clarke JP, Bardsley WG, Garrod DR. Distinct desmocollin isoforms occur in the same desmosomes and show reciprocally graded distributions in bovine nasal epidermis. Proc Natl Acad Sci U S A. 1996;93:7701–5.PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat Cui T, Chen Y, Yang L, Knosel T, Huber O, Pacyna-Gengelbach M, et al. The p53 target gene desmocollin 3 acts as a novel tumor suppressor through inhibiting EGFR/ERK pathway in human lung cancer. Carcinogenesis. 2012;33:2326–33.PubMedCrossRef Cui T, Chen Y, Yang L, Knosel T, Huber O, Pacyna-Gengelbach M, et al. The p53 target gene desmocollin 3 acts as a novel tumor suppressor through inhibiting EGFR/ERK pathway in human lung cancer. Carcinogenesis. 2012;33:2326–33.PubMedCrossRef
24.
Zurück zum Zitat Chen J, O'Shea C, Fitzpatrick JE, Koster MI, Koch PJ. Loss of Desmocollin 3 in skin tumor development and progression. Mol Carcinog. 2012;51:535–45.PubMedCrossRef Chen J, O'Shea C, Fitzpatrick JE, Koster MI, Koch PJ. Loss of Desmocollin 3 in skin tumor development and progression. Mol Carcinog. 2012;51:535–45.PubMedCrossRef
25.
Zurück zum Zitat Knosel T, Chen Y, Hotovy S, Settmacher U, Altendorf-Hofmann A, Petersen I. Loss of desmocollin 1-3 and homeobox genes PITX1 and CDX2 are associated with tumor progression and survival in colorectal carcinoma. Int J Color Dis. 2012;27:1391–9.CrossRef Knosel T, Chen Y, Hotovy S, Settmacher U, Altendorf-Hofmann A, Petersen I. Loss of desmocollin 1-3 and homeobox genes PITX1 and CDX2 are associated with tumor progression and survival in colorectal carcinoma. Int J Color Dis. 2012;27:1391–9.CrossRef
26.
Zurück zum Zitat Oshiro MM, Kim CJ, Wozniak RJ, Junk DJ, Munoz-Rodriguez JL, Burr JA, et al. Epigenetic silencing of DSC3 is a common event in human breast cancer. Breast Cancer Res. 2005;7:R669–80.PubMedPubMedCentralCrossRef Oshiro MM, Kim CJ, Wozniak RJ, Junk DJ, Munoz-Rodriguez JL, Burr JA, et al. Epigenetic silencing of DSC3 is a common event in human breast cancer. Breast Cancer Res. 2005;7:R669–80.PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Vasiliu D, Clamons S, McDonough M, Rabe B, Saha M. A regression-based differential expression detection algorithm for microarray studies with ultra-low sample size. PLoS One. 2015;10:e0118198.PubMedPubMedCentralCrossRef Vasiliu D, Clamons S, McDonough M, Rabe B, Saha M. A regression-based differential expression detection algorithm for microarray studies with ultra-low sample size. PLoS One. 2015;10:e0118198.PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Braga-Neto UM, Dougherty ER. Is cross-validation valid for small-sample microarray classification? Bioinformatics. 2004;20:374–80.PubMedCrossRef Braga-Neto UM, Dougherty ER. Is cross-validation valid for small-sample microarray classification? Bioinformatics. 2004;20:374–80.PubMedCrossRef
29.
Zurück zum Zitat Huang H, Jin T, Wang L, Wang F, Zhang R, Pan Y, et al. The RAS guanyl nucleotide-releasing protein RasGRP1 is involved in lymphatic development in zebrafish. J Biol Chem. 2013;288:2355–64.PubMedCrossRef Huang H, Jin T, Wang L, Wang F, Zhang R, Pan Y, et al. The RAS guanyl nucleotide-releasing protein RasGRP1 is involved in lymphatic development in zebrafish. J Biol Chem. 2013;288:2355–64.PubMedCrossRef
30.
Zurück zum Zitat Kortum RL, Rouquette-Jazdanian AK, Samelson LE. Ras and extracellular signal-regulated kinase signaling in thymocytes and T cells. Trends Immunol. 2013;34:259–68.PubMedCrossRef Kortum RL, Rouquette-Jazdanian AK, Samelson LE. Ras and extracellular signal-regulated kinase signaling in thymocytes and T cells. Trends Immunol. 2013;34:259–68.PubMedCrossRef
31.
Zurück zum Zitat To SQ, Knower KC, Clyne CD. NFkappaB and MAPK signalling pathways mediate TNFalpha-induced early growth response gene transcription leading to aromatase expression. Biochem Biophys Res Commun. 2013;433:96–101.PubMedCrossRef To SQ, Knower KC, Clyne CD. NFkappaB and MAPK signalling pathways mediate TNFalpha-induced early growth response gene transcription leading to aromatase expression. Biochem Biophys Res Commun. 2013;433:96–101.PubMedCrossRef
32.
Zurück zum Zitat Dugas JC, Ibrahim A, Barres BA. The T3-induced gene KLF9 regulates oligodendrocyte differentiation and myelin regeneration. Mol Cell Neurosci. 2012;50:45–57.PubMedPubMedCentralCrossRef Dugas JC, Ibrahim A, Barres BA. The T3-induced gene KLF9 regulates oligodendrocyte differentiation and myelin regeneration. Mol Cell Neurosci. 2012;50:45–57.PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Wang HQ, Xu ML, Ma J, Zhang Y, Xie CH. Frizzled-8 as a putative therapeutic target in human lung cancer. Biochem Biophys Res Commun. 2012;417:62–6.PubMedCrossRef Wang HQ, Xu ML, Ma J, Zhang Y, Xie CH. Frizzled-8 as a putative therapeutic target in human lung cancer. Biochem Biophys Res Commun. 2012;417:62–6.PubMedCrossRef
34.
Zurück zum Zitat Katoh M. WNT signaling in stem cell biology and regenerative medicine. Curr Drug Targets. 2008;9:565–70.PubMedCrossRef Katoh M. WNT signaling in stem cell biology and regenerative medicine. Curr Drug Targets. 2008;9:565–70.PubMedCrossRef
35.
Zurück zum Zitat Meier-Abt F, Milani E, Roloff T, Brinkhaus H, Duss S, Meyer DS, et al. Parity induces differentiation and reduces Wnt/Notch signaling ratio and proliferation potential of basal stem/progenitor cells isolated from mouse mammary epithelium. Breast Cancer Res. 2013;15:R36.PubMedPubMedCentralCrossRef Meier-Abt F, Milani E, Roloff T, Brinkhaus H, Duss S, Meyer DS, et al. Parity induces differentiation and reduces Wnt/Notch signaling ratio and proliferation potential of basal stem/progenitor cells isolated from mouse mammary epithelium. Breast Cancer Res. 2013;15:R36.PubMedPubMedCentralCrossRef
37.
38.
Zurück zum Zitat Su F, Pascal LE, Xiao W, Wang Z. Tumor suppressor U19/EAF2 regulates thrombospondin-1 expression via p53. Oncogene. 2010;29:421–31.PubMedCrossRef Su F, Pascal LE, Xiao W, Wang Z. Tumor suppressor U19/EAF2 regulates thrombospondin-1 expression via p53. Oncogene. 2010;29:421–31.PubMedCrossRef
39.
Zurück zum Zitat Wissmann C, Wild PJ, Kaiser S, Roepcke S, Stoehr R, Woenckhaus M, et al. WIF1, a component of the Wnt pathway, is down-regulated in prostate, breast, lung, and bladder cancer. J Pathol. 2003;201:204–12.PubMedCrossRef Wissmann C, Wild PJ, Kaiser S, Roepcke S, Stoehr R, Woenckhaus M, et al. WIF1, a component of the Wnt pathway, is down-regulated in prostate, breast, lung, and bladder cancer. J Pathol. 2003;201:204–12.PubMedCrossRef
40.
Zurück zum Zitat Ai L, Tao Q, Zhong S, Fields CR, Kim WJ, Lee MW, et al. Inactivation of Wnt inhibitory factor-1 (WIF1) expression by epigenetic silencing is a common event in breast cancer. Carcinogenesis. 2006;27:1341–8.PubMedCrossRef Ai L, Tao Q, Zhong S, Fields CR, Kim WJ, Lee MW, et al. Inactivation of Wnt inhibitory factor-1 (WIF1) expression by epigenetic silencing is a common event in breast cancer. Carcinogenesis. 2006;27:1341–8.PubMedCrossRef
41.
Zurück zum Zitat Russo J, Balogh GA, Chen J, Fernandez SV, Fernbaugh R, Heulings R, et al. The concept of stem cell in the mammary gland and its implication in morphogenesis, cancer and prevention. Front Biosci. 2006;11:151–72.PubMedCrossRef Russo J, Balogh GA, Chen J, Fernandez SV, Fernbaugh R, Heulings R, et al. The concept of stem cell in the mammary gland and its implication in morphogenesis, cancer and prevention. Front Biosci. 2006;11:151–72.PubMedCrossRef
42.
Zurück zum Zitat Christensen J, Bentz S, Sengstag T, Shastri VP, Anderle P. FOXQ1, a novel target of the Wnt pathway and a new marker for activation of Wnt signaling in solid tumors. PLoS One. 2013;8:e60051.PubMedPubMedCentralCrossRef Christensen J, Bentz S, Sengstag T, Shastri VP, Anderle P. FOXQ1, a novel target of the Wnt pathway and a new marker for activation of Wnt signaling in solid tumors. PLoS One. 2013;8:e60051.PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat Sehrawat A, Kim SH, Vogt A, Singh SV. Suppression of FOXQ1 in benzyl isothiocyanate-mediated inhibition of epithelial-mesenchymal transition in human breast cancer cells. Carcinogenesis. 2013;34:864–73.PubMedCrossRef Sehrawat A, Kim SH, Vogt A, Singh SV. Suppression of FOXQ1 in benzyl isothiocyanate-mediated inhibition of epithelial-mesenchymal transition in human breast cancer cells. Carcinogenesis. 2013;34:864–73.PubMedCrossRef
44.
Zurück zum Zitat Qiao Y, Jiang X, Lee ST, Karuturi RK, Hooi SC, Yu Q. FOXQ1 regulates epithelial-mesenchymal transition in human cancers. Cancer Res. 2011;71:3076–86.PubMedCrossRef Qiao Y, Jiang X, Lee ST, Karuturi RK, Hooi SC, Yu Q. FOXQ1 regulates epithelial-mesenchymal transition in human cancers. Cancer Res. 2011;71:3076–86.PubMedCrossRef
45.
46.
Zurück zum Zitat Tessema M, Yingling CM, Grimes MJ, Thomas CL, Liu Y, Leng S, et al. Differential epigenetic regulation of TOX subfamily high mobility group box genes in lung and breast cancers. PLoS One. 2012;7:e34850.PubMedPubMedCentralCrossRef Tessema M, Yingling CM, Grimes MJ, Thomas CL, Liu Y, Leng S, et al. Differential epigenetic regulation of TOX subfamily high mobility group box genes in lung and breast cancers. PLoS One. 2012;7:e34850.PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat Aliahmad P, de la Torre B, Kaye J. Shared dependence on the DNA-binding factor TOX for the development of lymphoid tissue-inducer cell and NK cell lineages. Nat Immunol. 2010;11:945–52.PubMedPubMedCentralCrossRef Aliahmad P, de la Torre B, Kaye J. Shared dependence on the DNA-binding factor TOX for the development of lymphoid tissue-inducer cell and NK cell lineages. Nat Immunol. 2010;11:945–52.PubMedPubMedCentralCrossRef
48.
Zurück zum Zitat Aliahmad P, Kadavallore A, de la Torre B, Kappes D, Kaye J. TOX is required for development of the CD4 T cell lineage gene program. J Immunol. 2011;187:5931–40.PubMedCrossRef Aliahmad P, Kadavallore A, de la Torre B, Kappes D, Kaye J. TOX is required for development of the CD4 T cell lineage gene program. J Immunol. 2011;187:5931–40.PubMedCrossRef
49.
Zurück zum Zitat Unoki M, Nishidate T, Nakamura Y. ICBP90, an E2F-1 target, recruits HDAC1 and binds to methyl-CpG through its SRA domain. Oncogene. 2004;23:7601–10.PubMedCrossRef Unoki M, Nishidate T, Nakamura Y. ICBP90, an E2F-1 target, recruits HDAC1 and binds to methyl-CpG through its SRA domain. Oncogene. 2004;23:7601–10.PubMedCrossRef
50.
Zurück zum Zitat Jin W, Chen L, Chen Y, Xu SG, Di GH, Yin WJ, et al. UHRF1 is associated with epigenetic silencing of BRCA1 in sporadic breast cancer. Breast Cancer Res Treat. 2010;123:359–73.PubMedCrossRef Jin W, Chen L, Chen Y, Xu SG, Di GH, Yin WJ, et al. UHRF1 is associated with epigenetic silencing of BRCA1 in sporadic breast cancer. Breast Cancer Res Treat. 2010;123:359–73.PubMedCrossRef
51.
Zurück zum Zitat Attia M, Forster A, Rachez C, Freemont P, Avner P, Rogner UC. Interaction between nucleosome assembly protein 1-like family members. J Mol Biol. 2011;407:647–60.PubMedCrossRef Attia M, Forster A, Rachez C, Freemont P, Avner P, Rogner UC. Interaction between nucleosome assembly protein 1-like family members. J Mol Biol. 2011;407:647–60.PubMedCrossRef
52.
Zurück zum Zitat Attia M, Rachez C, De Pauw A, Avner P, Rogner UC. Nap1l2 promotes histone acetylation activity during neuronal differentiation. Mol Cell Biol. 2007;27:6093–102.PubMedPubMedCentralCrossRef Attia M, Rachez C, De Pauw A, Avner P, Rogner UC. Nap1l2 promotes histone acetylation activity during neuronal differentiation. Mol Cell Biol. 2007;27:6093–102.PubMedPubMedCentralCrossRef
53.
Zurück zum Zitat Lackey L, Law EK, Brown WL, Harris RS. Subcellular localization of the APOBEC3 proteins during mitosis and implications for genomic DNA deamination. Cell Cycle. 2013;12:762–72.PubMedPubMedCentralCrossRef Lackey L, Law EK, Brown WL, Harris RS. Subcellular localization of the APOBEC3 proteins during mitosis and implications for genomic DNA deamination. Cell Cycle. 2013;12:762–72.PubMedPubMedCentralCrossRef
54.
Zurück zum Zitat Long J, Delahanty RJ, Li G, Gao YT, Lu W, Cai Q, et al. A common deletion in the APOBEC3 genes and breast cancer risk. J Natl Cancer Inst. 2013;105:573–9.PubMedPubMedCentralCrossRef Long J, Delahanty RJ, Li G, Gao YT, Lu W, Cai Q, et al. A common deletion in the APOBEC3 genes and breast cancer risk. J Natl Cancer Inst. 2013;105:573–9.PubMedPubMedCentralCrossRef
55.
Zurück zum Zitat Monks J, Geske FJ, Lehman L, Fadok VA. Do inflammatory cells participate in mammary gland involution? J Mammary Gland Biol Neoplasia. 2002;7:163–76.PubMedCrossRef Monks J, Geske FJ, Lehman L, Fadok VA. Do inflammatory cells participate in mammary gland involution? J Mammary Gland Biol Neoplasia. 2002;7:163–76.PubMedCrossRef
56.
Zurück zum Zitat Csanaky K, Doppler W, Tamas A, Kovacs K, Toth G, Reglodi D. Influence of terminal differentiation and PACAP on the cytokine, chemokine, and growth factor secretion of mammary epithelial cells. J Mol Neurosci. 2014;52:28–36.PubMedCrossRef Csanaky K, Doppler W, Tamas A, Kovacs K, Toth G, Reglodi D. Influence of terminal differentiation and PACAP on the cytokine, chemokine, and growth factor secretion of mammary epithelial cells. J Mol Neurosci. 2014;52:28–36.PubMedCrossRef
57.
Zurück zum Zitat Martinson HA, Jindal S, Durand-Rougely C, Borges VF, Schedin P. Wound healing-like immune program facilitates postpartum mammary gland involution and tumor progression. Int J Cancer. 2015;136:1803–13.PubMedCrossRef Martinson HA, Jindal S, Durand-Rougely C, Borges VF, Schedin P. Wound healing-like immune program facilitates postpartum mammary gland involution and tumor progression. Int J Cancer. 2015;136:1803–13.PubMedCrossRef
58.
Zurück zum Zitat Plaks V, Boldajipour B, Linnemann JR, Nguyen NH, Kersten K, Wolf Y, et al. Adaptive immune regulation of mammary postnatal organogenesis. Dev Cell. 2015;34:493–504.PubMedPubMedCentralCrossRef Plaks V, Boldajipour B, Linnemann JR, Nguyen NH, Kersten K, Wolf Y, et al. Adaptive immune regulation of mammary postnatal organogenesis. Dev Cell. 2015;34:493–504.PubMedPubMedCentralCrossRef
59.
Zurück zum Zitat Asztalos S, Gann PH, Hayes MK, Nonn L, Beam CA, Dai Y, et al. Gene expression patterns in the human breast after pregnancy. Cancer Prev Res (Phila). 2010;3:301–11.CrossRef Asztalos S, Gann PH, Hayes MK, Nonn L, Beam CA, Dai Y, et al. Gene expression patterns in the human breast after pregnancy. Cancer Prev Res (Phila). 2010;3:301–11.CrossRef
60.
Zurück zum Zitat Rotunno M, Sun X, Figueroa J, Sherman ME, Garcia-Closas M, Meltzer P, et al. Parity-related molecular signatures and breast cancer subtypes by estrogen receptor status. Breast Cancer Res. 2014;16:R74.PubMedPubMedCentralCrossRef Rotunno M, Sun X, Figueroa J, Sherman ME, Garcia-Closas M, Meltzer P, et al. Parity-related molecular signatures and breast cancer subtypes by estrogen receptor status. Breast Cancer Res. 2014;16:R74.PubMedPubMedCentralCrossRef
61.
Zurück zum Zitat Clarkson RW, Wayland MT, Lee J, Freeman T, Watson CJ. Gene expression profiling of mammary gland development reveals putative roles for death receptors and immune mediators in post-lactational regression. Breast Cancer Res. 2004;6:R92–109.PubMedCrossRef Clarkson RW, Wayland MT, Lee J, Freeman T, Watson CJ. Gene expression profiling of mammary gland development reveals putative roles for death receptors and immune mediators in post-lactational regression. Breast Cancer Res. 2004;6:R92–109.PubMedCrossRef
62.
Zurück zum Zitat Stein T, Morris JS, Davies CR, Weber-Hall SJ, Duffy MA, Heath VJ, et al. Involution of the mouse mammary gland is associated with an immune cascade and an acute-phase response, involving LBP, CD14 and STAT3. Breast Cancer Res. 2004;6:R75–91.PubMedCrossRef Stein T, Morris JS, Davies CR, Weber-Hall SJ, Duffy MA, Heath VJ, et al. Involution of the mouse mammary gland is associated with an immune cascade and an acute-phase response, involving LBP, CD14 and STAT3. Breast Cancer Res. 2004;6:R75–91.PubMedCrossRef
63.
Zurück zum Zitat Liu Q, Wuu J, Lambe M, Hsieh SF, Ekbom A, Hsieh CC. Transient increase in breast cancer risk after giving birth: postpartum period with the highest risk (Sweden). Cancer Causes Control. 2002;13:299–305.PubMedCrossRef Liu Q, Wuu J, Lambe M, Hsieh SF, Ekbom A, Hsieh CC. Transient increase in breast cancer risk after giving birth: postpartum period with the highest risk (Sweden). Cancer Causes Control. 2002;13:299–305.PubMedCrossRef
64.
Zurück zum Zitat Schumacher A, Heinze K, Witte J, Poloski E, Linzke N, Woidacki K, et al. Human chorionic gonadotropin as a central regulator of pregnancy immune tolerance. J Immunol. 2013;190:2650–8.PubMedCrossRef Schumacher A, Heinze K, Witte J, Poloski E, Linzke N, Woidacki K, et al. Human chorionic gonadotropin as a central regulator of pregnancy immune tolerance. J Immunol. 2013;190:2650–8.PubMedCrossRef
65.
Zurück zum Zitat Gadi VK. Fetal microchimerism in breast from women with and without breast cancer. Breast Cancer Res Treat. 2010;121:241–4.PubMedCrossRef Gadi VK. Fetal microchimerism in breast from women with and without breast cancer. Breast Cancer Res Treat. 2010;121:241–4.PubMedCrossRef
67.
Zurück zum Zitat Gadi VK, Nelson JL. Fetal microchimerism in women with breast cancer. Cancer Res. 2007;67:9035–8.PubMedCrossRef Gadi VK, Nelson JL. Fetal microchimerism in women with breast cancer. Cancer Res. 2007;67:9035–8.PubMedCrossRef
69.
Zurück zum Zitat Boyon C, Collinet P, Boulanger L, Rubod C, Lucot JP, Vinatier D. Fetal microchimerism: benevolence or malevolence for the mother? Eur J Obstet Gynecol Reprod Biol. 2011;158:148–52.PubMedCrossRef Boyon C, Collinet P, Boulanger L, Rubod C, Lucot JP, Vinatier D. Fetal microchimerism: benevolence or malevolence for the mother? Eur J Obstet Gynecol Reprod Biol. 2011;158:148–52.PubMedCrossRef
70.
Zurück zum Zitat Kallenbach LR, Johnson KL, Bianchi DW. Fetal cell microchimerism and cancer: a nexus of reproduction, immunology, and tumor biology. Cancer Res. 2011;71:8–12.PubMedPubMedCentralCrossRef Kallenbach LR, Johnson KL, Bianchi DW. Fetal cell microchimerism and cancer: a nexus of reproduction, immunology, and tumor biology. Cancer Res. 2011;71:8–12.PubMedPubMedCentralCrossRef
71.
Zurück zum Zitat Finak G, Bertos N, Pepin F, Sadekova S, Souleimanova M, Zhao H, et al. Stromal gene expression predicts clinical outcome in breast cancer. Nat Med. 2008;14:518–27.PubMedCrossRef Finak G, Bertos N, Pepin F, Sadekova S, Souleimanova M, Zhao H, et al. Stromal gene expression predicts clinical outcome in breast cancer. Nat Med. 2008;14:518–27.PubMedCrossRef
73.
Zurück zum Zitat Nagalla S, Chou JW, Willingham MC, Ruiz J, Vaughn JP, Dubey P, et al. Interactions between immunity, proliferation and molecular subtype in breast cancer prognosis. Genome Biol. 2013;14:R34.PubMedPubMedCentralCrossRef Nagalla S, Chou JW, Willingham MC, Ruiz J, Vaughn JP, Dubey P, et al. Interactions between immunity, proliferation and molecular subtype in breast cancer prognosis. Genome Biol. 2013;14:R34.PubMedPubMedCentralCrossRef
74.
Zurück zum Zitat Farmer P, Bonnefoi H, Anderle P, Cameron D, Wirapati P, Becette V, et al. A stroma-related gene signature predicts resistance to neoadjuvant chemotherapy in breast cancer. Nat Med. 2009;15:68–74.PubMedCrossRef Farmer P, Bonnefoi H, Anderle P, Cameron D, Wirapati P, Becette V, et al. A stroma-related gene signature predicts resistance to neoadjuvant chemotherapy in breast cancer. Nat Med. 2009;15:68–74.PubMedCrossRef
75.
Zurück zum Zitat Mahmoud SM, Paish EC, Powe DG, Macmillan RD, Grainge MJ, Lee AH, et al. Tumor-infiltrating CD8+ lymphocytes predict clinical outcome in breast cancer. J Clin Oncol. 2011;29:1949–55.CrossRefPubMed Mahmoud SM, Paish EC, Powe DG, Macmillan RD, Grainge MJ, Lee AH, et al. Tumor-infiltrating CD8+ lymphocytes predict clinical outcome in breast cancer. J Clin Oncol. 2011;29:1949–55.CrossRefPubMed
76.
Zurück zum Zitat Huang H, Hara A, Homma T, Yonekawa Y, Ohgaki H. Altered expression of immune defense genes in pilocytic astrocytomas. J Neuropathol Exp Neurol. 2005;64:891–901.PubMedCrossRef Huang H, Hara A, Homma T, Yonekawa Y, Ohgaki H. Altered expression of immune defense genes in pilocytic astrocytomas. J Neuropathol Exp Neurol. 2005;64:891–901.PubMedCrossRef
Metadaten
Titel
Genomic signature of parity in the breast of premenopausal women
verfasst von
Julia Santucci-Pereira
Anne Zeleniuch-Jacquotte
Yelena Afanasyeva
Hua Zhong
Michael Slifker
Suraj Peri
Eric A. Ross
Ricardo López de Cicco
Yubo Zhai
Theresa Nguyen
Fathima Sheriff
Irma H. Russo
Yanrong Su
Alan A. Arslan
Pal Bordas
Per Lenner
Janet Åhman
Anna Stina Landström Eriksson
Robert Johansson
Göran Hallmans
Paolo Toniolo
Jose Russo
Publikationsdatum
01.12.2019
Verlag
BioMed Central
Erschienen in
Breast Cancer Research / Ausgabe 1/2019
Elektronische ISSN: 1465-542X
DOI
https://doi.org/10.1186/s13058-019-1128-x

Weitere Artikel der Ausgabe 1/2019

Breast Cancer Research 1/2019 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.