Nothing Special   »   [go: up one dir, main page]

Skip to main content
Reproductive Sciences logoLink to Reproductive Sciences
. 2011 Mar;18(3):229–251. doi: 10.1177/1933719110386241

Molecular Evidence for Differences in Endometrium in Severe Versus Mild Endometriosis

Lusine Aghajanova 1, Linda C Giudice 1,
PMCID: PMC3118406  NIHMSID: NIHMS287162  PMID: 21063030

Abstract

Women with stage III/IV versus stage I/II endometriosis have lower implantation and pregnancy rates in natural and assisted reproduction cycles. To elucidate potential molecular mechanisms underlying these clinical observations, herein we investigated the transcriptome of eutopic endometrium across the menstrual cycle in the setting of severe versus mild endometriosis. Proliferative (PE), early secretory (ESE), and mid-secretory (MSE) endometrial tissues were obtained from 63 participants with endometriosis (19 mild and 44 severe). Purified RNA was subjected to microarray analysis using the Gene 1.0 ST Affymetrix platform. Data were analyzed with GeneSpring and Ingenuity Pathway Analysis and subsequently validated. Comparison of differentially regulated genes, analyzed by cycle phase, revealed dysregulation of progesterone and/or cyclic adenosine monophosphate (cAMP)-regulated genes and genes related to thyroid hormone action and metabolism. Also, members of the epidermal growth factor receptor (EGFR) signaling pathway were observed, with the greatest upregulation of EGFR in severe versus mild disease during the early secretory phase. The extracellular matrix proteoglycan versican (VCAN), which regulates cell proliferation and apoptosis, was the most highly expressed gene in severe versus mild disease. Upregulation of microRNA 21 (MIR21) and DICER1 transcripts suggests roles for microRNAs (miRNAs) in the pathogenesis of severe versus mild endometriosis, potentially through regulation of gene silencing and epigenetic mechanisms. These observed differences in transcriptomic signatures and signaling pathways may result in poorly programmed endometrium during the cycle, contributing to lower implantation and pregnancy rates in women with severe versus mild endometriosis.

Keywords: severe endometriosis, mild endometriosis, eutopic endometrium, microarray, transcriptome

Introduction

Endometriosis is a benign gynecologic disease characterized by endometrial-like tissue (epithelium and stroma) outside the uterus. It affects primarily women of reproductive age and presents with pelvic pain and infertility.1,2 Endometriosis is diagnosed mainly by visualization at surgery, and the revisedAmerican Society for Reproductive Medicine (ASRM) staging system recognizes minimal, mild, moderate, and severe (I-IV) stages of disease, based on the number and character of peritoneal lesions, ovarian and other organ involvement, and presence, type, and extent of adhesions.3,4 Although peritoneal, ovarian and rectovaginal endometriotic lesions are considered distinct entities with different pathogenesis,57 mild and severe stages of peritoneal endometriosis may also be distinct disorders, though the supporting data are limited. However, there is clinical evidence that embryonic implantation rates differ in women with severe versus mild endometriosis (see below), suggesting that the eutopic endometrium is different functionally and biochemically in these 2 types of endometriosis.

Women with moderate–severe endometriosis have more difficulty conceiving, compared to those with minimal–mild disease.8 Also, women with stage III/IV endometriosis have significantly lower implantation rates (13.7% vs 28.3%, respectively; P < .05) and pregnancy rates (22.6% vs 40.0%, respectively; P < .01) but not fertilization or miscarriage rates, compared to women with stage I/II endometriosis.9 A meta-analysis of 22 published studies on endometriosis and in vitro fertilization (IVF) outcomes showed that IVF pregnancy rates are significantly lower in women with severe versus mild endometriosis (13.84% vs 21.12%, respectively; P < .001),10 underscoring a potential endometrial origin of these differences. Also, participants with advanced disease demonstrate diminished ovarian response and higher cancellation rates in IVF cycles, but improved implantation, pregnancy, miscarriage, and delivery rates, after surgery, similar to those for women with tubal factor infertility,11 suggesting that removal of disease improves endometrial receptivity.

We have previously compared the transcriptome of eutopic endometrium from women with minimal/mild disease with the endometrium from women without disease during the window of implantation (mid-secretory endometrium [MSE])12 and also the endometrial transcriptome from women with moderate/severe disease compared with no disease in proliferative (PE), early secretory endometrium (ESE), and MSE.13 Based on these and other studies,14 endometrium from women with endometriosis appears to differ from that of disease-free women.12,13,15 Herein, we compared the transcriptome of eutopic endometrium from women with severe versus mild endometriosis at different times in the menstrual cycle, in an attempt to understand the differences and their potential roles contributing to the pathophysiology of infertility in women with endometriosis.

Materials and Methods

Study Participants

The study was approved by the Committee on Human Research of the University of California−San Francisco (UCSF) and the Stanford University Committee on the Use of Human Subjects in Medical Research. Samples were obtained from the National Institute of Health Specialized Cooperative Centers Program in Reproduction and Infertility Research (NIH SCCPRR) Human Endometrial Tissue and DNA Bank at UCSF. Endometrial tissue was obtained from 12 participants without endometriosis undergoing endometrial biopsy or hysterectomy for benign disorders not related to endometrial pathology (used in immunohistochemistry experiments) and from 63 participants with endometriosis undergoing endometrial biopsy for infertility evaluation or hysterectomy for treatment of severe pelvic pain and extensive endometriosis (Table 1 ). All participants were documented not to be pregnant and not to have had hormonal treatment for at least 3 months before surgery. Staging of endometriosis was performed according to the revised American Fertility Society classification system.3,4 Of the 63 participants, 19 had mild and 44 had severe endometriosis. The majority of endometriosis samples were obtained by endometrial biopsy, whereas the majority of control (no endometriosis) samples were obtained after hysterectomy (Table 1). (Note our previous studies demonstrated that sampling technique does not affect the endometrial transcriptome.16) The mean ages (years) of patients were: mild endometriosis group, 35.7 ± 1.4; severe endometriosis group, 35.2 ± 1.2l (P > .05). Menstrual cycle phase was determined based on histological evaluation of the tissue by 3 independent readers and according to the Noyes' criteria.17

Table 1.

Characteristics of Participants and Endometrial Tissue Samples in the Study

Participant ID Cycle Phase Experiment How Endometrium was Obtained Age Ethnicity
Whole tissue biopsy used for microarray analysis and validations
 Mild endometriosis
  609 PE Microarray, IHC Endometrial biopsy 27 Mixed
  621 PE Microarray, IHC Hysterectomy 37 Caucasian
  658 PE Microarray, QPCR, IHC Endometrial biopsy 36 Caucasian
  660 PE Microarray, QPCR Endometrial biopsy 46 Caucasian
  ST-007 PE Microarray, QPCR Endometrial biopsy 41 Unknown
  ST-012 PE Microarray, QPCR, Endometrial biopsy 41 Unknown
  ST-042 PE Microarray Endometrial biopsy 34 Caucasian
  ST-071 PE Microarray Endometrial biopsy 42 Caucasian
  ST-082 PE Microarray, IHC Endometrial biopsy 32 Caucasian
  ST-50 PE Microarray, QPCR Endometrial biopsy 39 Caucasian
  ST-080 ESE Microarray, QPCR, IHC Endometrial biopsy 43 Unknown
  ST-089 ESE Microarray, QPCR, IHC Endometrial biopsy 33 Caucasian
  ST-113 ESE Microarray, QPCR, IHC Endometrial biopsy 27 Caucasian
  550 MSE Microarray Endometrial biopsy 38 Mixed
  ST-009 MSE Microarray, QPCR, IHC Endometrial biopsy 31 Caucasian
  ST-014 MSE Microarray, QPCR, IHC Endometrial biopsy 28 Black
  ST-033 MSE Microarray, QPCR Endometrial biopsy 42 Caucasian
  ST-038 MSE Microarray, QPCR Endometrial biopsy 36 Caucasian
  ST-121 MSE Microarray, QPCR, IHC Endometrial biopsy 25 Caucasian
 Severe endometriosis
  26Aa PE Microarray Endometrial biopsy 31 Caucasian
  508a PE Microarray, QPCR Endometrial biopsy 25 Caucasian
  511 PE Microarray Endometrial biopsy 42 Caucasian
  575a PE Microarray, IHC Endometrial biopsy 26 Unknown
  587a PE Microarray, QPCR Endometrial biopsy 37 Caucasian
  589 PE Microarray, IHC Hysterectomy 48 Asian
  594a PE Microarray Endometrial biopsy 38 Caucasian
  595 PE Microarray Endometrial biopsy 37 Asian
  647a PE Microarray, QPCR Endometrial biopsy 39 Caucasian
  651a PE Microarray, QPCR Endometrial biopsy 37 Caucasian
  ST-049 PE Microarray Endometrial biopsy 29 Caucasian
  ST-076 PE Microarray Endometrial biopsy 30 Hispanic
  ST-084 PE Microarray, QPCR, IHC Endometrial biopsy 37 Caucasian
  ST-090 PE Microarray, QPCR, IHC Endometrial biopsy 42 Caucasian
  ST-70 PE Microarray Endometrial biopsy 22 Caucasian
  489a ESE Microarray, IHC Hysterectomy 39 Asian
  496a ESE Microarray, QPCR, IHC Endometrial biopsy 37 Caucasian
  517a ESE Microarray Endometrial biopsy 35 Asian
  599a ESE Microarray, IHC Endometrial biopsy 35 Black
  607 ESE Microarray, IHC Endometrial biopsy 24 Asian
  684 ESE Microarray, QPCR, IHC Hysterectomy 36 Caucasian
  27Aa ESE Microarray Endometrial biopsy 22 Caucasian
  ST-036 ESE Microarray Endometrial biopsy 45 Caucasian
  ST-065 ESE Microarray, QPCR Endometrial biopsy 34 Asian
  ST-112 ESE Microarray, QPCR, IHC Endometrial biopsy 38 Caucasian
  ST-127 ESE Microarray, IHC Endometrial biopsy 43 Caucasian
  ST-130 ESE Microarray, QPCR, IHC Endometrial biopsy 35 Caucasian
  516a MSE Microarray Endometrial biopsy 34 Asian
  526 MSE Microarray, QPCR, IHC Hysterectomy 48 Unknown
  540a MSE Microarray Endometrial biopsy 37 Caucasian
  543a MSE Microarray Endometrial biopsy 38 Caucasian
  544 MSE Microarray, QPCR, IHC Endometrial biopsy 46 Caucasian
  645a MSE Microarray Endometrial biopsy 39 Asian Indian
  678a MSE Microarray Hysterectomy 44 Asian
  33Aa MSE Microarray Endometrial biopsy 27 Caucasian
  72Aa MSE Microarray Endometrial biopsy 31 Caucasian
  73Aa MSE Microarray Endometrial biopsy 26 Caucasian
  97Aa MSE Microarray Endometrial biopsy 35 Unknown
  ST-037 MSE Microarray, QPCR, IHC Hysterectomy 32 Caucasian
  ST-039 MSE Microarray, QPCR Endometrial biopsy 44 Caucasian
  ST-078 MSE Microarray Endometrial biopsy 32 Caucasian
  ST-091 MSE Microarray, QPCR Endometrial biopsy 20 Caucasian
  ST-096 MSE Microarray, QPCR, IHC Endometrial biopsy 31 Caucasian
  ST-119 MSE Microarray, IHC Endometrial biopsy 41 Asian
 No endometriosis
  455b PE IHC Hysterectomy 39 Caucasian
  469 PE IHC Hysterectomy 42 Caucasian
  604 PE IHC Hysterectomy 44 Caucasian
  693 PE IHC Hysterectomy 46 Caucasian
  UC-24 ESE IHC Hysterectomy 45 Black
  UC-26 ESE IHC Hysterectomy 34 Caucasian
  629 ESE IHC Hysterectomy 46 Caucasian
  680 ESE IHC Hysterectomy 34 Caucasian
  463 MSE IHC Hysterectomy 48 Caucasian
  501 MSE IHC Hysterectomy 49 Caucasian
  610b MSE IHC Hysterectomy 50 Caucasian
  626b MSE IHC Hysterectomy 42 Caucasian

Abbreviations: QPCR, quantitative real-time reverse transcriptase−polymerase chain reaction; IHC, immunohistochemistry; PE, proliferative endometrium; ESE, early secretory endometrium; MSE, mid-secretory endometrium.

a Samples used in Burney et al, 2007.13

b Samples used in Talbi et al, 2006.16

Isolation of RNA and Preparation for Hybridization

Each endometrial tissue specimen was processed individually for microarray hybridization, as described earlier.13 Briefly, total RNA was extracted from whole-tissue specimens using the Trizol reagent (Invitrogen, Carlsbad, California), subjected to DNase treatment, and purified using the RNeasy Plus Kit (QIAGEN, Valencia, California). RNA purity was assessed by the A260/A280 ratio, and quality and integrity were assessed using the Agilent Bioanalyzer 2100 (Agilent Technologies, Santa Clara, California), with all samples having high-quality RNA (RNA Integrity Number (RIN) = 9.7-10).

RNA samples were prepared for microarray analysis according to the Affymetrix protocol (Affymetrix, Inc, Santa Clara, California), as described earlier.13,16 Briefly, for each sample, 5 µg of total RNA were reverse transcribed to complementary DNA (cDNA). Second strand DNA was generated using DNA polymerase, followed by overnight in vitro transcription to generate cRNA. After chemical fragmentation, biotinylated cRNAs were ready for hybridization. Quality of the final product was assessed in the Agilent Bioanalyzer. Each sample was hybridized to HU133 Plus 2.0 high-density oligonucleotide array (Affymetrix), with 54 600 genes and expressed sequence tags (ESTs), at the UCSF Genomic Core Facility. The data were scanned according to the protocol described in Assay Manual from Affymetrix.

Microarray Data Analysis

The .cel data files were imported into GeneSpring GX 10.0 software (Agilent Technologies) and processed using the robust multiarray analysis (RMA) algorithm for background adjustment, normalization, and log2-transformation of perfect match (PM) values.16 The data during each menstrual cycle phase (PE, ESE, and MSE were compared between the severe and mild endometriosis groups. The generated gene lists included only genes with >2.0-fold change (FC) and P < .05 by 1-way analysis of variance (ANOVA) with Tukey post hoc test and Benjamini-Hochberg multiple testing correction for false discovery rate.

Principal Component Analysis and Hierarchical Clustering

Principal component analysis (PCA) and hierarchical clustering were performed as described.15,16 Principal component analysis is an unbiased analysis performed in GeneSpring with all samples, using all 42 203 genes and 12 397 ESTs on Affymetrix Human HU133 Plus 2.0 arrays to look for similar expression patterns and underlying cluster structures. Hierarchical cluster analysis of differentially expressed genes from all samples was conducted using the smooth correlation distance measure algorithm (GeneSpring) to identify samples with similar patterns of gene expression. Compared to PCA, hierarchical clustering uses only informational genes—that is are differentially expressed among all experimental conditions.

Ingenuity Pathway Analysis

Gene symbols and FCs of the up- and downregulated genes in each pairwise comparison were imported into Ingenuity Pathway Analysis (IPA, Ingenuity Systems, Redwood City, California), as described earlier.15 For each comparison, associated top significantly regulated molecular and biological networks and canonical molecular pathways were identified. Only networks with the highest score were selected for the analysis. This was followed by functional analysis on the data set level and canonical pathway analysis. The significance of the association between the genes from the data set and the canonical pathway (in the IPA library) was presented as a ratio of the number of genes from the data set in a given pathway divided by the total number of molecules that make up the canonical pathway (Fisher exact test was used to calculate a P value). Pathways with P < .05 and ratio >0.05 were considered significant.

Microarray Validation by Real-Time Reverse Transcriptase−Polymerase Chain Reaction

Real-time reverse transcriptase−polymerase chain reaction (RT-PCR) was performed in duplicate using the SYBR Green PCR Mix (Fermentas Inc, Glen Burnie, Maryland), according to the manufacturer’s instructions. The housekeeping gene RPL19 was used as the normalizer. Numbers of mild endometriosis samples used for validation were n = 5, n = 3, and n = 5 for PE, ESE, and MSE, respectively, and in the severe endometriosis group, n = 6, n = 5, and n = 6 for PE, ESE, and MSE, respectively (Table 1 ). The following primer sequences were used: thyroxine deiodinase 2 (DIO2) sense 5′- TTGTACTTACTCTAAATTTCCCAAGG-3′ and antisense 5′-CATTGCCACTGTTGT CACCT-3′; insulin-like growth factor binding protein 5 (IGFBP5) sense 5′-TGCACCTGAGATGAGACAGG-3′ and antisense 5′-GCTTCATCCCGTACTTGTCC-3′; somatostatin (SST) sense 5′-CCCAGACTCCGTCAGTTTCT-3′ and antisense 5′-ATCATTCTCCGTCTGGTTGG-3′; transgelin (TAGLN) sense 5′-TTAGCTTTCCCCAGACATGG-3′ and antisense 5′-CGGTAGTGCCCATCATTCTT-3′; versican (VCAN) sense 5′-CCAGCCCCCTGTTGTAGAAA-3′ and antisense 5′’-ATTGAATTGTCCTTT GCTGATG-3′; solute carrier family 1, member 1 (SLC1A1) sense 5′-AACACTGCCTGTCACCTTCC-3′ and antisense 5′-GCACTCAGCACAATCACCAT-3′; epidermal growth factor receptor (EGFR) sense 5′-GAATGCATTTGCCAAGTCCT-3′ and antisense 5′-CGTCTATGCTGTCCTCAGTCA-3′; and RPL19 sense 5′-GCAGAT AATGGGAGGAGCC-3′ and antisense 5′- GCCCATCTTTGATGAGCTTC-3′. Polymerase chain reactions were run on the Mx4000 and Mx3005 quantitative real-time reverse transcriptase−polymerase chain reaction (QPCR) Stratagene systems (Agilent Technologies), using thermal cycling conditions, as described.15,18 Statistical analysis for the QRT-PCR results was performed using the nonparametric Mann-Whitney test. Significance was determined at P ≤ .05.

Immunohistochemistry

Immunostaining was performed for VCAN and EGFR using 4 µm thick paraffin-embedded endometrial tissue sections from women with mild and severe endometriosis: PE, n = 4 and n = 5, respectively; ESE, n = 3 and n = 7, respectively; and MSE, n = 3 and n = 5, respectively), as well as women without endometriosis (n = 4 in all phases). The samples were de-paraffinized in Xylene (Sigma-Aldrich, St Louis, Missouri) and rehydrated in decreasing concentrations of ethanol. All slides were incubated for 15 minutes in H2O2 (3% in methanol) to block endogenous peroxidase activity after antigen retrieval by boiling slides in citrate buffer (pH = 6.0). Thereafter the slides were blocked with normal horse serum for 45 minutes, followed by incubation with the primary antibody: overnight 4°C incubation with the rabbit polyclonal anti-VCAN antibody (Versican V0/V2 Neo, ThermoScientific, Waltham, Massachusetts) at 5 µg/mL concentration, and 1 hour at room temperature for the rabbit anti-human EGFR antibody (Santa Cruz Biotechnology, Inc, Santa Cruz, California, kind gift from Dr M Hsieh, UCSF) at 1:50 dilution.

In negative control slides, the primary antibody was replaced with nonimmune immunoglobulin G (IgG) of equivalent concentration from the same species. All slides were incubated with universal goat anti-rabbit/mouse secondary antibodies (Vector Laboratories Inc, Burlingame, California) for 30 minutes at room temperature. A freshly prepared diaminobenzidine-hydrogen peroxide solution (ImmPACT DAB kit, Vector Laboratories) was added to the slides, which were thereafter rinsed with distilled water. The slides were counterstained with haematoxylin (Vector Laboratories) and mounted with Clarion mounting medium (SigmaAldrich). A Leica microscope was used to visualize the immunostaining and to photograph the results. Sections of mouse ovarian and lung tissue (a kind gift from Dr Marco Conti, UCSF) were used as positive controls for VCAN immunostaining.19,20 Sections of 12-week human placental tissue served as a positive control for EGFR staining21; myometrium served as an internal positive control.22,23

Results

Cluster Analysis

Principal component analysis of all genes showed that mild and severe endometriosis samples cluster according to their cycle phase rather than the disease stage (Figure 1A), confirming previous observations of phase-dependent segregation when analyzing endometrial tissue or isolated cells.13,15 However, PCA followed by subsequent analysis of disease stage demonstrated that severe endometriosis samples cluster separately from mild endometriosis, regardless of cycle phase, although there was some overlap (Figure 1B).

Figure 1.

Figure 1.

Clustering analyses of samples from participants with mild and severe endometriosis. Panels A and B, Principal component analysis (PCA) of samples. A, Analyzed by menstrual cycle phase; and B, Analyzed by disease stage. PCA was applied to all endometrial samples that were characterized by the gene expression of all probes on the Affymetrix Gene 1.0 ST platform. C, Hierarchical clustering analysis of no-endometriosis and mild and severe endometriosis samples throughout the menstrual cycle, using the profiles of significantly regulated genes. PE indicates proliferative phase endometrium; ESE, early secretory phase endometrium; MSE, mid-secretory phase endometrium; m, mild endometriosis; s, severe endometriosis.

Unsupervised hierarchical clustering analysis was conducted using the profiles of significantly regulated genes in each study group (Figure 1C, clusterogram). Severe endometriosis samples clustered together and separately from mild endometriosis samples. Early secretory endometriosis from the mild endometriosis group clustered close to the mild PE group. Remarkably, even though clustering analysis of mild and severe endometriosis samples showed that they clustered separately from each other, signifying the difference between these 2 stages of endometriosis, PE as well as ESE and MSE samples from all groups demonstrated branching from the same stem, supporting the conclusion that cycle phase has greater impact than disease stage in sample clustering (Figure 1C).

Endometrial Transcriptome

Severe versus mild endometriosis

Comparison of severe versus mild endometriosis samples in the proliferative phase revealed 380 differentially regulated genes (P < .05, FC =2; Supplement Table 1), with 120 up- and 260 downregulated. Transcripts for several extracellular matrix (ECM) proteins and their receptors, such as VCAN, laminin-β1, fibrillin 1, and integrin-β1 (fibronectin receptor), were upregulated in severe endometriosis PE samples, as were heat shock proteins, DIO2 (the enzyme that converts thyroxine T4 to triiodothyronine T3), thioredoxin interacting protein (TXNIP), relaxin/insulin-like family peptide receptor 1, EGFR, microRNA 21 (MIR21), interferon-γ receptor 1, neuropilin, and others (Supplement Table 1).

Comparison of severe versus mild endometriosis samples in the early secretory phase revealed 817 differentially regulated genes (166 up- and 651 downregulated; Supplement Table 2 ). Although dysregulation of some genes persisted from the proliferative phase, some new genes were revealed, including upregulation of CYP26A1, IGF1, DICER1, DUSP1, KLF9, PAPPA, FOXO1A, neurotrophic tyrosine kinase receptor type 3, transducer of ERBB2 (TOB), and sulfatase 2 and downregulation of thyrotropin-releasing hormone (TRH), SST, lactotransferrin (LTF), TAGLN, Indian hedgehog homolog (IHH), BMP7, CXCL14, and others (Supplement Table 2). Some of the upregulated genes are progesterone and/or estradiol dependent, although some known progesterone-regulated genes (eg, IGFBP6, secretoglobin family 3A1, complement D, and glutathioine peroxidase 3 [GPX3]) were downregulated. These data suggest that the steroid hormone response and intracellular programs are disordered in both severe and mild forms of endometriosis in the early secretory phase.

Table 2.

The Most Represented Gene Ontology (GO) Categories in Severe Endometriosis

GO Biological Process GO Cellular Component GO Molecular Function
PE vs mild endometriosis PE
 Transcription Nucleus Nucleotide binding
 Transport Intracellular Protein binding
 Cell adhesion Cytoplasm DNA binding
 Nuclear mRNA splicing, via spliceosome Extracellular region Receptor activity
 Proteolysis Membrane Nucleic acid binding
 Translation Mitochondrion Catalytic activity
 Signal transduction Plasma membrane Actin binding
 Negative regulation of transcription from RNA polymerase II promoter Membrane fraction Binding
 Lipid metabolic process Integral to plasma membrane Structural constituent of ribosome
 Protein folding Golgi membrane Signal transducer activity
 Regulation of transcription, DNA-dependent Endoplasmic reticulum Structural molecule activity
 Skeletal system development Integral to membrane Zinc ion binding
 Mesoderm formation Golgi apparatus Magnesium ion binding
 Cell cycle Ruffle Receptor binding
 Protein amino acid phosphorylation Cytosol Insulin receptor binding
 Ubiquitin-dependent protein catabolic process Cytoskeleton RNA binding
 Carbohydrate metabolic process Extracellular space Transporter activity
 Mitochondrial electron transport, NADH to ubiquinone Filopodium NADH dehydrogenase activity
 Multicellular organismal development Mitochondrial inner membrane Hydrolase activity
 RNA processing Eukaryotic translation initiation factor 4F complex Ion channel activity
 mRNA processing Nucleolus Calcium ion binding
 Organ morphogenesis Cornified envelope Metalloendopeptidase activity
 Cell fate determination Endosome Transcription factor activity
 Angiogenesis Inner acrosomal membrane Ubiquitin-protein ligase activity
 DNA repair Microfibril Iron ion binding
ESE vs mild endometriosis ESE
 Transport Nucleus Protein binding
 Transcription Extracellular region Nucleotide binding
 Cell adhesion Cytoplasm DNA binding
 Proteolysis Membrane Nucleic acid binding
 Cell cycle Intracellular Catalytic activity
 Signal transduction Mitochondrion Receptor activity
 Translation Plasma membrane Actin binding
 Lipid metabolic process Membrane fraction Signal transducer activity
 Skeletal system development Endoplasmic reticulum Structural molecule activity
 Immune response Golgi membrane Transporter activity
 Nuclear mRNA splicing, via spliceosome Integral to plasma membrane Calcium ion binding
 Carbohydrate metabolic process Lysosome Cytokine activity
 Apoptosis Extracellular space RNA binding
 Ubiquitin-dependent protein catabolic process Cytosol Ion channel activity
 Protein amino acid phosphorylation Golgi apparatus Magnesium ion binding
 Metabolic process Soluble fraction Binding
 Protein folding Ubiquitin ligase complex Structural constituent of ribosome
 Protein modification process Nucleosome Zinc ion binding
 Regulation of cell growth Endosome Receptor binding
 rRNA processing Chromatin Hydrolase activity
 tRNA processing Ruffle Endopeptidase inhibitor activity
 Ossification Mediator complex Metalloendopeptidase activity
 DNA repair Cytoskeleton Ubiquitin-protein ligase activity
 Acute-phase response Integral to membrane of membrane fraction Phosphoprotein phosphatase activity
 Regulation of transcription, DNA-dependent Exosome (RNase complex) Antigen binding
MSE vs mild endometriosis MSE
 Transcription Nucleus Nucleotide binding
 Transport Cytoplasm Protein binding
 Signal transduction Extracellular region DNA binding
 Cell adhesion Intracellular Nucleic acid binding
 Translation Membrane Receptor activity
 Nuclear mRNA splicing, via spliceosome Mitochondrion Catalytic activity
 Proteolysis Plasma membrane Signal transducer activity
 Lipid metabolic process Endoplasmic reticulum Binding
 Protein amino acid phosphorylation Membrane fraction Actin binding
 Ubiquitin-dependent protein catabolic process Golgi membrane Transporter activity
 Negative regulation of transcription from RNA polymerase II promoter Integral to plasma membrane RNA binding
 Cell cycle Lysosome Calcium ion binding
 Multicellular organismal development Golgi apparatus Structural molecule activity
 DNA repair Cytosol Magnesium ion binding
 Angiogenesis Soluble fraction Structural constituent of ribosome
 Immune response Integral to membrane Zinc ion binding
 Skeletal system development Ubiquitin ligase complex Ion channel activity
 Protein folding Proteasome complex Ubiquitin-protein ligase activity
 Apoptosis Nucleosome Iron ion binding
 Carbohydrate metabolic process Ruffle Endopeptidase inhibitor activity
 Metabolic process Endosome Phosphoprotein phosphatase activity
 Regulation of cell growth Chromatin Receptor binding
 Protein modification process Telomeric region Iron ion transmembrane transporter activity
 Regulation of transcription, DNA-dependent Mitochondrion Enzyme inhibitor activity
 mRNA processing Coated pit hydrolase activity

Abbreviations: PE, proliferative endometrium; ESE, early secretory endometrium; MSE, mid-secretory endometrium.

Comparison of severe versus mild endometriosis samples in the mid-secretory phase revealed 1286 differentially regulated genes (Supplement Table 3 ), with 377 and 909genes being up- and downregulated, respectively. These data are consistent with the hierarchical clusterogram and indicate that the greatest differences between severe and mild endometriosis occur in the window of implantation (Figure 1C). Interestingly, some progesterone-regulated genes such as DKK1, MAOA, MAOB, CXCL14, IL15, IL1R1, IDO1, and CD55 were upregulated in this comparison group, although other progesterone-regulated genes, for example, KLF-13, IGFBP6, and members of the Notch-signaling pathway were downregulated (Supplemental Table 3). These data are consistent with dysregulation in the response to progesterone in MSE in both forms of endometriosis, as observed in ESE.

Table 3.

Top Networks Regulated in Endometriuum from Severe Versus Mild Endometriosis

Top 5 networks regulated in proliferative phase endometrium (PE) from severe endometriosis versus mild PE
ID Molecules in Network Score Focus Molecules Top Functions
1 ADAMTS9, Caspase, CDKN2A, Cyclin A, DBP, E2f, EN2, ERCC1, FOXL2, GAS2L1, GNA11, GSPT1, GTF2H4, GTSE1, HNRNPA2B1, HNRNPH1, Ifn gamma, IFNGR1, IL32, KRT17, LIN37, MAFF, MAP3K7IP2, NFkB (complex), NLRP1, PYCARD, RBCK1, RRAS, RRM1 (includes EG:6240), SUZ12, TANK, TFDP1, TFIIH, UHMK1, WTAP 52 29 Cell Death, Hair and Skin Development and Function, Organ Development
2 ANXA1, BAX, C12ORF10, Calpain, CBLC, DDX3X, DNAJA1, EGFR, FANCA, Fibrinogen, GNRH, HSP, Hsp70, Hsp90, HSP90AA1, HSP90AB1, HSP90B1, HSPA8, HSPH1, IFN Beta, IL1, LAMB1, LARP1, LRRFIP1, MACF1, MYOF, PAFAH1B3, PCM1, PI3K, PLA2, Proteasome, REEP6, SFRP1, SSB, YWHAZ 39 24 Cellular Compromise, Post-Translational Modification, Protein Folding
3 14-3-3, ADD3, Akt, ASAH1, BAD, CCNL1, Cdc2, CEL, CP, Ctbp, CTBP1, Cyclin E, DCN, DDX42, EXOSC4, FZD2, GADD45GIP1, HISTONE, HMG20B, HNRNPR, JDP2, MAP2K1/2, NFIC, p70 S6k, PDGF BB, PDPK1, PNN, PP2A, RBM5, RBMX, RPL13, SF3A2, SF3B1, SFRS1, SFRS7 39 25 RNA Post-Transcriptional Modification, Lipid Metabolism, Small Molecule Biochemistry
4 ATP2B4, C1QTNF2, CTTN, DIO2, EPHA2, ERK, FAK, FBN1, Fgf, Fgfr, FGFR3, FOSL2, GDI2, IL27RA, ITGB1, JAK, JAK1, KLF13, LMO4, NRTN, NUFIP1, Pak, Pdgf, Pdgfr, PI3K p85, PLC gamma, PSMD7, PTPN11, RAB1A, Raf, RLIM, Shc, STAT, STAT5a/b, VCAN 32 21 Cellular Development, Skeletal and Muscular System Development and Function, Cellular Movement
5
ARAP1, BRD4, Calmodulin, Ck2, CRIM1, CSDC2, GGA1, Histone h3, Ikb, IKK (complex), Insulin, MATR3, MIR21 (includes EG:406991), MRPL12, MSX2, MTUS1, MYCN, NOP2, NUDT1, PIK3R1, PSPC1, PTEN, RBM14, RNA polymerase II, RPL13, SFPQ, SLC39A4, SMC4, SORD, UBE2G2, UBE2I, Ubiquitin, XIST, ZNF146, ZNF451
27
19
Cell Cycle, Embryonic Development, Cancer
Top 5 networks regulated in early secretory phase endometrium (ESE) from severe endometriosis versus mild ESE
ID Molecules in Network Score Focus Molecules Top Functions
1 ADAMTS9, AP1S1, BCR, BEX2, BGN, C1q, C1QA, C1QB, C1S, CARD10, Complement component 1, CXCL16, ENPP1, FMOD, G0S2, IgG, IGKC, IGL@, Igm, IL32, KRT7, KRT13, KRT17, NFkB (complex), NFKBIZ, PIGR, PTPLAD1, RBCK1, SERINC3, SERPING1, SLC16A1, SLC3A1, SLC7A1, STAP2, TNFRSF18 42 29 Skeletal and Muscular System Development and Function, Amino Acid Metabolism, Dermatological Diseases
2 ALP, ALPP, ASS1, CCNO, CFD, DDX3X, DIO2, DKK1, DNMT3A, Fgf, FGF18, FOXL2, FOXS1, Frizzled, FRZB, FXYD5, FZD2, FZD8, FZD10, GAS1, HES1, MAFF, MIB2, MSX2, MUC4, NEDD4L, P38 MAPK, PDGF BB, PORCN, SLC30A5, SMAD6, SOX4, TOB1, Wnt, WNT4 42 29 Cell Development, Connective Tissue Development and Function, Skeletal and Muscular System Development
3 ADD3, AGTRAP, ATP1A2, BMP7, C1QTNF2, CCL5, CTSG, ERK, Fibrin, FXYD4, Growth hormone, Igf, Igfbp, IGFBP3, IGFBP5, IGFBP6, IL27RA, LCN2, Mmp, MMP7, Na+,K+ -ATPase, NADPH oxidase, NAMPT, NRTN, POSTN, PRPF4, RARRES2, SERPINA1, SERPINA3, Smad2/3-Smad4, STRA13, Tgf beta, TIMP1, VCAN, VitaminD3-VDR-RXR 30 24 Cellular Movement, Cancer, Gastrointestinal Disease
4 ABP1, Adaptor protein 2, ADRB2, ALDOA, Angiotensin II receptor type 1, Beta Arrestin, C12ORF10, CAV1, Clathrin, CMTM8, Creatine Kinase, DAB2, Dynamin, EGFR, FBP1, GFER, HDL, HSP90AB1, HSPA8, MACF1, MAT2A, Mek, MIR21 (includes EG:406991), NCK, NFIB, NUMA1, PLTP, PTPRS (includes EG:5802), SNX9 (includes EG:51429), Sos, SPDEF, TRAK1, TRAK2, TRH, Vegf 30 24 Cellular Assembly and Organization, Cardiac Hypertrophy, Cardiovascular Disease
5
ADCYAP1R1, AGR2, BLVRB, Cbp/p300, CYP26A1, DHRS13, FJX1, FOXO1, hCG, Histone h3, Histone h4, HMOX1, HOXB8, KLF6, LBH, LSR, MAF, MGMT, MTUS1, Nfat (family), NPTX2, NPTXR, Oxidoreductase, PDGFA, Pkc(s), PURA, Rxr, SFRP4, SP3, TAGLN, TBL1X, Thyroid hormone receptor, TNFRSF4, TSPO, XIST
30
26
Developmental Disorder, Genetic Disorder, Neurological Disease
Top 5 networks regulated in mid-secretory phase endometrium (MSE) from severe endometriosis versus mild MSE
ID Molecules in Network Score Focus Molecules Top Functions
1 AHNAK, ARID5B, B3GNT1, BGN, CAND2, CHI3L1, COL16A1, COL6A2, EHD2, ELN, FOXS1, FXYD6, H1FX, HNRPDL, KCNG1, KRT7, MFAP2, MPHOSPH9, MYOF, NPTX2, NPTXR, PDLIM4, PDZK1IP1, PKIG, PLOD2, PMEPA1, RAB25, RAMP2, RBPMS, SCG5, SPAG4, STARD10, TGFB1, WFS1, ZNF581 51 35 Cancer, Cell-To-Cell Signaling and Interaction, Skeletal and Muscular Disorders
2 ANXA11, APBA3, APEX2, AQP1, BIK, C12ORF10, CBLC, CCT2, DOCK5, EFEMP2, EGFR, FAM107A, GAS5, GPC1, HNRNPH1, HSP90AB1, HSPH1, LDOC1, LRRFIP1, MACF1, MBNL1, MPG, PAFAH1B3, PGK1, PHLDA1, PSMD7, PTPRS (includes EG:5802), RAB1A, RABAC1, SCAMP1, SFRP1, TRAK2, TUBB2A, WWP1, ZNF638 51 35 Cardiovascular System Development and Function, Cellular Development, Cell Growth and Proliferation
3 BCLAF1, C19ORF43, CAMK2N1, CCNL2, CHD1 (includes EG:1105), CLK1, DDX42, DEAF1, ERAL1, ERK, GNRH, HMG20B, HNRNPA2B1, HNRNPL, IL27RA, IL6ST, KLF13, KLHL22, LMO4, LOXL1, PRPF4, RBM5, RBMX, RLIM, SEMA3F, SF3A2, SF3B1, SFRS1, SFRS5, SFRS7, SFRS11, SFRS2IP, TRA2A, UBXN1, WSX1-gp130 42 32 RNA Post-Transcriptional Modification, Cancer, Cellular Growth and Proliferation
4 3 BETA HSD, AKR7A2, CCNL1, CNN1, CRIM1, CSRP2, DDX5, DUSP1, GLIS2, HELZ, HOXB8, HSD3B7, MAT2A, MEIS1, MIR21 (includes EG:406991), NAMPT, NBL1, NKIRAS2, PDGF BB, RNA polymerase II, RPL13, SERPINA3, SLC1A1, SP2, TAGLN, TEAD2, TEAD4, TIA1, TNNC1, TOB1, UNC5B, XAB2, XDH, YAP1, ZNF83 42 32 Cellular Assembly and Organization, Drug Metabolism, Genetic Disorder
5 ABCG1, ADAMTS9, AEBP1, ATF5, CARD10, CLIP2, CRISP3 (includes EG:10321), CXCL16, DBP, GFER, HDL, HES1, HEY2, LCN2, NCOA7, NFkB (complex), NFKBIZ, Notch, NOTCH2, NOTCH3, PLTP, PRDX2, PYCARD, RBCK1, RIOK3, RTKN, RTN4R, S100P, Secretase gamma, SLC3A1, SLC7A1, TAX1BP3, TCEA2, WTAP, ZMYND11 40 31 Amino Acid Metabolism, Molecular Transport, Small Molecule Biochemistry

Gene Ontology Categories in Severe Versus Mild Endometriosis Throughout the Menstrual Cycle

The most common gene ontology (GO) biological process groups in all comparisons were transcription, transport, cell adhesion, nuclear messenger RNA (mRNA) splicing, proteolysis, translation, and cell cycle, with angiogenesis and apoptosis processes having significant representation in the secretory (ESE and MSE) phase (Table 2). The main cellular components involved were nucleus, cytoplasm, extracellular and intracellular regions, and membranes (Table 2), demonstrating the ubiquitous participation of all cellular components in molecular differences between severe and mild endometriosis. The main GO molecular function categories included nucleotide binding, protein and DNA binding, receptor activity, actin binding, signal transducer activity, and others.

Microarray Validation by Real-Time RT-PCR

Some of the highly up- or downregulated genes, as well as genes dysregulated in all cycle phases between severe and mild endometriosis groups were selected randomly for validation using real-time RT-PCR: DIO2, IGFBP5), VCAN, SLC1A1, SST, TAGLN, and EGFR (Figure 2 ; Supplemental Tables 1-3). Most of the validated genes (VCAN, IGFBP5, SST, DIO2) follow the trend differences of the microarray results.

Figure 2.

Figure 2.

Quantitative real-time reverse transcriptase−polymerase chain reaction (QPCR) validation of microarray data. Panels A, VCAN; B, EGFR; C, SLC1A1; D, IGFBP5; E, SST; F, TAGLN; G, DIO2 gene expression in severe endometriosis expressed as fold change compared to the expression in mild endometriosis, throughout the menstrual cycle. Microarray data are presented in the insert. *Statistically significant differences (P < .05) between the same cycle phases in severe vs mild endometriosis determined by the (Mann-Whitney test). Error bars represent ± SEM. PE indicates proliferative phase endometrium; ESE, early secretory phase endometrium; MSE, mid-secretory phase endometrium; SEM, standard error of the mean; VCAN, versican; EGFR, epidermal growth factor receptor; SLC1A1, solute carrier family 1, member 1; IGFBP5, insulin-like growth factor binding protein 5; SST, somatostatin; TAGLN, transgelin; DIO2, thyroxine deiodinase 2.

Analysis of Networks and Canonical Pathways Regulated in Severe Versus Mild Endometriosis

Ingenuity pathway analysis of gene expression profiles revealed several associated network functions identified as different between severe versus mild endometriosis in proliferative, early secretory, and mid-secretory phase samples (Table 3). As expected, several genes are involved in more than 1 network/pathway. Comparison of canonical pathways regulated in severe versus mild endometriosis revealed large differences in eutopic endometrium in these 2 disease stages, as shown by the high number of regulated genes in the pathways. The major canonical pathways regulated are presented in Table 4 .

Table 4.

Top 50 Canonical Pathways

Ingenuity Canonical Pathways −log(P Value) P Value Ratio Molecules
Pathways regulated in proliferative endometrium (PE) from severe endometriosis vs mild PE
 Neuregulin signaling 6.25E00 .000001 0.11 ITGB1, BAD, PTPN11, HSP90AB1, RRAS, PIK3R1, DCN, HSP90AA1, PDPK1, PTEN, EGFR
 Prostate cancer signaling 4.83E00 .000015 0.09 BAD, TFDP1, PIK3C2A, HSP90AB1, RRAS, PIK3R1, HSP90AA1, PDPK1, PTEN
 Non-small cell lung cancer signaling 4.64E00 .000023 0.10 CDKN2A, BAD, TFDP1, PIK3C2A, RRAS, PIK3R1, PDPK1, EGFR
 PI3K/AKT signaling 4.35E00 .000045 0.07 ITGB1, JAK1, BAD, HSP90AB1, RRAS, PIK3R1, YWHAZ, HSP90AA1, PDPK1, PTEN
 Chronic myeloid leukemia signaling 4.27E00 .000054 0.09 CDKN2A, CTBP1, BAD, TFDP1, PTPN11, PIK3C2A, RRAS, PIK3R1, CBLC
 Myc-mediated apoptosis signaling 4.17E00 .000068 0.12 CDKN2A, BAD, PIK3C2A, RRAS, PIK3R1, YWHAZ, BAX
 Melanoma signaling 3.98E00 .000105 0.13 CDKN2A, BAD, PIK3C2A, RRAS, PIK3R1, PTEN
 IGF-1 signaling 3.69E00 .000204 0.08 BAD, PTPN11, PIK3C2A, RRAS, PIK3R1, YWHAZ, PDPK1, IGFBP5
 P70S6K signaling 3.51E00 .000309 0.07 GNAI2, JAK1, BAD, PIK3C2A, RRAS, PIK3R1, YWHAZ, PDPK1, EGFR
 Endometrial cancer signaling 3.45E00 .000355 0.11 BAD, PIK3C2A, RRAS, PIK3R1, PDPK1, PTEN
 Docosahexaenoic acid (DHA) signaling 3.39E00 .000407 0.11 BAD, PIK3C2A, PIK3R1, PDPK1, BAX
 FAK signaling 3.1E00 .000794 0.07 ITGB1, PIK3C2A, RRAS, PIK3R1, PDPK1, PTEN, EGFR
 PTEN signaling 2.89E00 .001288 0.07 ITGB1, BAD, RRAS, PIK3R1, PDPK1, PTEN, EGFR
 FLT3 signaling in hematopoietic progenitor cells 2.83E00 .001479 0.08 BAD, PTPN11, PIK3C2A, RRAS, PIK3R1, PDPK1
 Glioma signaling 2.75E00 .001778 0.06 CDKN2A, TFDP1, PIK3C2A, RRAS, PIK3R1, PTEN, EGFR
 CNTF signaling 2.75E00 .001778 0.09 JAK1, PTPN11, PIK3C2A, RRAS, PIK3R1
 Insulin receptor signaling 2.68E00 .002089 0.06 JAK1, BAD, PTPN11, PIK3C2A, RRAS, PIK3R1, PDPK1, PTEN
 Pancreatic adenocarcinoma signaling 2.6E00 .002512 0.06 CDKN2A, JAK1, BAD, TFDP1, PIK3C2A, PIK3R1, EGFR
 IL-2 signaling 2.56E00 .002754 0.09 JAK1, PTPN11, PIK3C2A, RRAS, PIK3R1
 Molecular mechanisms of cancer 2.49E00 .003236 0.04 CDKN2A, JAK1, PIK3C2A, BAD, TFDP1, RRAS, PIK3R1, MAP3K7IP2, GNA11, BAX, APH1A (includes EG:51107), GNAI2, PTPN11, FZD2
 FcγRIIB signaling in B lymphocytes 2.31E00 .004898 0.07 PIK3C2A, RRAS, PIK3R1, PDPK1
 JAK/Stat signaling 2.3E00 .005012 0.08 JAK1, PTPN11, PIK3C2A, RRAS, PIK3R1
 p53 signaling 2.29E00 .005129 0.07 CDKN2A, SCO2 (includes EG:9997), PIK3C2A, PIK3R1, BAX, PTEN
 Glucocorticoid receptor signaling 2.28E00 .005248 0.04 HSPA8, JAK1, PIK3C2A, HSP90AB1, RRAS, GTF2H4, PIK3R1, ANXA1, HSP90AA1, CCL5, UBE2I
 Angiopoietin signaling 2.27E00 .005370 0.07 BAD, PTPN11, PIK3C2A, RRAS, PIK3R1
 ERK5 signaling 2.24E00 .005754 0.07 BAD, PTPN11, RRAS, YWHAZ, EGFR
 Neurotrophin/TRK signaling 2.18E00 .006607 0.06 PTPN11, PIK3C2A, RRAS, PIK3R1, PDPK1
 IL-3 signaling 2.04E00 .009120 0.07 JAK1, BAD, PIK3C2A, RRAS, PIK3R1
 Agrin interactions at neuromuscular junction 2.04E00 .009120 0.07 ITGB1, RRAS, LAMB1, CTTN, EGFR
 EGF signaling 2.01E00 .009772 0.08 JAK1, PIK3C2A, PIK3R1, EGFR
 Aryl hydrocarbon receptor signaling 1.89E00 .012882 0.04 CDKN2A, NFIC, TFDP1, HSP90AB1, HSP90AA1, GSTT2, BAX
 14-3-3-mediated signaling 1.86E00 .013804 0.05 BAD, PIK3C2A, RRAS, PIK3R1, YWHAZ, BAX
 Aldosterone signaling in epithelial cells 1.83E00 .014791 0.05 HSPA8, PIK3C2A, PIK3R1, HSP90AA1, PDPK1
 EIF2 signaling 1.79E00 .016218 0.05 PIK3C2A, RRAS, PIK3R1, EIF4G3, PDPK1
 Thrombopoietin signaling 1.77E00 .016982 0.06 PTPN11, PIK3C2A, RRAS, PIK3R1
 Mitotic roles of polo-like kinase 1.77E00 .016982 0.06 ANAPC4, HSP90AB1, HSP90AA1, CDC16
 IL-9 signaling 1.73E00 .018621 0.08 JAK1, PIK3C2A, PIK3R1
 Bladder cancer signaling 1.69E00 .020417 0.05 CDKN2A, FGFR3, TFDP1, RRAS, EGFR
 Colorectal cancer metastasis signaling 1.68E00 .020893 0.04 JAK1, BAD, PIK3C2A, RRAS, PIK3R1, IFNGR1, BAX, FZD2, EGFR
 G beta gamma signaling 1.63E00 .023442 0.04 GNAI2, RRAS, GNA11, PDPK1, EGFR
 IL-15 signaling 1.6E00 .025119 0.06 JAK1, PIK3C2A, RRAS, PIK3R1
 Hypoxia signaling in the Cardiovascular system 1.58E00 .026303 0.06 HSP90AB1, HSP90AA1, PTEN, UBE2I
 GM-CSF signaling 1.58E00 .026303 0.06 PTPN11, PIK3C2A, RRAS, PIK3R1
 IL-4 signaling 1.53E00 .029512 0.06 JAK1, PIK3C2A, RRAS, PIK3R1
 Macropinocytosis signaling 1.53E00 .029512 0.06 ITGB1, PIK3C2A, RRAS, PIK3R1
 Erythropoietin signaling 1.51E00 .030903 0.05 PIK3C2A, RRAS, PIK3R1, PDPK1
 Fc Epsilon RI signaling 1.51E00 .030903 0.05 PTPN11, PIK3C2A, RRAS, PIK3R1, PDPK1
 Huntington disease signaling 1.48E00 .033113 0.03 HSPA8, PIK3C2A, PIK3R1, GNA11, STX16, PDPK1, BAX, EGFR
 HGF signaling 1.47E00 .033884 0.05 CDKN2A, PTPN11, PIK3C2A, RRAS, PIK3R1
 iCOS-iCOSL signaling in T Helper Cells 1.46E00 .034674 0.04 BAD, PIK3C2A, PIK3R1, PDPK1, PTEN
Pathways regulated in early secretory endometrium (ESE) from severe endometriosis vs mild ESE
 Complement system 3.54E00 .00029 0.19 CFD, SERPING1, C1S, CD55, C1QA, CD46, C1QB
 Hepatic fibrosis/hepatic stellate cell activation 2.92E00 .00120 0.10 VCAM1, FN1, PDGFA, IFNGR1, IGFBP5, BAX, CCL5, PGF, MYL9 (includes EG:10398), IGF1, TIMP1, IGFBP3, EGFR
 IL-8 signaling 2.72E00 .00191 0.08 NAPEPLD, VCAM1, PIK3C2A, RRAS, PIK3R1, BAX, IRAK1, PGF, EIF4EBP1, ROCK2, HMOX1, CCND2, RHOD, RHOF, EGFR
 Clathrin-mediated endocytosis signaling 2.71E00 .00195 0.08 ACTR2, PIK3C2A, PDGFA, PIK3R1, ITGB8, PGF, HSPA8, SNX9 (includes EG:51429), IGF1, FGF18, RAB5C, DAB2, CTTN, ITGB5
 Docosahexaenoic acid (DHA) signaling 2.67E00 .00214 0.13 BAD, FOXO1, PIK3C2A, PIK3R1, BIK, BAX
 IGF-1 signaling 2.65E00 .00224 0.10 IGFBP6, IGF1, BAD, FOXO1, PIK3C2A, RRAS, PIK3R1, IGFBP3, IGFBP5, SFN
 Prostate cancer signaling 2.41E00 .00389 0.09 BAD, TFDP1, FOXO1, PIK3C2A, HSP90AB1, RRAS, SRD5A1, PIK3R1, CREB3L4
 Myc-mediated apoptosis signaling 2.23E00 .00589 0.12 IGF1, BAD, PIK3C2A, RRAS, PIK3R1, BAX, SFN
 NRF2-mediated oxidative stress response 2.1E00 .00794 0.08 AKR7A2, MGST1, PIK3C2A, RRAS, PIK3R1, MAF, CLPP, MAFF, HMOX1, DNAJC4, GSTT2, GSTO2, FKBP5, PTPLAD1
 Ubiquinone biosynthesis 2.07E00 .00851 0.06 NDUFB11, NDUFS8, NDUFS7, MGMT, NDUFB7, NDUFA3, ALDH6A1
 IL-4 signaling 1.92E00 .01202 0.10 HLA-DQB1, SOCS1, HLA-DMA, JAK1, PIK3C2A, RRAS, PIK3R1
 VEGF signaling 1.81E00 .01549 0.08 ROCK2, BAD, FOXO1, PIK3C2A, RRAS, PIK3R1, SFN, PGF
 Pancreatic adenocarcinoma signaling 1.77E00 .01698 0.08 HMOX1, NAPEPLD, JAK1, BAD, TFDP1, PIK3C2A, PIK3R1, PGF, EGFR
 Virus entry via endocytic pathways 1.75E00 .01778 0.08 PIK3C2A, RRAS, PIK3R1, CD55, CAV1, ITGB8, ITGB5, FOLR1
 Caveolar-mediated endocytosis signaling 1.72E00 .01905 0.09 CD55, RAB5C, CAV1, COPE, ITGB8, ITGB5, EGFR
 Axonal guidance signaling 1.65E00 .02239 0.06 ACTR2, FZD10, PIK3C2A, UNC5A, RRAS, PDGFA, PIK3R1, GNA11, EFNA4, PGF, MYL9 (includes EG:10398), ROCK2, FZD8, IGF1, GLIS2, RHOD, NTRK3, WNT4, BMP7, EFNB3, RTN4R, FZD2, GLIS1
 Role of NANOG in mammalian embryonic stem cell pluripotency 1.62E00 .02399 0.09 FZD8, FZD10, JAK1, PIK3C2A, RRAS, PIK3R1, WNT4, BMP7, TCF7L1, FZD2
 Fructose and mannose metabolism 1.56E00 .02754 0.03 AKR7A2, GMDS, GALK1, FBP1, ALDOA
 Colorectal cancer metastasis signaling 1.55E00 .02818 0.06 FZD10, MMP7, JAK1, PIK3C2A, BAD, RRAS, PIK3R1, IFNGR1, BAX, PGF, FZD8, RHOD, WNT4, RHOF, FZD2, EGFR
 Interferon signaling 1.55E00 .02818 0.13 SOCS1, JAK1, IFITM1, IFNGR1
 Glucocorticoid receptor signaling 1.52E00 .03020 0.06 VCAM1, JAK1, PIK3C2A, RRAS, PIK3R1, CCL5, SLPI, HSPA8, HSP90AB1, DUSP1, GTF2H4, CDKN1C, FKBP5, POLR2I, UBE2I, ADRB2
 VDR/RXR activation 1.52E00 .03020 0.09 IGFBP6, FOXO1, PDGFA, IGFBP3, IGFBP5, HES1, CCL5
 ILK signaling 1.51E00 .03090 0.06 MYL9 (includes EG:10398), PARVB, FN1, PIK3C2A, RHOD, PIK3R1, CREB3L4, ITGB8, RHOF, PPP1R14B, ITGB5, PGF
 Basal cell carcinoma signaling 1.47E00 .03388 0.10 FZD8, FZD10, GLIS2, WNT4, BMP7, FZD2, GLIS1
 PI3K/AKT signaling 1.46E00 .03467 0.07 JAK1, BAD, FOXO1, HSP90AB1, RRAS, PIK3R1, SFN, EIF4EBP1, THEM4
 Human embryonic stem cell pluripotency 1.44E00 .03631 0.07 FZD8, FZD10, PIK3C2A, PDGFA, NTRK3, PIK3R1, SMAD6, WNT4, BMP7, FZD2
 Wnt/β-catenin signaling 1.44E00 .03631 0.07 SOX4, FZD8, SFRP4, FZD10, MMP7, FRZB, CDH3, WNT4, TCF7L1, PIN1, DKK1, FZD2
 Macropinocytosis signaling 1.42E00 .03802 0.08 PIK3C2A, RRAS, PDGFA, PIK3R1, ITGB8, ITGB5
 Non-small cell lung cancer signaling 1.39E00 .04074 0.08 BAD, TFDP1, PIK3C2A, RRAS, PIK3R1, EGFR
 Riboflavin metabolism 1.39E00 .04074 0.05 ACP5, ENPP3, ENPP1
 FLT3 signaling in hematopoietic progenitor cells 1.31E00 .04898 0.08 BAD, PIK3C2A, RRAS, PIK3R1, CREB3L4, EIF4EBP1
 Glutathione metabolism 1.29E00 .05129 0.05 GPX3, MGST1, GPX1, GSTT2, GSTO2
 Selenoamino acid metabolism 1.28E00 .05248 0.04 MGMT, PAPSS1 (includes EG:9061), MAT2A
 p53 signaling 1.27E00 .05370 0.08 SCO2 (includes EG:9997), CCND2, PIK3C2A, PIK3R1, RPRM, BAX, SFN
 IL-2 signaling 1.26E00 .05495 0.09 SOCS1, JAK1, PIK3C2A, RRAS, PIK3R1
 IL-3 signaling 1.26E00 .05495 0.08 JAK1, BAD, FOXO1, PIK3C2A, RRAS, PIK3R1
 Amyotrophic lateral sclerosis signaling 1.25E00 .05623 0.06 IGF1, PIK3C2A, PIK3R1, RAB5C, GPX1, BAX, PGF
 14-3-3-mediated signaling 1.24E00 .05754 0.07 BAD, FOXO1, PIK3C2A, RRAS, PIK3R1, BAX, SFN, PLCL1
 PDGF signaling 1.23E00 .05888 0.08 JAK1, PIK3C2A, RRAS, PDGFA, PIK3R1, CAV1
 Acute phase response signaling 1.22E00 .06026 0.06 HMOX1, SOCS1, SERPING1, FN1, RBP7, RRAS, C1S, PIK3R1, SERPINA3, SERPINA1, IRAK1
 Integrin signaling 1.21E00 .06166 0.06 ACTR2, PARVB, PIK3C2A, RHOD, RRAS, PIK3R1, CAV1, ITGB8, RHOF, TSPAN4, ITGB5, RAP2A
 Nitric oxide signaling in the cardiovascular system 1.21E00 .06166 0.07 PIK3C2A, HSP90AB1, PIK3R1, CAV1, SLC7A1, PGF
 Molecular mechanisms of cancer 1.21E00 .06166 0.05 FZD10, JAK1, PIK3C2A, BAD, TFDP1, RRAS, PIK3R1, GNA11, SMAD6, BAX, APH1A (includes EG:51107), FZD8, CCND2, FOXO1, RHOD, IHH, BMP7, RHOF, FZD2, RAP2A
 HMGB1 signaling 1.19E00 .06457 0.07 VCAM1, PIK3C2A, RHOD, RRAS, PIK3R1, IFNGR1, RHOF
 Nicotinate and nicotinamide metabolism 1.17E00 .06761 0.05 ENPP3, PRKX, ENPP1, QPRT, NAMPT, HIPK1, IRAK1
 Leukocyte extravasation signaling 1.14E00 .07244 0.06 ROCK2, CLDN10, MMP7, VCAM1, PIK3C2A, ICAM3, TIMP1, PIK3R1, RDX, MLLT4, CTTN
 Glioma signaling 1.11E00 .07762 0.06 IGF1, TFDP1, PIK3C2A, RRAS, PDGFA, PIK3R1, EGFR
 Methionine metabolism 1.1E00 .07943 0.04 DNMT3A, IL4I1, MAT2A
 mTOR signaling 1.1E00 .07943 0.06 HMOX1, NAPEPLD, PIK3C2A, RHOD, RRAS, PIK3R1, RHOF, PGF, EIF4EBP1
 Melanoma signaling 1.1E00 .07943 0.09 BAD, PIK3C2A, RRAS, PIK3R1
Pathways regulated in mid-secretory endometrium (MSE) from severe endometriosis vs mild MSE
 Neuregulin signaling 3.13E00 .00074 0.14 ITGB1, BAD, PIK3R1, DCN, PDPK1, PTEN, ERBB2IP (includes EG:55914), PRKCI, HSP90AB1, PTPN11, CDK5, HSP90AA1, PSEN1, EGFR
 Acute phase response signaling 2.63E00 .00234 0.11 IL6ST, SOCS1, MAP2K7, C1S, PIK3R1, PDPK1, CP, SERPINA3, MAP3K5 (includes EG:4217), IL1R1, TCF3, IRAK1, HMOX1, SOD2, RIPK1, PTPN11, C4BPA, MAP3K7, CRABP2, SERPINA1
 Docosahexaenoic acid (DHA) SIGNALING 2.37E00 .00427 0.16 BAD, FOXO1, PIK3C2A, PIK3R1, BIK, PDPK1, BAX
 Wnt/β-catenin signaling 2.34E00 .00457 0.12 SOX4, SOX7, FZD10, FRAT1, TCF7L1, TCF3, SOX17, CSNK1E, FZD8, TGFB1, MAP3K7, DKK3, CD44, PIN1, SOX18, SFRP1, DKK1, FZD2, FZD7
 Germ cell-sertoli cell junction signaling 2.14E00 .00724 0.11 EPN3, ITGB1, MAP2K7, PIK3C2A, CDC42, TJP1, PIK3R1, TUBG1, TUBB2A, PDPK1, MAP3K5 (includes EG:4217), IQGAP1, GSN, RHOQ, TGFB1, MAP3K7, PPAP2B, ACTN1
 Clathrin-mediated endocytosis signaling 2.11E00 .00776 0.10 ITGB1, ACTR2, PIK3C2A, RAB5A, CDC42, PDGFA, PIK3R1, CLTB, HSPA8, MET, ARRB1, FGF18, RAB5C, DAB2, TFRC, CTTN, ITGB5
 Prostate cancer signaling 2.09E00 .00813 0.12 BAD, TFDP1, FOXO1, PIK3C2A, HSP90AB1, SRD5A1, PIK3R1, HSP90AA1, PDPK1, CREB3L4, PTEN
 Mitochondrial dysfunction 1.94E00 .01148 0.09 NDUFS7, XDH, NDUFA13, APH1A (includes EG:51107), MAOB, NDUFB11, SOD2, NDUFS8, TXN2, NDUFB7, NDUFA3, CYC1, UQCRC1, PSEN1, MAOA
 Virus entry via endocytic pathways 1.89E00 .01288 0.12 ITGB1, PRKCI, CDC42, PIK3C2A, PIK3R1, CLTB, CD55, CAV1, TFRC, ITGB5, FOLR1
 Macropinocytosis signaling 1.86E00 .01380 0.13 ITGB1, MET, PRKCI, RAB5A, CDC42, PIK3C2A, PDGFA, PIK3R1, ITGB5
 Leukocyte extravasation signaling 1.83E00 .01479 0.09 ITGB1, PIK3C2A, CDC42, PIK3R1, RDX, THY1, ROCK2, GNAI2, CLDN23, PRKCI, PTPN11, ICAM3, EZR, CD44, MMP11, CTTN, ACTN1, TIMP2
 VDR/RXR activation 1.78E00 .01660 0.13 IGFBP6, SPP1, PRKCI, FOXO1, PDGFA, IGFBP5, NCOR2, HES1, CCL5, KLF4
 Notch signaling 1.77E00 .01698 0.16 RFNG, NOTCH2, NOTCH3, HEY2, HES1, APH1A (includes EG:51107), PSEN1
 NRF2-mediated oxidative stress response 1.5E00 .03162 0.09 AKR7A2, MAP2K7, PIK3C2A, PIK3R1, SLC35A2, DNAJC13, DNAJC3, MAP3K5 (includes EG:4217), DNAJA1, CLPP, MAFF, MAFG, HMOX1, PRKCI, SOD2, DNAJC4, MAP3K7
 Huntington disease signaling 1.43E00 .03715 0.08 MAP2K7, CAPN6, PIK3C2A, PIK3R1, GNA11, HSPA9, PDPK1, CREB3L4, BAX, GNG7, HDAC5, HSPA8, PRKCI, GNG11, CDK5, STX16, NCOR2, POLR2I, EGFR
 Hepatic fibrosis/hepatic stellate cell activation 1.42E00 .03802 0.10 PDGFA, FGFR1, FGFR2, IGFBP5, IL1R1, BAX, CCL5, MET, COL1A2, TGFB1, COL3A1, TIMP2, EGFR
 Insulin receptor signaling 1.42E00 .03802 0.09 JAK1, BAD, PIK3C2A, PIK3R1, PDPK1, VAMP2, PTEN, EIF4EBP1, PRKCI, RHOQ, FOXO1, PTPN11, PPP1R12A
 IGF-1 signaling 1.42E00 .03802 0.10 IGFBP6, PRKCI, BAD, FOXO1, PTPN11, PIK3C2A, PIK3R1, YWHAZ, PDPK1, IGFBP5
 Neurotrophin/TRK signaling 1.39E00 .04074 0.10 MAP2K7, CDC42, PTPN11, PIK3C2A, PIK3R1, PDPK1, CREB3L4, MAP3K5 (includes EG:4217)
 G beta gamma signaling 1.39E00 .04074 0.09 GNAI2, GNAS, GNG11, PRKCI, CDC42, GNA11, CAV1, PDPK1, GNG7, EGFR
 PI3K/AKT signaling 1.38E00 .04169 0.09 ITGB1, JAK1, BAD, FOXO1, HSP90AB1, PIK3R1, YWHAZ, HSP90AA1, PDPK1, MAP3K5 (includes EG:4217), PTEN, EIF4EBP1
 Semaphorin signaling in neurons 1.38E00 .04169 0.13 ROCK2, ITGB1, MET, RHOQ, CDK5, DPYSL4, NRP1
 Mitotic roles of polo-like kinase 1.38E00 .04169 0.11 ANAPC4, HSP90AB1, TGFB1, CDC7, HSP90AA1, CDC16, STAG2
 Histidine metabolism 1.37E00 .04266 0.05 PRPS2, ALDH3B2 (includes EG:222), MAOB, MGMT, ABP1, MAOA
 SAPK/JNK signaling 1.36E00 .04365 0.10 MAP2K7, GNG11, RIPK1, TRD@, CDC42, PIK3C2A, MAP3K7, PIK3R1, MAP3K5 (includes EG:4217), GNG7
 Type I diabetes mellitus signaling 1.34E00 .04571 0.10 SOCS1, HLA-DMA, MAP2K7, JAK1, RIPK1, TRD@, MAP3K7, IL1R1, MAP3K5 (includes EG:4217), HSPD1, IRAK1
 FGF signaling 1.3E00 .05012 0.10 MET, PTPN11, PIK3C2A, FGF18, PIK3R1, FGFR1, FGFR2, CREB3L4, MAP3K5 (includes EG:4217)
 Chronic myeloid leukemia signaling 1.25E00 .05623 0.10 CTBP1, RBL2, BAD, TFDP1, PTPN11, PIK3C2A, TGFB1, PIK3R1, CBLC, HDAC5
 RAR activation 1.25E00 .05623 0.08 CYP26A1, PIK3R1, NR2F2, PDPK1, MAP3K5 (includes EG:4217), PTEN, PRKCI, DUSP1, GTF2H4, TGFB1, CRABP2, RDH5, NCOR2, CARM1, SCAND1
 Phenylalanine metabolism 1.25E00 .05623 0.05 ALDH3B2 (includes EG:222), MAOB, ABP1, MAOA, PRDX2
 14-3-3-mediated signaling 1.24E00 .05754 0.10 PRKCI, BAD, FOXO1, PIK3C2A, PIK3R1, TUBB2A, TUBG1, YAP1, YWHAZ, BAX, MAP3K5 (includes EG:4217)
 Caveolar-mediated endocytosis signaling 1.23E00 .05888 0.10 ITGB1, RAB5A, CD55, RAB5C, CAV1, COPE, ITGB5, EGFR
 Ubiquinone biosynthesis 1.17E00 .06761 0.06 NDUFB11, NDUFS8, NDUFS7, MGMT, NDUFB7, NDUFA3, NDUFA13
 RAN signaling 1.17E00 .06761 0.13 KPNB1, RANBP2, RAN
 ILK signaling 1.11E00 .07762 0.08 MUC1, ITGB1, PIK3C2A, CDC42, PIK3R1, FERMT2, PDPK1, CREB3L4, PTEN, PARVB, RHOQ, PPAP2B, CHD1 (includes EG:1105), ACTN1, ITGB5
 Human embryonic stem cell pluripotency 1.07E00 .08511 0.08 FZD8, GNAS, FZD10, PIK3C2A, PDGFA, TGFB1, PIK3R1, FGFR1, FGFR2, PDPK1, FZD2, FZD7
 B Cell receptor signaling 1.06E00 .08710 0.08 MAP2K7, BAD, CDC42, PIK3C2A, PIK3R1, CREB3L4, MAP3K5 (includes EG:4217), PTEN, PTPRC, PTPN11, CARD10, MAP3K7, PAG1
 Ephrin receptor signaling 1.04E00 .09120 0.08 ITGB1, ACTR2, CDC42, PDGFA, GNA11, CREB3L4, GNG7, EFNA4, ROCK2, GNAI2, EPHB6, GNAS, GNG11, PTPN11, EFNB3
 Pentose phosphate pathway 9.86E-01 .10328 0.04 PRPS2, PGLS, FBP1, ALDOA
 Melanoma signaling 9.75E-01 .10593 0.11 BAD, PIK3C2A, PIK3R1, CHD1 (includes EG:1105), PTEN
 CCR5 signaling in macrophages 9.7E-01 .10715 0.08 GNAI2, GNAS, GNG11, PRKCI, TRD@, CCL5, GNG7
 FLT3 signaling in hematopoietic progenitor cells 9.45E-01 .11350 0.10 BAD, PTPN11, PIK3C2A, PIK3R1, PDPK1, CREB3L4, EIF4EBP1
 Oxidative phosphorylation 9.28E-01 .11803 0.08 ATP6V0E2, ATP6V0B, ATP5D, NDUFS7, TCIRG1, ATP6V1A, NDUFA13, NDUFB11, NDUFS8, NDUFB7, NDUFA3, CYC1, UQCRC1
 IL-8 signaling 9.27E-01 .11830 0.07 PIK3C2A, PIK3R1, BAX, GNG7, EIF4EBP1, IRAK1, ROCK2, GNAI2, HMOX1, GNAS, GNG11, PRKCI, RHOQ, EGFR
 Fructose and mannose metabolism 9.1E-01 .12303 0.04 AKR7A2, TSTA3, GMPPA, FBP1, ALDOA, FUK
 Cdc42 signaling 9E-01 .12589 0.08 ITGB1, ANAPC4, ACTR2, PRKCI, TRD@, CDC42, CDC42EP5, PPP1R12A, CDC16, IQGAP1
 Arginine and proline metabolism 8.96E-01 .12706 0.04 CKB, MAOB, VNN1, GAMT, LOXL1, ABP1, MAOA
 HGF signaling 8.69E-01 .13521 0.09 MET, MAP2K7, PRKCI, CDC42, PTPN11, PIK3C2A, MAP3K7, PIK3R1, MAP3K5 (includes EG:4217)
 EGF signaling 8.5E-01 .14125 0.10 MAP2K7, JAK1, PIK3C2A, PIK3R1, EGFR
 Nitric oxide signaling in the cardiovascular system 8.49E-01 .14158 0.08 PIK3C2A, HSP90AB1, PIK3R1, CAV1, HSP90AA1, SLC7A1, CACNA1A

Major differences in neuregulin signaling, which involves members of the EGFR family, were observed in the proliferative and mid-secretory phases between severe versus mild endometriosis (Figure 3 ). Epidermal growth factor receptor mRNA was upregulated in severe versus mild endometriosis in PE and ESE (Figure 2B), indicating its involvement in severe disease, and confirming our earlier reports of the involvement of EGF family in the pathophysiology of severe endometriosis.13,24

Figure 3.

Figure 3.

Neuregulin pathway regulation in proliferative endometrium (PE) and mid-secretory endometrium (MSE) from participants with severe vs mild endometriosis analyzed by ingenuity pathway analysis (IPA). Red color indicates upregulation of a gene; green color, down-regulation.

Epidermal Growth Factor Receptor Protein Immunoreactivity

As presented in Figure 4 and Table 5 , EGFR protein was expressed throughout the menstrual cycle in women with mild as well as severe endometriosis. Interestingly, the most dramatic difference in EGFR protein immunoreactivity was observed in the early secretory phase, where strong stromal expression was observed in severe compared to mild endometriosis, consistent with the real-time RT-PCR data (Figure 2B, Figure 4E,H, Table 5). There was a slight increase in epithelial EGFR immunostaining in the proliferative phase of severe endometriosis samples (Figure 4D,G, Table 5), whereas immunostaining was similar in MSE samples, regardless of the endometriosis stage (Figure 4F, I, Table 5). Immunohistochemical analysis of EGFR in endometrial samples from women without endometriosis throughout the menstrual cycle revealed weak expression of this protein in epithelial and/or stromal compartments (in particular, in ESE stroma), compared to the endometrium from women with endometriosis (Figure 4A-C, Table 5).

Figure 4.

Figure 4.

Epidermal growth factor receptor (EGFR) immunoreactivity in endometrial tissue from women without endometriosis (A-C), women with mild (D-F), and severe (G-I) endometriosis in the proliferative phase ([PE] n = 4, n = 4, and n = 5, respectively; A, D, G), early secretory phase ([ESE] n = 4, n = 3, and n = 7, respectively; B, E, H), and mid-secretory phase ([MSE] n = 4, n = 3, and n = 5, respectively; C, F, I) of the cycle. Human myometrial tissue was used as an internal positive control (on the same slide as endometrium, adjacent to the basalis endometrium, J). Human 12-week gestation placental tissue (K) was used as an additional positive control. L indicates negative control, nonimmune IgG-treated human endometrium; Le, luminal epithelium; ge, glandular epithelium; st, stroma; myo, myometrium; IgG, immunoglobulin G. Magnification ×200.

Table 5.

Semiquantitative Evaluation of VCAN and EGFR Immunostaining in Human Endometrial Tissue Sections From Women With Mild And Severe Endometriosis, As Well As Without Endometriosis

Protein PE Mild Endometriosis
ESE Mild Endometriosis
MSE Mild Endometriosis
PE Severe Endometriosis
ESE Severe Endometriosis
MSE Severe Endometriosis
Epithelium Stroma Epithelium Stroma Epithelium Stroma Epithelium Stroma Epithelium stroma Epithelium Stroma
EGFR + ++ + +/++ ++ ++ ++ ++ + +++ ++ ++
VCAN −/+ +++ ++ + ++ ++ +++ + ++ + +++ ++
PE no endometriosis
ESE no endometriosis
MSE no endometriosis

Epithelium
Stroma
Epithelium
Stroma
Epithelium
Stroma
EGFR ++ ++ + + ++ +
VCAN ++ + +/++ + ++ +

Abbreviations: PE, proliferative phase endometrium; ESE, early secretory phase endometrium; MSE, mid-secretory endometrium; VACN, versican; EGFR, epidermal growth factor receptor; −, no staining, −/+, a few stained cells; +, faint staining; ++, moderate staining; +++, strong staining.

Versican Protein Immunoreactivity

In the proliferative phase, there was a strong stromal and epithelial VCAN immunostaining in severe and mild endometriosis, respectively (Figure 5D,G , Table 5). In ESE, similar VCAN immunostaining was observed in the stroma and epithelial compartments (Figure 5E,H, Table 5), and epithelial VCAN immunostaining in MSE tended to be stronger in the severe endometriosis samples (Figure 5F,I, Table 5). Of note, diffuse stromal reactivity of VCAN in both cellular and extracellular compartments was observed. Remarkable was the VCAN immunoreactivity in the vasculature—in the smooth muscle layer and endothelial cells, regardless of disease status, stage, or cycle phase. Versican immunostaining in endometrial tissue from women without endometriosis throughout the menstrual cycle demonstrated overall weaker expression compared to samples from women with mild or severe endometriosis. This was particularly evident in PE epithelium and stroma in patients with severe and mild endometriosis, respectively, as well as MSE epithelium in severe endometriosis samples (Figure 5A-C, Table 5). Positive and negative controls demonstrated specificity of the observed results (Figure 5J-L).

Figure 5.

Figure 5.

Versican (VCAN) immunoreactivity in endometrial tissue from women without endometriosis (A-C), and women with mild (D-F), and severe (G-I) endometriosis in the proliferative ([PE] n = 4, n = 4, and n = 5, respectively; A, D, G), early secretory ([ESE]; n = 4, n = 3, and n = 7, respectively; B, E, H), and mid-secretory ([MSE]; n = 4, n = 3, and n = 5, respectively; C, F, I) phases of the menstrual cycle. Mouse lung tissue (J) and mouse ovary (K) were used as positive controls. L indicates negative control, nonimmune IgG-treated human endometrium; Le, luminal epithelium; ge, glandular epithelium; st, stroma; v, blood vessel. Magnification ×200.

Discussion

General Comments

The main finding of this study is the demonstrated difference in global gene expression in eutopic endometrium from participants with severe versus mild endometriosis, throughout the menstrual cycle. These 2 endometriosis stages are distinct in their clinical presentation, as well as therapeutic and surgical management, although the corresponding scientific literature is limited. The data herein underscore significant molecular and signaling pathway differences between these 2 stages of endometriosis in distinct hormonal milieu, suggesting that eutopic endometrium in severe versus mild endometriosis has different functional capacities.

Comparison of severe versus mild endometriosis samples by cycle phase revealed the dysregulation of several cyclic adenosine monophosphate (cAMP) and/or progesterone regulated gene, such as downregulation of IHH, SST, and TAGLN in ESE and upregulation of DKK1, MAO, IL15, and IL1R1 in MSE. Upregulation of DIO2 and downregulation of TRH transcripts between severe and mild endometriosis samples indicate potential involvement of thyroid hormone homeostasis and metabolism in the pathophysiology of this endometrial disorder.

Women with severe endometriosis experience higher rates of implantation failure during IVF treatment cycles. Of the 25 human receptivity-related genes identified by analysis of endometrial tissue from healthy fertile women,25 TAGLN and calponin 1 transcripts were dysregulated in MSE from women with severe versus mild endometriosis, suggesting their potential role in the impaired implantation process in women with severe disease.

Dysregulation of Neuregulin Signaling and EGFR in Severe Versus Mild Endometriosis

Of interest is the association of neuregulin signaling with endometriosis. Neuregulin signaling involves ligands for the transmembrane tyrosine kinase receptors ERBB1 (EGFR), ERBB2, ERBB3, and ERBB4—members of the EGFR family.26 Ligand binding activates intracellular signaling cascades and the induction of cellular responses including proliferation, migration, differentiation, and survival or apoptosis in different organs and systems.27 Neuregulin genes, though not regulated themselves in the current study, influence proliferation, migration, and differentiation of epithelial, neuronal, glial, cardiac, and other types of cells.2830 This canonical pathway was highly regulated herein in PE and MSE between severe and mild endometriosis samples. Neuregulin (also known as heregulin) signals through HER3 and HER4 receptors; although no changes were observed herein between disease stages or in different cycle phases.

Epidermal growth factor receptor is the major player of neuregulin-signaling pathway. Epidermal growth factor receptor (ERBB1) expression in normal eutopic endometrium on the mRNA and protein levels during different menstrual cycle phases was demonstrated herein and confirms earlier reports.3133 We have observed that EGFR gene expression is increased in eutopic endometrium of women with severe endometriosis compared to women without disease in ESE and MSE, but not PE, and is not regulated in mild endometriosis versus nonendometriosis samples throughout the cycle (Aghajanova et al, unpublished data). Herein, we have found that EGFR is dysregulated in severe versus mild endometriosis throughout the menstrual cycle on both mRNA and protein levels, with the most dramatic difference (upregulation) in ESE. Furthermore, transducer of ERBB2 was upregulated in ESE. Thus, the present study demonstrates differences in EGFR expression between different stages of endometriosis and also supports earlier studies noting involvement of EGF family members in the pathophysiology of endometriosis.13,25,34 Interestingly, EGFR is a tumor marker, particularly for epithelial tumors such as colon cancer, lung cancer, prostate cancer, breast cancer, or other solid tumors.26,35 Whether it is a marker of a severity of endometriosis remains to be determined.

Dysregulation of ECM Molecules in Severe Versus Mild Endometriosis

This is the first study to demonstrate mRNA expression and immunoreactivity of the ECM proteoglycan VCAN in human endometrium. In participants without endometriosis, VCAN immunoreactivity was weak, especially in PE; whereas, strong immunoreactivity was observed in endometrial stroma from women with severe endometriosis and in epithelium of samples from women with mild disease. Versican can bind to integrins on the cell surface,36 stimulating cell proliferation and inhibiting apoptosis.37 Versican has multiple functions and interactions in different model systems. For example, overexpression of VCAN in a pheochromocytoma cell line upregulates EGFR.38 Also, VCAN expression is increased in endothelial cells with increased migrating capacity.39 Thus, high levels of VCAN may promote an invasive phenotype of endometrial cells in endometriosis by affecting their proliferation, apoptosis, adhesion, and migration, and also may participate in or be causative of the upregulation of EGFR in endometriosis. These functions in endometriosis await further investigation.

Dysregulation of MicroRNAs in Severe Versus Mild Endometriosis

MicroRNA (miRNA) 21 (MIR21) was found to be upregulated on the array, herein, in eutopic endometrium throughout the menstrual cycle in severe versus mild endometriosis. It has recently been shown to be upregulated in eutopic endometrium of women with versus without endometriosis.40 Some of the predicted target genes for this miRNA are the tumor-suppressor gene PTEN (downregulated 2.18- and 2.3-fold in severe vs mild endometriosis in PE and MSE, respectively), PDCD4, E2F1, and TGFBRII.4143 Interestingly, downregulation of MIR21 inhibits expression of EGFR in human glioblastoma cells.44 Whether there is such a mechanism operating in human endometrial stromal fibroblasts (hESF) remains to be determined.

Herein, we observed the upregulation of DICER1 in ESE and MSE from severe versus mild endometriosis (Supplement Tables 2 and 3). The transcript for DICER1 (dicer1, ribonuclease type III), which is a repressor of gene expression due to its involvement in the biogenesis of microRNAs and small interfering RNAs, demonstrates cyclic variation throughout the normal human menstrual cycle.45 Female mice with a conditional knockout of Dicer1 in mesenchyme-derived cells of the oviducts and uterus are sterile, in part, due to uterine defects.46 Although endometrial stromal Dicer1 expression was absent, the decidualization process was not compromised,46 consistent with the recent finding that DICER1 knockdown in hESF does not affect decidualization.45 Increased expression of DICER1 in secretory endometrium from women with severe versus mild endometriosis may lead to downregulation of apoptosis-associated genes and dysregulation of adhesion molecules, leading to resistance to apoptosis and increased migratory functions in endometrial cells, as observed with endothelial cells.47

Dysregulation of Canonical Pathways in Severe Versus Mild Endometriosis

Several pathways regulated between severe and mild endometriosis are of interest. Severe endometriosis samples exhibited dysregulation of second-messenger signaling pathways, including PI3K/AKT, JAK/STAT, SPK/JNK, and MAPK, confirming recent reports.48 Regulation of neurotrophin/TRK (neurotrophic tyrosine kinase) signaling in PE and MSE and axonal signaling in ESE are consistent with the presence of nerve fibers in eutopic endometrium and perhaps their role in the pathogenesis of endometriosis-associated pain.49,50 However, other functions of these pathways (and their members) may be operating in endometrium, not related to pain. The current study demonstrates differences in these pathways between severe and mild stages of endometriosis, although pain and stage are not necessarily correlated.51,52

Of note is the involvement of cancer-associated pathways, such as prostate, endometrial, bladder, colorectal, pancreatic cancer, and basal cell carcinoma signaling, suggesting commonalities in the pathophysiology between severe endometriosis and epithelial cancers.

Wnt signaling, NRF2-mediated oxidative stress response signaling (nuclear factor [erythroid-derived 2]-like 2, involved in apoptosis and the oxidative stress response), and retinoid X receptor (RXR) signaling were significantly regulated in secretory endometrium (ESE and MSE; Supplememtal Table 2), probably indicating the differences in the endometrial response to progesterone between severe and mild endometriosis.

Summary

Taken together, these data demonstrate the complexity of the processes and gene interactions and pathways involved in the endometrium of women with endometriosis and the molecular differences in the setting of severe versus mild disease. Whether these differences account for the observed differences in clinical presentations of women with severe versus mild endometriosis, that is lower implantation and pregnancy rates in women with severe disease, remain to be determined. The signaling pathways identified may serve for development of targeted therapies to correct the phenotype at the endometrial level.

Footnotes

The authors declared no potential conflicts of interests with respect to the authorship and/or publication of this article.

The authors disclosed receipt of the following financial support for the research and/or authorship of this article: the Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD)/NIH through cooperative agreement U54HD055764-04 as part of the Specialized Cooperative Centers Program in Reproduction and Infertility Research.

References

  • 1. Giudice LC, Kao LC. Endometriosis. Lancet. 2004;364(9447):1789–1799 [DOI] [PubMed] [Google Scholar]
  • 2. Taylor RN, Yu J, Torres PB, et al. Mechanistic and therapeutic implications of angiogenesis in endometriosis. Reprod Sci. 2009;16(2):140–146 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. The American Fertility Society Revised American Fertility Society classification of endometriosis. Fertil Steril. 1985;43(3):351–352 [DOI] [PubMed] [Google Scholar]
  • 4. Rock JA. The revised American Fertility Society classification of endometriosis: reproducibility of scoring. ZOLADEX Endometriosis Study Group. Fertil Steril. 1995;63(5):1108–1110 [DOI] [PubMed] [Google Scholar]
  • 5. Nisolle M, Donnez J. Peritoneal endometriosis, ovarian endometriosis, and adenomyotic nodules of the rectovaginal septum are three different entities. Fertil Steril. 1997;68(4):585–596 [DOI] [PubMed] [Google Scholar]
  • 6. Donnez J, Nisolle M, Smoes P, Gillet N, Beguin S, Casanas-Roux F. Peritoneal endometriosis and “endometriotic” nodules of the rectovaginal septum are two different entities. Fertil Steril. 1996;66(3):362–368 [PubMed] [Google Scholar]
  • 7. Matsuzaki S, Maleysson E, Darcha C. Analysis of matrix metalloproteinase-7 expression in eutopic and ectopic endometrium samples from patients with different forms of endometriosis. Hum Reprod. 2010;25(3):742–750 [DOI] [PubMed] [Google Scholar]
  • 8. D’Hooghe TM, Debrock S, Hill JA, Meuleman C. Endometriosis and subfertility: is the relationship resolved?. Semin Reprod Med. 2003;21(2):243–254 [DOI] [PubMed] [Google Scholar]
  • 9. Kuivasaari P, Hippelainen M, Anttila M, Heinonen S. Effect of endometriosis on IVF/ICSI outcome: stage III/IV endometriosis worsens cumulative pregnancy and live-born rates. Hum Reprod. 2005;20(11):3130–3135 [DOI] [PubMed] [Google Scholar]
  • 10. Barnhart K, Dunsmoor-Su R, Coutifaris C. Effect of endometriosis on in vitro fertilization. Fertil Steril. 2002;77(5):1148–1155 [DOI] [PubMed] [Google Scholar]
  • 11. Matalliotakis IM, Cakmak H, Mahutte N, Fragouli Y, Arici A, Sakkas D. Women with advanced-stage endometriosis and previous surgery respond less well to gonadotropin stimulation, but have similar IVF implantation and delivery rates compared with women with tubal factor infertility. Fertil Steril. 2007;88(6):1568–1572 [DOI] [PubMed] [Google Scholar]
  • 12. Kao LC, Germeyer A, Tulac S, et al. Expression profiling of endometrium from women with endometriosis reveals candidate genes for disease-based implantation failure and infertility. Endocrinology. 2003;144(7):2870–2881 [DOI] [PubMed] [Google Scholar]
  • 13. Burney RO, Talbi S, Hamilton AE, et al. Gene expression analysis of endometrium reveals progesterone resistance and candidate susceptibility genes in women with endometriosis. Endocrinology. 2007;148(8):3814–3826 [DOI] [PubMed] [Google Scholar]
  • 14. Bulun SE. Endometriosis. N Engl J Med. 2009;360(3):268–279 [DOI] [PubMed] [Google Scholar]
  • 15. Aghajanova L, Horcajadas JA, Weeks JL, et al. The protein kinase A pathway-regulated transcriptome of endometrial stromal fibroblasts reveals compromised differentiation and persistent proliferative potential in endometriosis. Endocrinology. 2010;151(3):1341–1355 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Talbi S, Hamilton AE, Vo KC, et al. Molecular phenotyping of human endometrium distinguishes menstrual cycle phases and underlying biological processes in normo-ovulatory women. Endocrinology. 2006;147(3):1097–1121 [DOI] [PubMed] [Google Scholar]
  • 17. Noyes RW, Hertig AT, Rock J. Dating the endometrial biopsy. Fertil Steril. 1950;1:3–25 [DOI] [PubMed] [Google Scholar]
  • 18. Aghajanova L, Hamilton A, Kwintkiewicz J, Vo KC, Giudice LC. Steroidogenic enzyme and key decidualization marker dysregulation in endometrial stromal cells from women with versus without endometriosis. Biol Reprod. 2009;80(1):105–114 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Russell DL, Ochsner SA, Hsieh M, Mulders S, Richards JS. Hormone-regulated expression and localization of versican in the rodent ovary. Endocrinology. 2003;144(3):1020–1031 [DOI] [PubMed] [Google Scholar]
  • 20. Kaplan F, Comber J, Sladek R, et al. The growth factor midkine is modulated by both glucocorticoid and retinoid in fetal lung development. Am J Respir Cell Mol Biol. 2003;28(1):33–41 [DOI] [PubMed] [Google Scholar]
  • 21. Bulmer JN, Thrower S, Wells M. Expression of epidermal growth factor receptor and transferrin receptor by human trophoblast populations. Am J Reprod Immunol. 1989;21(3-4):87–93 [DOI] [PubMed] [Google Scholar]
  • 22. Smith K, LeJeune S, Harris AH, Rees MC. Epidermal growth factor receptor in human uterine tissues. Hum Reprod. 1991;6(9):619–622 [DOI] [PubMed] [Google Scholar]
  • 23. Heiner JS, Cai L, Ding H, Rutgers JK. Myometrial expression of mRNA encoding epidermal growth factor receptor (EGFR) throughout the menstrual cycle. Am J Reprod Immunol. 1994;32(3):152–156 [DOI] [PubMed] [Google Scholar]
  • 24. Velarde MC, Aghajanova L, Nezhat CR, Giudice LC. Increased mitogen-activated protein kinase kinase/extracellularly regulated kinase activity in human endometrial stromal fibroblasts of women with endometriosis reduces 3,’5’-cyclic adenosine 5’-monophosphate inhibition of cyclin D1. Endocrinology. 2009;150(10):4701–4712 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Horcajadas JA, Pellicer A, Simón C. Wide genomic analysis of human endometrial receptivity: new times, new opportunities. Hum Reprod Update. 2007;13(1):77–86 [DOI] [PubMed] [Google Scholar]
  • 26. Hynes NE, Lane HA. ERBB receptors and cancer: the complexity of targeted inhibitors. Nat Rev Cancer. 2005;5(5):341–354 [DOI] [PubMed] [Google Scholar]
  • 27. Earp HS, 3rd, Calvo BF, Sartor CI. The EGF receptor family—multiple roles in proliferation, differentiation, and neoplasia with an emphasis on HER4. Trans Am Clin Climatol Assoc. 2003;114:315–333 [PMC free article] [PubMed] [Google Scholar]
  • 28. Birchmeier C, Nave KA. Neuregulin-1, a key axonal signal that drives Schwann cell growth and differentiation. Glia. 2008;56(4):1491–1497 [DOI] [PubMed] [Google Scholar]
  • 29. Esper RM, Pankonin MS, Loeb JA. Neuregulins: versatile growth and differentiation factors in nervous system development and human disease. Brain Res Rev. 2006;51(2):161–175 [DOI] [PubMed] [Google Scholar]
  • 30. Xu Y, Li X, Zhou M. Neuregulin-1/ErbB signaling: a druggable target for treating heart failure. Curr Opin Pharmacol. 2009;9(2):214–219 [DOI] [PubMed] [Google Scholar]
  • 31. Imai T, Kurachi H, Adachi K, et al. Changes in epidermal growth factor receptor and the levels of its ligands during menstrual cycle in human endometrium. Biol Reprod. 1995;52(4):928–938 [DOI] [PubMed] [Google Scholar]
  • 32. Ejskjaer K, Sorensen BS, Poulsen SS, Mogensen O, Forman A, Nexo E. Expression of the epidermal growth factor system in human endometrium during the menstrual cycle. Mol Hum Reprod. 2005;11(8):543–551 [DOI] [PubMed] [Google Scholar]
  • 33. Aghajanova L, Bjuresten K, Altmae S, Landgren BM, Stavreus-Evers A. HB-EGF but not amphiregulin or their receptors HER1 and HER4 is altered in endometrium of women with unexplained infertility. Reprod Sci. 2008;15(5):484–492 [DOI] [PubMed] [Google Scholar]
  • 34. Ejskjaer K, Sorensen BS, Poulsen SS, Mogensen O, Forman A, Nexo E. Expression of the epidermal growth factor system in eutopic endometrium from women with endometriosis differs from that in endometrium from healthy women. Gynecol Obstet Invest. 2009;67(2):118–126 [DOI] [PubMed] [Google Scholar]
  • 35. Hynes NE, MacDonald G. ErbB receptors and signaling pathways in cancer. Curr Opin Cell Biol. 2009;21(2):177–184 [DOI] [PubMed] [Google Scholar]
  • 36. Wu YJ, La Pierre DP, Wu J, Yee AJ, Yang BB. The interaction of versican with its binding partners. Cell Res. 2005;15(7):483–494 [DOI] [PubMed] [Google Scholar]
  • 37. Sheng W, Wang G, Wang Y, et al. The roles of versican V1 and V2 isoforms in cell proliferation and apoptosis. Mol Biol Cell. 2005;16(3):1330–1340 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Wu Y, Chen L, Cao L, Sheng W, Yang BB. Overexpression of the C-terminal PG-M/versican domain impairs growth of tumor cells by intervening in the interaction between epidermal growth factor receptor and beta1-integrin. J Cell Sci. 2004;117(11):2227–2237 [DOI] [PubMed] [Google Scholar]
  • 39. Cattaruzza S, Schiappacassi M, Ljungberg-Rose A, et al. Distribution of PG-M/versican variants in human tissues and de novo expression of isoform V3 upon endothelial cell activation, migration, and neoangiogenesis in vitro. J Biol Chem. 2002;277(49):47626–47635 [DOI] [PubMed] [Google Scholar]
  • 40. Luo X, Prucha MS, Chegini N. The expression, regulation and function of miR-21 in the endometrium and endometriosis. Reprod Sci. 2010;17(3):349A [Google Scholar]
  • 41. Pan Q, Luo X, Chegini N. MicroRNA 21: response to hormonal therapies and regulatory function in leiomyoma, transformed leiomyoma and leiomyosarcoma cells. Mol Hum Reprod. 2010;16(3):215–227 [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 42. Zhang JG, Wang JJ, Zhao F, Liu Q, Jiang K, Yang GH. MicroRNA-21 (miR-21) represses tumor suppressor PTEN and promotes growth and invasion in non-small cell lung cancer (NSCLC). Clin Chim Acta. 2010;411(11-12):846–852 [DOI] [PubMed] [Google Scholar]
  • 43. Qi L, Bart J, Tan LP, et al. Expression of miR-21 and its targets (PTEN, PDCD4, TM1) in flat epithelial atypia of the breast in relation to ductal carcinoma in situ and invasive carcinoma. BMC Cancer. 2009;9:163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Zhou X, Ren Y, Moore L, et al. Downregulation of miR-21 inhibits EGFR pathway and suppresses the growth of human glioblastoma cells independent of PTEN status. Lab Invest. 2010;90(2):144–155 [DOI] [PubMed] [Google Scholar]
  • 45. Ran L, Arias P, Han D, Andreu-Vieyra C, Matzuk M, Hawkins S. Characterization of DICER gene expression in the human reproductive tract. Reprod Sci. 2010;17(2):137A [Google Scholar]
  • 46. Nagaraja AK, Andreu-Vieyra C, Franco HL, et al. Deletion of Dicer in somatic cells of the female reproductive tract causes sterility. Mol Endocrinol. 2008;22(10):2336–2352 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Asada S, Takahashi T, Isodono K, et al. Downregulation of Dicer expression by serum withdrawal sensitizes human endothelial cells to apoptosis. Am J Physiol Heart Circ Physiol. 2008;295(6):2512–2521 [DOI] [PubMed] [Google Scholar]
  • 48. Zhang H, Zhao X, Liu S, Li J, Wen Z, Li M. 17betaE2 promotes cell proliferation in endometriosis by decreasing PTEN via NFkappaB-dependent pathway. Mol Cell Endocrinol. 2010;317(1-2):31–43 [DOI] [PubMed] [Google Scholar]
  • 49. Tokushige N, Markham R, Russell P, Fraser IS. High density of small nerve fibres in the functional layer of the endometrium in women with endometriosis. Hum Reprod. 2006;21(3):782–787 [DOI] [PubMed] [Google Scholar]
  • 50. Tokushige N, Markham R, Russell P, Fraser IS. Different types of small nerve fibers in eutopic endometrium and myometrium in women with endometriosis. Fertil Steril. 2007;88(4):795–803 [DOI] [PubMed] [Google Scholar]
  • 51. Porpora MG, Koninckx PR, Piazze J, Natili M, Colagrande S, Cosmi EV. Correlation between endometriosis and pelvic pain. J Am Assoc Gynecol Laparosc. 1999;6(4):429–434 [DOI] [PubMed] [Google Scholar]
  • 52. Vercellini P, Fedele L, Aimi G, Pietropaolo G, Consonni D, Crosignani PG. Association between endometriosis stage, lesion type, patient characteristics and severity of pelvic pain symptoms: a multivariate analysis of over 1000 patients. Hum Reprod. 2007;22(1):266–271 [DOI] [PubMed] [Google Scholar]

Articles from Reproductive Sciences are provided here courtesy of Society for Reproductive Investigation

RESOURCES