Abstract
Objective:
To identify the transcriptomic changes of ectopic lesions and eutopic endometrial tissues during the progression of endometriosis.
Hypothesis:
Our hypothesis is that development and progression of endometriosis change transcriptome in the eutopic endometrium and ectopic lesions
Design:
Laboratory study.
Setting:
Academic Medical Center
Animals:
Four fertile and four subfertile Pgrcre/+Rosa26mTmG/+ mice with endometriosis, and four sham mice for each group of endometriosis mice as control. These mice underwent either surgery to induce endometriosis or sham surgery. Fertile sham and mice with endometriosis were used one month after surgery, while subfertile ones were used three months after surgery.
Interventions:
Early and chronic effect of endometriosis on transcriptomics of ectopic lesions and eutopic endometrium
Main Outcome Measures:
RNA-Sequencing analysis (RNA-Seq) and identification of differential expressed genes and pathways in the ectopic lesions and eutopic uteri from mice with endometriosis and sham mice at day of pregnancy 3.5.
Results:
Our mouse model recapitulates the transcriptomic changes of ectopic lesions in human. RNA-seq was performed in ectopic lesions and eutopic uteri from mice with or without endometriosis during progression of disease. Estradiol, inflammation, angiogenesis, and fibrosis pathways were consistently elevated in all the ectopic lesions compared to eutopic endometrium. Cholesterol/glucose synthesis and stem cell pluripotency pathways were more enhanced in ectopic lesions from subfertile mice compared to their eutopic endometrium. Dysregulation of infiltration of macrophage, dendritic, T and B cells were validated by using immunohistochemistry in ectopic lesions. multiple ligand-receptor pairs between the ectopic and eutopic endometrium were altered compared to the sham endometrium. Suppressed WNT and EGF pathways were only found in the eutopic endometrium from subfertile not fertile mice compared to sham.
Conclusions:
Our mouse endometriosis model recapitulates the transcriptomics of ectopic lesions in humans. Our transcriptomic analysis during endometriosis progression in our mouse model will help understand the pathophysiology of endometriosis.
Keywords: Endometriosis, ectopic lesions, eutopic, infertility, transcriptome, RNA-Seq
Introduction
Endometriosis is defined as the presence of endometrial-like tissues outside of the uterus. It affects about 10% of women at reproductive age and is associated with both impaired fertility and pelvic pain(1). Up to 30–50% of women with endometriosis are infertile(2) which has been attributed to the distortion of pelvic anatomy, non-receptive endometrium, compromised folliculogenesis and ovulation(3). As many as 70% of women with endometriosis experience abdominopelvic pain(4) secondary to elevated inflammation and altered innervation(5). However, the etiological and pathophysiological mechanism underlying the development of endometriosis and its symptoms remains poorly understood. Furthermore, there are no established diagnostic biomarkers, especially for the early stages of endometriosis. A history of menstrual symptoms and chronic pelvic pain provides the basis for suspecting endometriosis. No individual serum marker has yet been shown to be both sensitive and specific for diagnosis or monitoring of endometriosis. Although several screening tools and tests have been proposed and tested, none are currently validated to accurately identify or predict individuals or populations that are most likely to have the disease. Ovarian endometrioma, adhesions and deep nodular forms of disease often require ultrasonography or magnetic resonance imaging (MRI) to detect(1). Histologic verification, usually following surgical/laparoscopic visualization, can be useful in confirming diagnosis, particularly for the most common superficial lesions(6). MicroRNAs play crucial roles in endometriosis and circulating MicroRNAs have the potential to diagnose the endometriosis at early stage(7, 8). Nanoparticles accumulate at the endometriotic lesions which have both diagnosis and therapy potentials(9). Therefore, further research is needed to develop non-invasive tests for diagnosis of endometriosis. Current treatment includes suppression of estrogen action or surgery, both associated with substantial side effects and a high recurrence rate(1). Considering the great economic, physical, and psychological burdens of endometriosis on individuals and society, studies of molecular mechanisms are imperative.
A number of whole-tissue transcriptome studies have utilized DNA microarray and RNA sequencing to better understand the differences in signaling pathways between eutopic and ectopic endometrium in women with endometriosis as compared to endometrium of healthy women. Endometriosis is characterized by altered immune, angiogenic, and neurogenic processes(1). Hall and coworkers analyzed paired eutopic and ectopic lesions from nine patients by microarray and identified a gene expression signature consistent with increased activity of inflammatory pathways(10). Crispi et al. used microarray analysis to compare eight samples of paired eutopic and ectopic endometrial tissues together with six samples of control endometrium from healthy women in the proliferative phase. These investigators found that a significant alteration in expression of genes involved in organogenesis in the ectopic tissues(11). Another study using microarray compared gene expression in the eutopic and ectopic endometrial samples of 18 fertile women with ovarian (stage 3 to 4) endometriosis(12). Both proliferative and secretory stage samples were included(12). These investigators found that immune, cell remodeling, neurophysiological pathways were affected in endometriotic tissues(12). Another genome-wide study of gene expression used RNA-sequencing to evaluate eutopic and ectopic lesions from ten women with endometriosis and endometrium from nine healthy women(13). These investigators detected evidence of altered extracellular matrix, complement, and coagulation pathways in women with endometriosis(13). Microarray analysis compared eutopic endometrial tissue from 21 women with moderate to severe endometriosis to 16 women without endometriosis and identified a subset of progesterone regulated genes that are altered, suggesting progesterone resistance(14). The largest microarray analysis, including 148 endometrial samples from women with or without endometriosis, demonstrated immune activation, steroid and growth factor signaling disruption in the eutopic endometrium of women with endometriosis compared to controls(15).
More recently, single cell RNA-sequencing and spatial transcriptomic analysis has provided further clues about cellular and spatial changes occurring in the presence of endometriosis(16–20). Despite these efforts, perhaps due to differences in cycle phase, between-individual, between-lesion, and study design heterogeneity, no conserved endometriosis biomarker genes have yet been found. Therefore, we have focused on translational animal model with well-controlled experimental conditions that can potentially provide valuable insight into the effects of endometriosis on disease phenotype, gene expression, and molecular pathways vital to its progression(21).
Nonhuman primates such as the baboon and rhesus macaques have proven to be good translational animal models that share similar reproductive physiology and anatomy to humans, including menstruation and naturally occurring endometriosis (22, 23). However, the long-term disease development process, costly animal husbandry and ethical concerns limit non-human primate usage(21). Therefore, the mouse has been widely used as the preclinical model for biomedical research due to its low costs and easy manipulation(24). Recent advances in the murine model of endometriosis enhance both its feasibility and translational potential. Endometriosis can be induced in the mouse by intraperitoneal injection, surgical engraftment and subcutaneous placement of donor endometrial tissues from littermate mice or women with endometriosis(24). Fluorescent protein-labeled endometrium has been used to enhance feasibility of detection and quantitation of endometriotic lesions(24). We recently developed a mouse endometriosis model with a fluorescence reporter using Pgrcre/+Rosa26mTmG/+ that seems to enhance translational potential by mimicking both the ectopic morphology and fertility defects seen in women with endometriosis(25). As we observed in human endometriosis, we did not observe any morphological changes of the ectopic lesions between one month and three months after endometriosis induction. One month after endometriosis induction, the endometriosis mice show comparable embryo implantation sites at gestation day (GD) 7.5(25). However, three months after endometriosis induction, 63.6% mice with endometriosis revealed implantation failure(25). Interestingly, there is a time lag between lesion establishment and infertility in the mouse model, likely similar to, but much faster than that of human disease suggesting an endometriosis progression that is positively associated with infertility.
In early pregnancy the endometrium undergoes changes coordinated by the ovarian hormones progesterone (P4) and estrogen (E2)(26–29). In rodents, E2 stimulates proliferation of uterine epithelial cells on gestation day (GD) 0.5. Upon formation of the corpus luteum (GD 2.5), P4 inhibits E2-mediated proliferation of uterine epithelial cells and leads to uterine stromal cell proliferation(30). On GD 3.5, an acute E2 spike in concert with elevated P4 levels further stimulates uterine stromal proliferation and induces uterine glands to produce leukemia inhibitory factor (LIF), which is critical for uterine receptivity(31). Implantation begins at GD 4.5 with attachment of the blastocyst to the luminal epithelium, followed by invasion of the embryonic trophoblasts(32). Upon invasion, uterine stromal cells decidualize (a cellular transformation characterized by morphological changes and development of a secretory phenotype), which creates a local environment supportive of the implanting embryo(33, 34). In humans, the endometrium is also tightly regulated by P4 and E2, including proliferation, secretion, and menstrual shedding. The endometrium becomes receptive for implantation during the mid-secretory phase, and decidualized stromal cells are seen in the late-secretory phase(35, 36). Therefore GD3.5 will be a good timepoint to check both estrogen and progesterone responses of eutopic endometrium and ectopic lesions(37, 38). More importantly, although the length of pregnancy is much longer in humans than mouse, the hormone regulations during early pregnancy shares similar patterns between human and mice(39).
Therefore, we undertook a transcriptomic analysis of the murine model at GD3.5 and its temporal progression allowing analysis of changes relevant to fertility and compared these transcriptomes to those obtained from women to better understand the translational potential of our mouse model and to uncover information relevant to human disease.
MATERIALS AND METHODS
Animals
All mice were maintained in the animal facility of the Institutional Animal Care and Use Committee of University of Missouri, and all animal experimental procedures were approved. Mice were bred on a mixed background of the mouse strains C57Bl/6 and SV129 under controlled humidity and temperature.
Induction of Endometriosis and Tissue Collection
Endometriosis was surgically induced as previously described(25). Briefly, 8-weeks-old female Pgrcre/+Rosa26mTmG/+, used as uterine tissue donors, were injected with 100ng 17β-estradiol (E2) for three consecutive days. One uterine horn of each donor mouse was cut into small fragments, mixed with 0.5ml 1X PBS and injected into the peritoneal cavity of a Rosa26mTmG/+ recipient littermate. For the sham-surgery control group, 0.5ml 1X PBS, alone, was injected into the peritoneal cavity. One and three months after endometriosis induction, the endometriosis and sham mice were mated with C57BL/6 male mice. The morning of a vaginal plug was designated as day 0.5 of gestation (GD 0.5). Mice were sacrificed at GD 3.5. Rosa26mTmG/+ mice have a double-fluorescent Cre reporter and express membrane-targeted tandem dimer Tomato (mT) prior to Cre-mediated excision, and express membrane-targeted green fluorescent proteins (mG) after excision(40). While uteri and ectopic lesions with Pgr-positive uterine cells express mG in the Pgrcre/+Rosa26mTmG/+mice, other tissues with only Pgr-negative cells express mT. Endometriosis-like lesions with mG in the mice were collected under a fluorescence microscope by dissection along with eutopic endometrium. Only endometrial tissues were collected from the sham mice. Samples were either fixed with 4% (vol/vol) paraformaldehyde for histology or immunohistochemistry or snap frozen and stored at −80 °C for RNA extraction. According to our previous report(25), mice after 1-month (1M) endometriosis induction are fertile, while 63.6% mice after 3-month (3M) endometriosis induction were infertile. For each group, four 1M and 3M endometriosis mice along with four 1M and 3M sham (control) mice were used. Since the mice were dissected at GD3.5, the pregnancy status cannot be accurately predicted. A schematic diagram of this experiment is at Fig. 1A.
Figure 1.
Mouse endometriosis models recapitulate the molecular changes seen in human endometriotic lesions. A. Schematic diagram of endometriosis induction and D3.5 tissue collection. B. Hierarchy cluster of gene expression pattens in mouse endometriosis models. C. T scores of mouse ectopic gene signatures and transcriptome of multiple human endometriotic lesions. Red dot indicates the T score calculated between ectopic gene signature at 1M and the transcriptome of human endometriosis samples. Green dot indicates the T score calculated between ectopic gene signature at 3M and the transcriptome of human endometriosis samples. Positive T score means the ectopic lesions at 1M or 3M are similar to the human sample. 3MEC: 3M ectopic lesions; 1MEC: 1M ectopic lesions; 3MEU: 3M eutopic endometrium; 1MEU: 1M eutopic endometrium; 3MS: 3M sham endometrium; 1MS: 1M sham endometrium; 3M: 3 months; 1M: 1 month. *, p<0.05, compared to control endometrium (CE); #, p<0.05, 3 month compared to 1 month.
RNA isolation
The total RNA was isolated from each sample using the RNeasy Total RNA Isolation Kit (Qiagen) following the manufacture’s instructions. RNA purity and concentration were assessed using UV spectrospoy with a NanoDrop™ instrument. All RNA samples were further analyzed by the Van Andel Institute Genomics Core to confirm sample concentration and purity (RIN > 8.0; concentration 100–200ng/ul) before RNA sequencing. Multiple ectopic lesions from the same mouse were pooled together as one sample for RNA isolation.
RNA-seq analysis
For RNA sequencing, libraries were prepared from 500 ng of total RNA using the KAPA mRNA Hyperprep kit (Kapa Biosystems, Wilmington, MA USA). RNA was sheared to 300–400 bp. Prior to PCR amplification, cDNA fragments were ligated to IDT for Illumina TruSeq UD Indexed adapters (Illumina Inc, San Diego CA, USA). Quality and quantity of the finished libraries were assessed using a combination of Agilent DNA High Sensitivity chip (Agilent Technologies, Inc.) and QuantiFluor® dsDNA System (Promega Corp., Madison, WI, USA). Individually indexed libraries were pooled and 50 bp, paired end sequencing was performed on an Illumina NovaSeq6000 sequencer using an S2, 100 bp sequencing kit (Illumina Inc., San Diego, CA, USA) to an average depth of 50 M reads per sample. Base calling was done by Illumina RTA3 and output of NCS was demultiplexed and converted to FastQ format with Illumina Bcl2fastq software v1.9.0.
The raw reads were initially filtered by removing low quality reads (average quality scores < 20) and aligned to the mm39 genome and assembled using Star 2.7.9a(41). Expression values of RNA-Seq were expressed as counts. Differentially expressed genes (DEGs) were identified by the “exact negative binomial test” in the EdgeR software package(42). The DEGs between 1M eutopic and 1M ectopic tissues or between 3M eutopic and 3M ectopic tissues were set as max counts >50, fold change of >2 (up-regulated) or <−2 (down-regulated), False Discovery Rate (FDR) < 0.05. Because the great within-group variation in sham and eutopic groups, a less stringent threshold was set for the DEGs between 1M sham and 1M eutopic tissues or between 3M sham and 3M eutopic tissues: max counts >50, fold change of >1.5 (up-regulated) or <−1.5 (down-regulated), p value <0.05. A Principle Component Analysis (PCA) map were generated by plotMDS in EdgeR(42). The expressions of DEGs were normalized, scaled, clustered to generate a heatmap using the ComplexHeatmap package(43). The significantly enriched pathways and upstream regulators based on the DEGs were identified using Ingenuity pathway analysis (IPA, Qiagen). The dotplot was generated by Tidyverse package(44). The full list of mouse ligand-receptor pairs were downloaded from CelltalkDB(45) to compare with the mouse DEG lists.
Comparison with human endometriosis dataset
The signatures of mouse ectopic lesion at 1 or 3M were determined as the DEGs between ectopic lesions and eutopic endometrium with fold change of >2 (High) or <−2 (Low). The correlations of the gene signatures with the published human endometriosis dataset GSE141549 (46) were predicted by T score using the SEMIP R package (47)
Real-time PCR
Total 1ug of RNA was reverse transcribed to cDNA using M-MLV reverse transcriptase (Invitrogen) according to the manufacturer’s instructions. Quantitative real time PCR was performed with SYBR green and specific primers to examine the relative expression of genes of interest. Experimental gene expression data were normalized against the constitutively expressed gene, Rpl7 ribosomal RNA. Table S1 shows the primer sequences used.
Immunohistochemistry
Immunohistochemistry analysis was performed as previously described(25). Briefly, dewaxed hydrated paraffin-embedded tissue sections were blocking by 10% normal goat serum and then the slides were incubated with F4/80 (cs-70076, 1:250), CD3ε (cs-78588, 1:200), CD11c (cs-97585, 1:300), CD19 (cs-90176, 1:800) antibody at 4°C overnight. The slides were incubated with biotinylated goat anti-rabbit secondary antibody (BA-1000, 1:500 Vector Laboratories) for 1h, Horseradish Peroxidase (HRP) antibody (434323, 1:1000, Fisher Scientific) for 1h, and the immune signals were stained by DAB substrate (SK-4100, Vector Laboratories). The slides were counter-stained with hematoxylin. The endometrial tissues from three different mice for each group were sectioned for immunohistochemistry. For eutopic and sham endometrium at 1M or 3M, one cross section from each endometrium was evaluated at high-powered fields for immune marker staining. Because our endometriosis mice developed various number of ectopic lesions(25), 1–8 cross sections of ectopic lesions per mouse were evaluated at high-powered fields. Mouse liver and spleen has been used as positive and negative control. RL performed the staining and analyzed the images.
Statistical Analysis
Student’s t test with unequal variance was used to compare the real-time PCR and T score. A p value of <0.05 was defined, a priori, as significant.
Data availability
The RNA-Seq analysis data generated in this study have been deposited in the NCBI Gene
Expression Omnibus database under accession code GSE240392.
RESULTS
Mouse endometriosis model recapitulates the molecular changes of human endometriotic lesions
Previously we reported a novel mouse endometriosis model with subfertility using Pgrcre/+Rosa26mTmG/+ mice(25). In this model, the established peritoneal endometriotic lesions showed comparable histology to human disease. At 1M after disease establishment, the mice were fertile, but at 3M after disease establishment, the mice demonstrated severely reduced fertility due to defects of implantation and decidualization(25). In order to identify the molecular mechanisms of the endometriosis progression and its associated infertility, we performed RNA-seq analysis of the eutopic endometrium and ectopic lesions from mice at 1M or 3M after endometriosis induction, plus the endometrium of age-matched sham-surgery mice, used as controls (Fig. 1A). We collected the sham, eutopic endometrium and ectopic lesions at GD 3.5. Hierarchy cluster analysis of all the RNA-seq samples indicated that 1M and 3M ectopic lesions had similar gene expression patterns which were quite different from the eutopic and sham endometrium (Fig. 1B). In comparison with the eutopic endometrium, we identified 2,487 DEGs in the 1M ectopic lesions from fertile mice and 5,185 DEGs in the 3M ectopic lesions from subfertile mice (Table S2, 3). Previously we reported that the histologic appearance of the ectopic lesions in our mouse model is very similar to human endometriotic lesions(25). T score analysis(47) uses the gene signature from our 1M and 3M mouse ectopic lesions and gene expression profile from the published human endometriosis datasets(46) to predict the similarity of between mouse and human data. A positive T score indicates the mouse and human data had similar gene expression patterns. Since the human dataset contains the transcriptome from different endometriosis lesion types, we compared the mouse gene signature with the different types of human lesions. Here we would like to emphasize that due to the ethical issues, it is very hard to collect human endometrium during early pregnancy. We actually compared our pregnant mice with non-pregnant human. Despite of the significant variation of immune and hormone environments between pregnancy and non-pregnancy, we found that gene-expression patterns in the ectopic lesions from mice showed a positive T score with the transcriptome in human peritoneal lesions, deep-infiltrating lesions, and ovarian endometriomas, but a negative T score with the healthy and eutopic human endometrium (Fig. 1C) suggesting mouse ectopic lesion share similar expression pattern with several types of human ectopic lesions regardless of the pregnancy status. Furthermore, ectopic lesion at 3M consistently had higher positive T score compared to ectopic lesions at 1M suggesting 3M ectopic lesions were more similar to the lesions in human datasets (Fig. 1C). These results indicated that ectopic lesions from mice recapitulated the molecular changes of human endometriotic tissues.
Conserved gene expression patterns between fertile (1M) and subfertile (3M) mouse ectopic lesions
In comparison to the age matched eutopic endometrium, 2,093 DEGs were identified in both 1M and 3M ectopic lesions and, notably, differential expression were changed in the same directions, implying conserved gene networks during endometriosis progression (Fig. 2A). Pathway analysis of these conserved DEGs identified multiple widely accepted endometriosis-related pathways. Enhanced estrogen signaling and deceased and altered progesterone signaling (progesterone resistance) has been regarded as a hallmark of human disease, strongly impacting disease pathophysiology(1). These changes in steroid receptor signaling were mirrored in our mouse model (Fig. 2B). Similar to human(48, 49), enhanced estrogen signaling is the result of inhibited Estrogen receptor α (ESR1) and activated Estrogen receptor β (ESR2) signaling (Fig. 2B). Several other human endometriosis relevant pathways(50–53) including immune signaling associated with macrophage activation, enhanced T cell and B cell receptor signaling, angiogenesis associated with increased HIF1α, angiotensin, and endothelin signaling, fibrosis-related wound healing and myocyte hypertrophy, complement and coagulation cascade members were all increased in our mouse lesions. PI3K/MAPK kinases, which have been proposed as potential non-hormonal therapeutic targets for treating endometriosis(54) which were also enhanced in mouse ectopic lesion. The hypertrophic neuron growth and pronociceptive factors in mouse ectopic lesion was consistent with the elevated innervation in human endometriotic lesions(55) (Fig. 2C, Table S4).
Figure 2.
Highly conserved and distinct gene signatures are found between 1M and 3M mouse ectopic lesions. A. Venn diagram of DEGs in 1M and 3M mouse ectopic lesions. Red indicates DEGs in 1MEC compared to 1MEU. Green indicates DEGs in 3MEC compared to 3MEU. Yellow indicates overlap between the two DEG sets. B. E2 hyperactivation and P4 resistance in mouse ecotpic lesions. Top altered pathways (C, E) and upstream regulators (D, F) in the conserved DEGs between 1M and 3M ectopic lesions (C, D) and unique DEG of 3M ectopic lesions (E, F). 3MEC: 3M ectopic lesions; 1MEC: 1M ectopic lesions; 3MEU: 3M eutopic endometrium; 1MEU: 1M eutopic endometrium; 3M: 3 month; 1M: 1 month; -logP: -log(p value); FC: fold change of gene expression.
Based on the conserved DEGs in 1M and 3M ectopic lesion, potential upstream signaling regulators were identified (Fig. 2D, Table S5) and included pro-inflammatory cytokines such as TNF, IL33, TGFB1, and IL21; Increased CFB, Complement Factor B and complement components C3, the central component in complement activation, and the C5AR1, Complement C5a Receptor 1,all suggest increased inflammatory processes, while PI3 kinase p110 delta protein, PIK3CD is strongly associated with kinase activation; plasminogen activator, PLAU, and its receptor, PLAUR, and Matrix Metallopeptidase MMP13 are important for tissue remodeling and thus may play roles in fibrosis. Additionally, brain derived neurotrophic factor (BDNF), nitric oxide synthase, NOS2, can play important roles in promoting neurogenesis and angiogenesis associated with endometriosis pathophysiology.
Unique gene signatures in subfertile (3M) mouse ectopic lesions
In order to gain insight into the change in fertility between 1M and 3M and to understand molecular change during disease progression, we identified the DEGs between 1M and 3M in the ectopic lesions. There are no obvious morphological changes from 1M to 3M after endometriosis induction between in the ectopic lesions. However, 3,093 DEGs were distinct to the 3M timepoint, indicating a significant transcriptomic difference between ectopic lesions during endometriosis progression, associated with acquired subfertility. Pathway analysis of the 3M unique DEG identified activated metabolism signaling associated with cholesterol, glucose, and estrogen; myocyte signaling related to cardiac hypertrophy, oxytocin, MAPK and stem cell pluripotency and inhibited sonic hedgehog signaling (Fig. 2E, Table S6). Analysis of potential upstream regulators (Fig. 2F, Table S7) identified cholesterol-steroid metabolism, luteinizing hormone receptor LHCGR, steroidogenic factor NR5A1, leptin LEP were all activated; for myocyte, muscle specific transcription factor MYOD1, MYOG, and oxytocin receptor OXTR were all stimulated; The expression of stem cell markers such as RUNX2, TP63, PAX3 were all increased. All these results suggested an endometriosis progression from 1M to 3M ectopic at molecular levels.
Elevated immune signaling in 1M and 3M ectopic lesions
Endometriosis is characterized by the ectopic focal inflammation(50). We found that multiple immune pathways are activated in our mouse ectopic lesions compared to the eutopic endometrium at both 1M and 3M (Fig. 2C). A gene expression heat map further showed enriched dendritic cell, macrophage, T and B cell markers in the 1M and 3M mouse ectopic lesions compared to the eutopic endometrium (Fig. 3A), which suggested the abnormal accumulation of multiple immune cells in the ectopic lesions. In order to confirm the gene expression data, we performed immunohistochemistry using antibodies against CD11c(56), F4/80(57), CD3ε(58) and CD19(59) as markers for dendritic cells, macrophage, T cell and B cell, respectively. As expected, we found abundant membrane and cytoplasmic staining of CD11c and CD19 in spleen and F4/80 staining in liver, and cytoplasmic staining of CD3ε in spleen, while much lower expression of CD11c, CD19, CD3ε in liver and F4/80 in spleen which is consistent with previous publications(60–66) (Fig. S1). We found (Fig 3B,) that CD11c, CD3 and CD19 positive cells were rarely detected in the sham and eutopic endometrium but abundantly present in multiple regions of 1M and 3M ectopic lesions. Further, sham and eutopic endometrium contained F4/80 positive cells, but these were increased in several regions of the ectopic tissues. Therefore, the increased infiltration of immune cells was detected in the 1M and 3M ectopic lesions, paralleling prior studies in human lesions showing increased immune cell infiltration(67, 68).
Figure 3.
Elevated immune signaling in mouse ectopic lesions. A. Heatmap of dendritic, macrophage, T and B cell related genes in 1M and 3M mouse eutopic and ectopic lesions. B. Representative immunohistochemistry of CD11c, F4/80, CD3ε and CD19 in sham and eutopic endometrium, 1M and 3M ectopic lesions. 3MEC: 3M ectopic lesions; 1MEC: 1M ectopic lesions; 3MEU: 3M eutopic endometrium; 1MEU: 1M eutopic endometrium; 3M: 3 months; 1M: 1 month.
Fertility related transcriptomic changes in 3M eutopic endometrium from subfertile mice
The molecular mechanism of endometriosis associated fertility remains largely unknown in which uterine defects play one of the important roles. Our endometriosis mouse models showed severe subfertility 3M after endometriosis induction in contrast to the normal fertility seen in the control mice and those 1M after endometriosis induction(25). Further studies revealed that the reduced fertility in 3M endometriosis mice was directly caused by uterine decidualization defects(25). To understand endometriosis induced pregnancy related uterine defects, we compared the transcriptome of eutopic with sham endometrium at pregnancy D3.5. The PCA plot and hierarchy cluster heatmap, we noticed all 1M sham (n=2), 1M eutopic (n=4), 3M control (n=3) and 2 of 4 3M eutopic samples were clustered together. Separately, one 3M sham, two 1M sham, and two 3M eutopic samples were spread out (Fig. S2A, B). The variable fertility in endometriosis mice(25) and sham mice might explain the within-group transcriptomic differences. In accordance with the normal fertility at 1M and severe subfertility at 3M, there was very limited overlap between 1M and 3M DEGs, in which 92% (650/608) DEGs of 3M eutopic endometrium were only altered at 3M not 1M (Fig. S2C, Table S8, 9). Analysis of upstream regulators and pathways revealed activation of ESR1 and IL21 signaling as well as suppression of Wnt-β/catenin signaling CTNNB1, TGFβ signaling BMP4, and EGF (Fig. S2D, Table S10), which play crucial roles in embryo implantation(69–71). Since immune and EGF pathways were altered in the eutopic endometrium, we also performed qPCR to validate the reduced expression of the cytokine, IL15, the cytokine receptor Il20rb, the increased expression of immune and EGF related Serpina3n, and the EGF regulator Egfl6, in the 3M eutopic compared to sham uterus (Fig. S2E).
The interactions between ectopic and eutopic endometrium
In our endometriosis models, we found that the presence of endometrial tissues outside of the uterus severely disrupted uterine functions during pregnancy(25). A long-standing question in endometriosis research is, how can ectopic lesions disrupt eutopic endometrial functions? We hypothesized interactions between ectopic lesion ligands and eutopic endometrial receptors may play a role. Although elevated inflammatory mediators, inflammatory cells, and growth factors (Fig. 2) may play a systemic role, it is likely that higher levels of these factors in the peritoneal fluid could have an increased role in the pelvic region. Additionally, we found the steroid synthetic enzymes, especially for progesterone and estrogen, were increased in the 3M ectopic lesions which may increase the local estrogen and possibly progesterone concentrations, though progesterone is metabolized to estrogen (Fig. 4A). Meanwhile, the receptors of estrogen and progesterone, ESR1 and PGR, maintained high levels of expression in the eutopic uterus. Therefore, a hyper-estrogen (and possibly progesterone) environment in the eutopic endometrium may be induced by the ectopic lesions (Fig. 4A).
Figure 4.
Interactions between ectopic and eutopic endometrium. (A) Heatmap of E2 and P4 synthesis and receptor related gene expression. (B) Schematic diagram of the overall eutopic and ectopic endometrial interactions. The thickness of the line is proportional to the strength of the interactions. The arrow points from the ligand producer to receptor producer. C. Dot plot of the top altered ligand-receptor pairs between eutopic and ectopic endometrium. 3MEC: 3M ectopic lesions; 1MEC: 1M ectopic lesions; 3MEU: 3M eutopic endometrium; 1MEU: 1M eutopic endometrium; 3M: 3 months; 1M: 1 month.
Although peritoneal endometriotic lesions cannot physically contact the eutopic endometrium, the peritoneal fluid may circulate some ligand-receptor pairs between ectopic and eutopic endometrium to mediate the paracrine interactions. To quantify the ligand receptor interactions, we set the sham endometrium as the base line and calculated the expression fold changes of the ligand and receptor in the eutopic and ectopic endometrium. Eventually, we found a great variety of ligand and receptor mRNA were altered in the ectopic lesions (ligand: 220 (1M), 289 (3M); receptor: 203 (1M), 309 (3M), Fig. S3A), while much fewer were changed in the eutopic endometrium (ligand: 15 (1M), 31 (3M); receptor: 7 (1M), 31 (3M), Fig. S3A). Among them, more than 50% ligands and receptors were upregulated in the ectopic lesion, but less than 50% were upregulated in the eutopic endometrium (Fig. S3B). Because of the abundantly altered ligand-receptor expression, the interactions within the ectopic lesions were more frequently changed compared to the eutopic endometrium (Fig. 4B). Meanwhile, many secreted ligand including cytokines, growth factors, and lipoprotein etc. and their receptors were differentially expressed in the ectopic and eutopic endometrium, which may greatly affect the interactions between these two endometrial tissues (Fig. 4C).
DISCUSSION
Endometriosis is a chronic inflammatory disease that involves both the peripheral immune system and endometrial tissues(72). Elevated inflammation has been reported in the transcriptomic analysis of human endometriotic tissues(10, 15). Recent advancement of scRNA-seq and spatial transcriptome further emphasized the pro-inflammatory microenvironment in different types of endometriotic cells (16–20). Normal endometrium also contains abundant immune cells to support the uterine remodeling during menstrual cycle and pregnancy(73). The dysregulated immune cells are central to the pathophysiology of endometriosis(72). Macrophages and dendritic cells are increased in the ectopic lesions and the peritoneal fluid of women with endometriosis to promote the organogenesis, angiogenesis and neurogenesis of ectopic lesion and may also contribute to the infertility and pelvic pain(72, 74–77). There are many contradictory results about the density of different types of T cell and B cell in the eutopic and ectopic endometrial tissues, peritoneal fluid, and peripheral blood in which reduce, increase or non-change has all been reported which may affect the immune tolerance(72, 73). Our endometriosis mouse model also showed activated immune signaling and abundant expression of immune cell markers in the ectopic lesions, including genes associated with macrophage, dendritic and T cell, etc. More interestingly, we noticed the clonal immune cell accumulation in some regions of ectopic lesion suggesting the heterogeneous immune infiltration in the endometriotic tissues. The tissue heterogeneity provides one explanation for the conflicting immune results in human endometrial samples. Therefore, a systematic study using spatial transcriptome will be a better method to reveal the complex immune cell distribution and its crosstalk with adjacent microenvironment in endometriotic tissues.
Endometrium is tightly regulated by estrogen and progesterone through their receptors ESRs and PGR. Disrupted steroid signaling causes multiple uterine diseases. Among them, progesterone resistance and estrogen dominance is the hallmark of endometriosis(1). The reduced PGR expression, especially the undetectable PGR-B isoform, may cause the progesterone resistance(78, 79). It has been reported that ESR1 and ESR2 were both expressed in the endometrium, while a switch from ESR1 to ESR2 dominancy was observed in endometriotic tissues(80). Besides, the steroid ligand, estrogen and progesterone levels are both increased in the endometriotic tissues due to the elevated expression of steroidogenesis enzymes(81, 82). The higher estrogen levels and ESR2 expression may contribute to the estrogen dominance. However, since many of these results were obtained from the human ovarian endometrioma, the confounding factor from ovarian tissues cannot be fully ruled out. In our endometriosis mouse model, the ectopic lesions has only been observed in the pelvic not the ovarian locations. But the steroidogenesis enzymes and Esr2 expression were increased while Esr1 and Pgr were decreased in the ectopic lesions confirming that these are conserved characteristics in endometriotic lesions.
We used the GD3.5 mouse uterus instead of the non-pregnant uterus based on three major reasons. First, we used uterine samples from mice at GD3.5 since they have similar ovarian steroid conditions as the early secretory phase of humans. A key aspect of endometrial biology is the elaborate crosstalk that occurs between these tissue compartments during early pregnancy via paracrine factors regulated by progesterone and estrogen. Dysregulating this crosstalk often leads to early pregnancy loss(83–87). The main differences in gene expression between normal and eutopic endometrium from women with endometriosis is predominantly in the early secretory phase(14, 88). Therefore, we used eutopic and ectopic endometrial tissues to identify gene pathways and networks that differ among fertile control and subfertile mice with endometriosis at GD 3.5. Second, the pregnancy events and hormone environments during early pregnancy are well-controlled. In mice, the ovulatory estrogen surge at GD0.5 was followed by increased progesterone levels and one nidatory estrogen at GD3.5 (89, 90). Therefore, GD3.5 will be a good timepoint to check both progesterone and estrogen responses of the eutopic endometrium and ectopic lesions. Although the length of pregnancy is much longer in humans than mice, the hormone regulations during early pregnancy shares similar patterns between humans and mice(39). Third, it is difficult to interpret the results from non-pregnant mice because non-pregnant mice continuously undergo variable estrous cycles that last 4–5 days. More importantly, mice do not have a progesterone dominance during the estrous stage that is comparable to the human secretory phase. Endometriosis disrupts coordinated progesterone responses in the endometrium. This progesterone resistance results in an inadequate antagonism of estrogen action, increased inflammation, and inadequate differentiation of the stroma and remodeling of the endometrium, all of which can lead to a non-receptive endometrium for embryo implantation(84). However, the mechanism of progesterone resistance in endometriosis is not fully understood. Therefore, GD3.5 with high progesterone levels could be a better time point to study the progesterone resistance mechanism in endometriosis mouse model.
One limitation of this study is there are great within-group variations in the sham and eutopic groups. We previously reported that our endometriosis mice displayed a range of fertility from normal to infertile, which is similar to that seen in women with endometriosis(25). 63.6% of mice with endometriosis at 3M had implantation failure which were not correlated with the number or mass of the ectopic lesions (25). Therefore, some mice have non-receptive endometrium and others have receptive endometrium at pre-implantation stage GD3.5. We speculate that these different endometrial conditions result in variations of transcriptomics within the group in eutopic and sham endometrium. Consequently, much less DEGs were detected with statistically lower significance in the eutopic compared to the sham endometrium. While this subfertility phenotype of our animal model mimics to endometriosis-associated infertility in some women with endometriosis, there is a limitation for identification of dysregulated genes and pathways in mixture of non-receptive and receptive endometrium from mice with endometriosis. Furthermore, the identified DEGs could be indirect effects of genetic, hormonal and immune system changes associated with conception. Future studies will include an increased number of endometriosis mice, unsupervised clustering, and collecting endometrial tissues at later pregnancy stages when fertility defects are more obvious as these may improve our understanding of the heterogeneous endometrial transcriptome between fertile or infertile eutopic endometrium.
The uterus is supplied with various types of neurons including sensory, postganglionic sympathetic and parasympathetic neurons(91). Multiple neurotrophic and neuro growth factors, (including brain-derived neurotrophic factor (BDNF)(92), neuropeptide Y (NPY)(93), and nerve growth factor (NGF)(94)) are synthesized in situ in the endometrium. They have a role for neurogenesis as well as modulation of epithelium proliferation, muscle contraction and prostaglandin production. The nerve fibers are dense in myometrium and reach the functional layers in the fetus uterus(95). However, they are confined in the basal layer of adult endometrium. Furthermore, the nerve fibers are detected in the functional layer of women with endometriosis(96) or with pain symptoms(97). There are more nerve fibers in ectopic lesions than peritoneum(98), which may sensitize 73% women with endometriosis toward the pelvic pain(4). In addition to the pain, the innervated neurons interact with immune cells, blood vessels and endometrial cells to promote the inflammation and angiogenesis in endometriotic tissues(55). Right now, there are no long-term relief of endometriosis-associated pain treatment without adverse effects on fertility(99). Our endometriosis mouse model is manifested with neurotrophic factors, cytokines and vascular growth factors in the ectopic lesions implying its application in improving research of endometriosis-associated pain.
CONCLUSION
In this study, we performed transcriptomic analysis in ectopic lesions and eutopic endometrial tissues from both fertile and subfertile mice with endometriosis. We identified the positive correlation of the gene signatures between the mouse and human in ectopic lesions. Conserved gene networks were activated in all the ectopic lesions including estradiol, immune, fibrosis, and angiogenesis pathways. The interactions mediated through hormone, cytokine, and growth factor as well as their corresponding receptors were predicted between the ectopic and eutopic endometrium. EGF and WNT signaling were more suppressed in the eutopic endometrium from subfertile mice. Our results revealed that our mouse endometriosis model recapitulates the important transcriptomic changes of endometriosis progression in human ectopic lesions including the essential regulator network and intensive inter-communications between ectopic and eutopic endometrium. Our preclinical animal model for endometriosis will be invaluable to understand etiology and pathophysiology on endometriosis.
Supplementary Material
Acknowledgement
The authors thank the Van Andel Genomics Core for providing RNA-seq services and Krystina Dunston for proofreading.
Statement:
The research and writing of this publication were supported in part by the Eunice Kennedy Shriver National Institute of Child Health & Human Development of the National Institutes of Health under Award Number R01HD084478, R01HD102170, R01HD101243, and P01HD106485 to J.W.J. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.
Footnotes
Disclosure Statement: Drs. Lessey and Young disclose licensed intellectual property related to endometriosis diagnosis.
Attestation Statement: Data in the study has not been previously published. Data will be made available to the editors of the journal for review of query upon request.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Zondervan KT, Becker CM , and Missmer SA Endometriosis. N Engl J Med 2020:1244–56. [DOI] [PubMed] [Google Scholar]
- 2.Bulletti C, Coccia ME, Battistoni S, Borini A. Endometriosis and infertility. J Assist Reprod Genet 2010;27:441–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Bonavina G, Taylor HS. Endometriosis-associated infertility: From pathophysiology to tailored treatment. Front Endocrinol (Lausanne) 2022;13:1020827. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Ballard KD, Seaman HE, de Vries CS, Wright JT. Can symptomatology help in the diagnosis of endometriosis? Findings from a national case-control study - Part 1. Bjog-Int J Obstet Gy 2008;115:1382–91. [DOI] [PubMed] [Google Scholar]
- 5.Maddern J, Grundy L, Castro J, Brierley SM. Pain in Endometriosis. Front Cell Neurosci 2020;14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.European Society for Human R, Embryology Guideline Group on POI, Webber L, Davies M, Anderson R, Bartlett J et al. ESHRE Guideline: management of women with premature ovarian insufficiency. Hum Reprod 2016;31:926–37. [DOI] [PubMed] [Google Scholar]
- 7.Bjorkman S, Taylor HS. MicroRNAs in endometriosis: biological function and emerging biomarker candidatesdagger. Biol Reprod 2019;100:1135–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Bendifallah S, Dabi Y, Suisse S, Jornea L, Bouteiller D, Touboul C et al. MicroRNome analysis generates a blood-based signature for endometriosis. Sci Rep 2022;12:4051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Moses AS, Demessie AA, Taratula O, Korzun T, Slayden OD, Taratula O. Nanomedicines for Endometriosis: Lessons Learned from Cancer Research. Small 2021;17:e2004975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Hull ML, Escareno CR, Godsland JM, Doig JR, Johnson CM, Phillips SC et al. Endometrial-peritoneal interactions during endometriotic lesion establishment. Am J Pathol 2008;173:700–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Crispi S, Piccolo MT, D’Avino A, Donizetti A, Viceconte R, Spyrou M et al. Transcriptional profiling of endometriosis tissues identifies genes related to organogenesis defects. J Cell Physiol 2013;228:1927–34. [DOI] [PubMed] [Google Scholar]
- 12.Khan MA, Sengupta J, Mittal S, Ghosh D. Genome-wide expressions in autologous eutopic and ectopic endometrium of fertile women with endometriosis. Reprod Biol Endocrinol 2012;10:84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Predeus AV, Vashukova ES, Glotov AS, Danilova MM, Osinovskaya NS, Malysheva OV et al. Next-Generation Sequencing of Matched Ectopic and Eutopic Endometrium Identifies Novel Endometriosis-Related Genes. Russ J Genet+ 2018;54:1358–65. [Google Scholar]
- 14.Burney RO, Talbi S, Hamilton AE, Vo KC, Nyegaard M, Nezhat CR et al. Gene expression analysis of endometrium reveals progesterone resistance and candidate susceptibility genes in women with endometriosis. Endocrinology 2007;148:3814–26. [DOI] [PubMed] [Google Scholar]
- 15.Tamaresis JS, Irwin JC, Goldfien GA, Rabban JT, Burney RO, Nezhat C et al. Molecular classification of endometriosis and disease stage using high-dimensional genomic data. Endocrinology 2014;155:4986–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Tan Y, Flynn WF, Sivajothi S, Luo D, Bozal SB, Dave M et al. Single-cell analysis of endometriosis reveals a coordinated transcriptional programme driving immunotolerance and angiogenesis across eutopic and ectopic tissues. Nat Cell Biol 2022;24:1306–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Fonseca MAS, Haro M, Wright KN, Lin X, Abbasi F, Sun J et al. Single-cell transcriptomic analysis of endometriosis. Nat Genet 2023;55:255–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Ma J, Zhang L, Zhan H, Mo Y, Ren Z, Shao A et al. Single-cell transcriptomic analysis of endometriosis provides insights into fibroblast fates and immune cell heterogeneity. Cell Biosci 2021;11:125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Zhu S, Wang A, Xu W, Hu L, Sun J, Wang X. The heterogeneity of fibrosis and angiogenesis in endometriosis revealed by single-cell RNA-sequencing. Biochim Biophys Acta Mol Basis Dis 2023;1869:166602. [DOI] [PubMed] [Google Scholar]
- 20.Huang X, Wu L, Pei T, Liu D, Liu C, Luo B et al. Single-cell transcriptome analysis reveals endometrial immune microenvironment in minimal/mild endometriosis. Clin Exp Immunol 2023;212:285–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Lagana AS, Garzon S, Franchi M, Casarin J, Gullo G, Ghezzi F. Translational animal models for endometriosis research: a long and windy road. Ann Transl Med 2018;6:431. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Nishimoto-Kakiuchi A, Netsu S, Okabayashi S, Taniguchi K, Tanimura H, Kato A et al. Spontaneous endometriosis in cynomolgus monkeys as a clinically relevant experimental model. Hum Reprod 2018;33:1228–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Fazleabas AT, Brudney A, Gurates B, Chai D, Bulun S. A modified baboon model for endometriosis. Ann N Y Acad Sci 2002;955:308–17; discussion 40–2, 96–406. [DOI] [PubMed] [Google Scholar]
- 24.Burns KA, Pearson AM, Slack JL, Por ED, Scribner AN, Eti NA et al. Endometriosis in the Mouse: Challenges and Progress Toward a ‘Best Fit’ Murine Model. Front Physiol 2021;12:806574. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Yoo JY, Kim TH, Shin JH, Marquardt RM, Muller U, Fazleabas AT et al. Loss of MIG-6 results in endometrial progesterone resistance via ERBB2. Nature communications 2022;13:1101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Patel B, Elguero S, Thakore S, Dahoud W, Bedaiwy M, Mesiano S. Role of nuclear progesterone receptor isoforms in uterine pathophysiology. Human reproduction update 2015;21:155–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Vasquez YM. Estrogen-regulated transcription: Mammary gland and uterus. Steroids 2018;133:82–6. [DOI] [PubMed] [Google Scholar]
- 28.Hamilton KJ, Hewitt SC, Arao Y, Korach KS. Estrogen Hormone Biology. Curr Top Dev Biol 2017;125:109–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Wetendorf M, DeMayo FJ. The progesterone receptor regulates implantation, decidualization, and glandular development via a complex paracrine signaling network. Molecular and cellular endocrinology 2012;357:108–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Franco HL, Jeong JW, Tsai SY, Lydon JP, DeMayo FJ. In vivo analysis of progesterone receptor action in the uterus during embryo implantation. Seminars in cell & developmental biology 2008;19:178–86. [DOI] [PubMed] [Google Scholar]
- 31.Stewart CL, Kaspar P, Brunet LJ, Bhatt H, Gadi I, Kontgen F et al. Blastocyst implantation depends on maternal expression of leukaemia inhibitory factor. Nature 1992;359:76–9. [DOI] [PubMed] [Google Scholar]
- 32.Lee KY, DeMayo FJ. Animal models of implantation. Reproduction 2004;128:679–95. [DOI] [PubMed] [Google Scholar]
- 33.Ochoa-Bernal MA, Fazleabas AT. Physiologic Events of Embryo Implantation and Decidualization in Human and Non-Human Primates. International journal of molecular sciences 2020;21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Okada H, Tsuzuki T, Murata H. Decidualization of the human endometrium. Reprod Med Biol 2018;17:220–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.von Wolff M, Thaler CJ, Strowitzki T, Broome J, Stolz W, Tabibzadeh S. Regulated expression of cytokines in human endometrium throughout the menstrual cycle: dysregulation in habitual abortion. Molecular human reproduction 2000;6:627–34. [DOI] [PubMed] [Google Scholar]
- 36.Dimitriadis E, Stoikos C, Tan YL, Salamonsen LA. Interleukin 11 signaling components signal transducer and activator of transcription 3 (STAT3) and suppressor of cytokine signaling 3 (SOCS3) regulate human endometrial stromal cell differentiation. Endocrinology 2006;147:3809–17. [DOI] [PubMed] [Google Scholar]
- 37.Caligioni CS. Assessing reproductive status/stages in mice. Curr Protoc Neurosci 2009;Appendix 4:Appendix 4I. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Wall EG, Desai R, Khant Aung Z, Yeo SH, Grattan DR, Handelsman DJ et al. Unexpected Plasma Gonadal Steroid and Prolactin Levels Across the Mouse Estrous Cycle. Endocrinology 2023;164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Wang H, Dey SK. Roadmap to embryo implantation: clues from mouse models. Nat Rev Genet 2006;7:185–99. [DOI] [PubMed] [Google Scholar]
- 40.Muzumdar MD, Tasic B, Miyamichi K, Li L, Luo L. A global double-fluorescent Cre reporter mouse. Genesis 2007;45:593–605. [DOI] [PubMed] [Google Scholar]
- 41.Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 2013;29:15–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Robinson MD, McCarthy DJ, Smyth GK. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 2010;26:139–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Gu ZG, Eils R, Schlesner M. Complex heatmaps reveal patterns and correlations in multidimensional genomic data. Bioinformatics 2016;32:2847–9. [DOI] [PubMed] [Google Scholar]
- 44.Wickham H, Averick M, Bryan J, Chang W, McGowan LD, François R et al. Welcome to the tidyverse. Journal of Open Source Software 2019;4:1686. [Google Scholar]
- 45.Shao X, Liao J, Li C, Lu X, Cheng J, Fan X. CellTalkDB: a manually curated database of ligand-receptor interactions in humans and mice. Brief Bioinform 2021;22. [DOI] [PubMed] [Google Scholar]
- 46.Gabriel M, Fey V, Heinosalo T, Adhikari P, Rytkonen K, Komulainen T et al. A relational database to identify differentially expressed genes in the endometrium and endometriosis lesions. Sci Data 2020;7:284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Li J, Bushel PR, Lin L, Day K, Wang T, DeMayo FJ et al. Structural Equation Modeling of In silico Perturbations. Front Genet 2021;12:727532. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Brandenberger AW, Lebovic DI, Tee MK, Ryan IP, Tseng JF, Jaffe RB et al. Oestrogen receptor (ER)-alpha and ER-beta isoforms in normal endometrial and endometriosis-derived stromal cells. Mol Hum Reprod 1999;5:651–5. [DOI] [PubMed] [Google Scholar]
- 49.Fujimoto J, Hirose R, Sakaguchi H, Tamaya T. Expression of oestrogen receptor-alpha and - beta in ovarian endometriomata. Mol Hum Reprod 1999;5:742–7. [DOI] [PubMed] [Google Scholar]
- 50.Guo B, Chen JH, Zhang JH, Fang Y, Liu XJ, Zhang J et al. Pattern-recognition receptors in endometriosis: A narrative review. Front Immunol 2023;14:1161606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Chung MS, Han SJ. Endometriosis-Associated Angiogenesis and Anti-angiogenic Therapy for Endometriosis. Front Glob Womens Health 2022;3:856316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Garcia Garcia JM, Vannuzzi V, Donati C, Bernacchioni C, Bruni P, Petraglia F. Endometriosis: Cellular and Molecular Mechanisms Leading to Fibrosis. Reprod Sci 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Yu L, Shen H, Ren X, Wang A, Zhu S, Zheng Y et al. Multi-omics analysis reveals the interaction between the complement system and the coagulation cascade in the development of endometriosis. Sci Rep 2021;11:11926. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.McKinnon BD, Kocbek V, Nirgianakis K, Bersinger NA, Mueller MD. Kinase signalling pathways in endometriosis: potential targets for non-hormonal therapeutics. Human reproduction update 2016;22:382–403. [DOI] [PubMed] [Google Scholar]
- 55.Godin SK, Wagner J, Huang P, Bree D. The role of peripheral nerve signaling in endometriosis. FASEB Bioadv 2021;3:802–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Wu J, Wu H, An J, Ballantyne CM, Cyster JG. Critical role of integrin CD11c in splenic dendritic cell capture of missing-self CD47 cells to induce adaptive immunity. Proc Natl Acad Sci U S A 2018;115:6786–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Austyn JM, Gordon S. F4/80, a monoclonal antibody directed specifically against the mouse macrophage. Eur J Immunol 1981;11:805–15. [DOI] [PubMed] [Google Scholar]
- 58.Grosjean C, Quessada J, Nozais M, Loosveld M, Payet-Bornet D, Mionnet C. Isolation and enrichment of mouse splenic T cells for ex vivo and in vivo T cell receptor stimulation assays. STAR Protoc 2021;2:100961. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.He X, Klasener K, Iype JM, Becker M, Maity PC, Cavallari M et al. Continuous signaling of CD79b and CD19 is required for the fitness of Burkitt lymphoma B cells. EMBO J 2018;37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Johnson BA 3rd, Kahler DJ, Baban B, Chandler PR, Kang B, Shimoda M et al. B-lymphoid cells with attributes of dendritic cells regulate T cells via indoleamine 2,3-dioxygenase. Proc Natl Acad Sci U S A 2010;107:10644–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Wang JQ, Tang Y, Li QS, Xiao M, Li M, Sheng YT et al. PARG regulates the proliferation and differentiation of DCs and T cells via PARP/NF‑kappaB in tumour metastases of colon carcinoma. Oncol Rep 2019;41:2657–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Gao LM, Liu WP, Yang QP, Li HF, Chen JJ, Tang Y et al. Aggressive natural killer-cell leukemia with jaundice and spontaneous splenic rupture: a case report and review of the literature. Diagn Pathol 2013;8:43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Melis M, Tang XH, Trasino SE, Patel VM, Stummer DJ, Jessurun J et al. Effects of AM80 compared to AC261066 in a high fat diet mouse model of liver disease. PLoS One 2019;14:e0211071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Taylor PR, Martinez-Pomares L, Stacey M, Lin HH, Brown GD, Gordon S. Macrophage receptors and immune recognition. Annu Rev Immunol 2005;23:901–44. [DOI] [PubMed] [Google Scholar]
- 65.Galle-Treger L, Helou DG, Quach C, Howard E, Hurrell BP, Muench GRA et al. Autophagy impairment in liver CD11c cells promotes non-alcoholic fatty liver disease through production of IL-23. Nature Communications 2022;13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Mehal WZ, Azzaroli F, Crispe IN. Immunology of the healthy liver: old questions and new insights. Gastroenterology 2001;120:250–60. [DOI] [PubMed] [Google Scholar]
- 67.Jones RK, Bulmer JN, Searle RF. Phenotypic and functional studies of leukocytes in human endometrium and endometriosis. Hum Reprod Update 1998;4:702–9. [DOI] [PubMed] [Google Scholar]
- 68.Klein NA, Pergola GM, Tekmal RR, Montoya IA, Dey TD, Schenken RS. Cytokine regulation of cellular proliferation in endometriosis. Ann N Y Acad Sci 1994;734:322–32. [DOI] [PubMed] [Google Scholar]
- 69.Mohamed OA, Jonnaert M, Lebelle-Dumais CL, Kuroda K, Clarke HJ, Dufort D. Uterine Wnt/β-catenin signaling is required for implantation. P Natl Acad Sci USA 2005;102:8579–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Ni N, Li Q. TGFbeta superfamily signaling and uterine decidualization. Reprod Biol Endocrinol 2017;15:84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Large MJ, Wetendorf M, Lanz RB, Hartig SM, Creighton CJ, Mancini MA et al. The epidermal growth factor receptor critically regulates endometrial function during early pregnancy. PLoS Genet 2014;10:e1004451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Vallve-Juanico J, Houshdaran S, Giudice LC. The endometrial immune environment of women with endometriosis. Human reproduction update 2019;25:564–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Meyer N, Zenclussen AC. Immune Cells in the Uterine Remodeling: Are They the Target of Endocrine Disrupting Chemicals? Front Immunol 2020;11:246. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Hogg C, Horne AW, Greaves E. Endometriosis-Associated Macrophages: Origin, Phenotype, and Function. Front Endocrinol (Lausanne) 2020;11:7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Qiaomei Z, Ping W, Yanjing Z, Jinhua W, Shaozhan C, Lihong C. Features of peritoneal dendritic cells in the development of endometriosis. Reprod Biol Endocrinol 2023;21:4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Schulke L, Berbic M, Manconi F, Tokushige N, Markham R, Fraser IS. Dendritic cell populations in the eutopic and ectopic endometrium of women with endometriosis. Hum Reprod 2009;24:1695–703. [DOI] [PubMed] [Google Scholar]
- 77.Laginha PA, Arcoverde FVL, Riccio LGC, Andres MP, Abrao MS. The role of dendritic cells in endometriosis: A systematic review. J Reprod Immunol 2022;149:103462. [DOI] [PubMed] [Google Scholar]
- 78.Attia GR, Zeitoun K, Edwards D, Johns A, Carr BR, Bulun SE. Progesterone receptor isoform A but not B is expressed in endometriosis. J Clin Endocrinol Metab 2000;85:2897–902. [DOI] [PubMed] [Google Scholar]
- 79.Bedaiwy MA, Dahoud W, Skomorovska-Prokvolit Y, Yi L, Liu JH, Falcone T et al. Abundance and Localization of Progesterone Receptor Isoforms in Endometrium in Women With and Without Endometriosis and in Peritoneal and Ovarian Endometriotic Implants. Reprod Sci 2015;22:1153–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Yilmaz BD, Bulun SE. Endometriosis and nuclear receptors. Hum Reprod Update 2019;25:473–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Attar E, Tokunaga H, Imir G, Yilmaz MB, Redwine D, Putman M et al. Prostaglandin E2 via steroidogenic factor-1 coordinately regulates transcription of steroidogenic genes necessary for estrogen synthesis in endometriosis. J Clin Endocrinol Metab 2009;94:623–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Noble LS, Simpson ER, Johns A, Bulun SE. Aromatase expression in endometriosis. J Clin Endocrinol Metab 1996;81:174–9. [DOI] [PubMed] [Google Scholar]
- 83.Lessey BA, Young SL. What exactly is endometrial receptivity? Fertility and sterility 2019;111:611–7. [DOI] [PubMed] [Google Scholar]
- 84.Lessey BA, Kim JJ. Endometrial receptivity in the eutopic endometrium of women with endometriosis: it is affected, and let me show you why. Fertility and sterility 2017;108:19–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Fox C, Morin S, Jeong JW, Scott RT Jr., Lessey BA Local and systemic factors and implantation: what is the evidence? Fertility and sterility 2016;105:873–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Bulun SE, Yilmaz BD, Sison C, Miyazaki K, Bernardi L, Liu S et al. Endometriosis. Endocrine reviews 2019;40:1048–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Bulun SE, Monsivais D, Kakinuma T, Furukawa Y, Bernardi L, Pavone ME et al. Molecular biology of endometriosis: from aromatase to genomic abnormalities. Semin Reprod Med 2015;33:220–4. [DOI] [PubMed] [Google Scholar]
- 88.McKinnon B, Mueller M, Montgomery G. Progesterone Resistance in Endometriosis: an Acquired Property? Trends in endocrinology and metabolism: TEM 2018;29:535–48. [DOI] [PubMed] [Google Scholar]
- 89.McCormack JT, Greenwald GS. Evidence for a preimplantation rise in oestradiol-17beta levels on day 4 of pregnancy in the mouse. J Reprod Fertil 1974;41:297–301. [DOI] [PubMed] [Google Scholar]
- 90.Psychoyos A Hormonal control of ovoimplantation. Vitam Horm 1973;31:201–56. [DOI] [PubMed] [Google Scholar]
- 91.Monica Brauer M, Smith PG. Estrogen and female reproductive tract innervation: cellular and molecular mechanisms of autonomic neuroplasticity. Auton Neurosci 2015;187:1–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Lim W, Bae H, Bazer FW, Song G. Brain-derived neurotrophic factor improves proliferation of endometrial epithelial cells by inhibition of endoplasmic reticulum stress during early pregnancy. J Cell Physiol 2017;232:3641–51. [DOI] [PubMed] [Google Scholar]
- 93.Jana B, Calka J, Palus K. Inflammation changes the expression of neuropeptide Y receptors in the pig myometrium and their role in the uterine contractility. PLoS One 2020;15:e0236044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Maranesi M, Parillo F, Leonardi L, Rebollar PG, Alonso B, Petrucci L et al. Expression of nerve growth factor and its receptors in the uterus of rabbits: functional involvement in prostaglandin synthesis. Domest Anim Endocrinol 2016;56:20–8. [DOI] [PubMed] [Google Scholar]
- 95.Pinsard M, Mouchet N, Dion L, Bessede T, Bertrand M, Darai E et al. Anatomic and functional mapping of human uterine innervation. Fertil Steril 2022;117:1279–88. [DOI] [PubMed] [Google Scholar]
- 96.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;21:782–7. [DOI] [PubMed] [Google Scholar]
- 97.Zhang X, Lu B, Huang X, Xu H, Zhou C, Lin J. Innervation of endometrium and myometrium in women with painful adenomyosis and uterine fibroids. Fertil Steril 2010;94:730–7. [DOI] [PubMed] [Google Scholar]
- 98.Tokushige N, Markham R, Russell P, Fraser IS. Nerve fibres in peritoneal endometriosis. Hum Reprod 2006;21:3001–7. [DOI] [PubMed] [Google Scholar]
- 99.Tejada MA, Antunez C, Nunez-Badinez P, De Leo B, Saunders PT, Vincent K et al. Rodent Animal Models of Endometriosis-Associated Pain: Unmet Needs and Resources Available for Improving Translational Research in Endometriosis. Int J Mol Sci 2023;24. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
The RNA-Seq analysis data generated in this study have been deposited in the NCBI Gene
Expression Omnibus database under accession code GSE240392.