Abstract

Uterine glands and, by inference, their secretions impact uterine receptivity, blastocyst implantation, stromal cell decidualization, and placental development. Changes in gland function across the menstrual cycle are primarily governed by the steroid hormones estrogen (E2) and progesterone (P4) but can also be influenced by extrinsic factors from the stroma. Using a human endometrial epithelial organoid system, transcriptome and proteome analyses identified distinct responses of the organoids to steroid hormones and prostaglandin E2 (PGE2). Notably, P4 and PGE2 modulated the basolateral secretion of organoid proteins, particularly cystatin C (CST3), serpin family A member 3 (SERPINA3), and stanniocalcin 1 (STC1). CST3, but not SERPINA3 or STC1, attenuated the in vitro stromal decidualization response to steroid hormones and PGE2. These findings provide evidence that uterine gland-derived factors impact stromal cell decidualization, which has implications for pregnancy establishment and fertility in women.

Introduction

The inner lining of the uterus, termed the endometrium, is replete with glands that are hypothesized to be essential for the establishment of pregnancy through interactions with the embryo and other endometrial cells (Burton et al., 2002; Hempstock et al., 2004; Filant and Spencer, 2013; Kelleher et al., 2017, 2018). In women, the endometrium must undergo cyclic changes in response to preovulatory and postovulatory steroid hormones to prepare for embryo implantation and pregnancy establishment. Predominantly, hormones from the ovary [estrogen (E2) and progesterone (P4)] co-ordinate the acquisition of endometrial receptivity that involves secretory transformation of the luminal and glandular epithelium (GE), differentiation of the stroma into decidual cells, recruitment of specialized immune cells, and remodeling of the vasculature (Evans et al., 2016). Perturbations in any of those early pregnancy events can lead to adverse ripple effects that compromise pregnancy outcomes (Lédée-Bataille et al., 2002; Klemmt et al., 2006; Al-Sabbagh et al., 2012; Lucas et al., 2016; Peter Durairaj et al., 2017; Fitzgerald et al., 2018).

Differentiation of the endometrial stromal cells into decidual cells (DC) is critical for implantation and pregnancy establishment in women (Cooke et al., 1997; Li et al., 2011; Chen et al., 2013). Stromal cells undergo spontaneous decidualization during the mid-secretory phase of the menstrual cycle in response to rising levels of P4 from the ovary. Progesterone is required for stromal cell decidualization and increased intracellular cAMP hastens that process in vitro (Gellersen and Brosens, 2003). Factors, such as prostaglandin E2 (PGE2) that utilize the cAMP/protein kinase A pathway, can also activate the core gene regulatory network of DCs in concert with P4 (Stadtmauer and Wagner, 2022) and this was previously found to enhance stromal cell decidualization (Frank et al., 1994). PGE2 is produced by both endometrial epithelial and stromal cells (Smith and Kelly, 1988; Alecozay et al., 1991); however, PGE2 regulation of endometrial gland function is not well investigated. Disordered decidualization is linked to reproductive pathologies such as recurrent pregnancy loss (Klemmt et al., 2006; Velarde et al., 2009; Aghajanova et al., 2010; Sherwin et al., 2010; Al-Sabbagh et al., 2012; Macklon and Brosens, 2014; Lucas et al., 2016), implantation failure (Peter Durairaj et al., 2017), and pre-eclampsia (Garrido-Gomez et al., 2017). Uterine glands and their products have long been implicated in uterine receptivity and blastocyst implantation in domestic animals, rodents, and humans (Kelleher et al., 2019c); however, the concept that uterine glands secrete paracrine-acting factors into the stroma that impact decidualization is relatively new and primarily based on studies of glandless mice (Kelleher et al., 2017, 2018). Collective evidence supports the concept that epithelial-stromal crosstalk is essential for uterine function and pregnancy establishment.

During the past few years, 3D endometrial epithelial organoid culture models have been established that recapitulate in vivo epithelial responses to steroid hormones and are viable models to study human uterine gland function (Boretto et al., 2017; Turco et al., 2017; Fitzgerald et al., 2019; Rawlings et al., 2021). The objectives in this study were to: modify the endometrial organoid culture system to remove wingless-type MMTV integration site family, member (WNT)-activating factors determine the effects of steroid hormones and PGE2 on uterine gland function; and test the hypothesis that uterine gland-derived factors impact stromal cell decidualization. These studies provide new evidence that uterine glands secrete proteins in response to P4 and PGE2 that directly impact stromal cell decidualization.

Materials and methods

Establishment and maintenance of human organoid and primary endometrial stromal cultures

All experiments involving human subjects were approved by the Institutional Review Board of the University of Missouri, and written informed consent was obtained from each donor. Donor 1 was 25 years old, undergoing a bilateral tubal ligation, and was menstrual cycle day 7 (mid-proliferative phase). Donor 2 was 24 years old, having a diagnostic laparoscopy, and left salpingectomy and oophorectomy, and was cycle day 13. Donor 3 was 25 years old, undergoing permanent sterilization and cycle day 12. All donors were not taking any steroid-modulating medications.

Upon collection, endometrial tissue biopsies were immediately placed in Dulbecco’s modified Eagle’s medium: nutrient mixture F-12 medium (DMEM/F-12) (Gibco, 11320-033, ThermoFisher Scientific, Waltham, MA, USA) supplemented with 10% fetal bovine serum (FBS) (Sigma-Aldrich, St. Louis, MO, USA) and 1% antibiotic-antimycotic (Anti-Anti, Gibco, 15240-062) at 4°C. Isolation of endometrial epithelial cells and establishment of organoids was performed using previously described methods (Fitzgerald et al., 2019). To collect stromal cells, filtrate from the 100-micron cell strainer was sequentially passed through 40-micron and 10-micron cell strainers. The resulting filtrate was pelleted by centrifugation (3000×g for 5 min at 4°C), resuspended in DMEM/F-12 supplemented with 1% Anti-Anti and 10% charcoal stripped FBS (CSFBS), placed in a flask, and incubated at 37°C. Sequential seeding was performed to ensure the purity of stromal cells in this first flask; the medium from the first flask and any unattached cells were then transferred to a second flask, and medium replaced in the first flask.

Hormone treatment of organoids

To examine the steroid hormone response, organoids were passaged, resuspended in Matrigel, and plated in 12-well plates (three droplets per well). Organoids were established for 4 days in expansion medium (ExM) (Supplementary Table SI). Organoids were then treated with either vehicle as a control (100% ethanol) or 10 nM estradiol (E2, Sigma, E1024) for 2 days in ExM without N-2 supplement (Fig. 1A). Next, organoids were treated with either vehicle control, 10 nM E2, 10 nM E2 and 1 µM medroxyprogesterone acetate (MPA, Sigma, PHR1589) (E2 + MPA), 1 µM MPA, or 1 µM MPA and 1 µM prostaglandin E2 (PGE2, Sigma, P0409) in base medium alone (Supplementary Table SI details base media content) for 6 days with media changed every 2 days. Each treatment was performed in triplicate wells, and organoids derived from three individual donors were used. MPA, a synthetic progestin and progesterone receptor (PGR) agonist, and PGE2 were utilized based on their requirement for, or enhancement of, human stromal cell decidualization in vitro (Frank et al., 1994; Stadtmauer and Wagner, 2022). The rationale for conducting hormone treatments in base media starting on Day 6 is to avoid compromising determination of the basolateral secretome, because the ExM contains proteins [epidermal growth factor (EGF), Noggin, Respondin-1, fibroblast growth factor (FGF)10, hepatocyte growth factor (HGF)] that would interfere with protein analysis by mass spectrometry.

Effects of steroid hormone and PGE2 treatment on human endometrial epithelial organoids. (A) Organoids were seeded in Matrigel and grown for 4 days in expansion media (ExM), followed by treatment with vehicle as a control or estrogen (E2) for 2 days. Next, the ExM was replaced with base media, and organoids were treated for 6 days with either vehicle as a control, E2, E2 + medroxyprogesterone acetate (MPA), MPA alone, or MPA + prostaglandin E2 (PGE2) for 6 days. (B) Gross morphology of organoids on Days 1 and 12. Scale bar = 1000 µm. (C) Immunofluorescent localization of forkhead box A2 (FOXA2), acetylated tubulin (AcTUB), and ectonucleotide pyrophosphatase/phosphodiesterase 3 (ENPP3) proteins in Day 12 organoids. Nuclei were visualized with Hoechst stain. The white arrows denote AcTUB-positive cilia in organoid cells. Scale bar = 75 µm.
Figure 1.

Effects of steroid hormone and PGE2 treatment on human endometrial epithelial organoids. (A) Organoids were seeded in Matrigel and grown for 4 days in expansion media (ExM), followed by treatment with vehicle as a control or estrogen (E2) for 2 days. Next, the ExM was replaced with base media, and organoids were treated for 6 days with either vehicle as a control, E2, E2 + medroxyprogesterone acetate (MPA), MPA alone, or MPA + prostaglandin E2 (PGE2) for 6 days. (B) Gross morphology of organoids on Days 1 and 12. Scale bar = 1000 µm. (C) Immunofluorescent localization of forkhead box A2 (FOXA2), acetylated tubulin (AcTUB), and ectonucleotide pyrophosphatase/phosphodiesterase 3 (ENPP3) proteins in Day 12 organoids. Nuclei were visualized with Hoechst stain. The white arrows denote AcTUB-positive cilia in organoid cells. Scale bar = 75 µm.

Immunofluorescence analyses

Medium was removed and organoids were fixed using zinc formalin for 15 min. Organoids were washed with PBS, embedded in Tissue-Tek O.C.T. Compound (Sakura Finetek USA, Torrance, CA, USA) frozen at −80°C, and sectioned at 7 µm. Sections were air dried for 10 min at 37°C then post-fixed in zinc formalin for 5 min and washed in PBS. Permeabilization was performed with 0.1% Tween 20 (PBST) for 20 min at room temperature and washed using PBS. Antigen retrieval was performed by incubating organoid sections in boiling Reveal Decloaker (RV1000M, Biocare Medica, Pacheco, CA, USA) for 30 min. After cooling for 45 min, organoid sections were briefly washed in Milli-Q H2O, followed by PBST for 20 min and two washes of PBS (10 min each). Sections were blocked with 2% normal goat serum (ThermoFisher Scientific, 01-6201) in PBS for 30 min at room temperature, followed by overnight incubation with primary antibody at 4°C (Supplementary Table SII). Sections were washed twice with PBS (10 min each) and incubated with either Alexa Fluor 488-conjugated secondary antibody (112-545-143, Jackson ImmunoResearch, West Grove, PA, USA) or Alexa Fluor 594-conjugated secondary antibody (111-585-144, Jackson ImmunoResearch) in 2% normal goat serum in PBS for 1 h at room temperature. Sections were then incubated with Hoechst (H3570, Invitrogen) to visualize nuclei and imaged with a Leica DM5500 B upright microscope using the Leica Application Suite X (LAS X).

Real-time quantitative PCR analysis

Total RNA was extracted from stromal cells using a standard TRIzol-based protocol and Direct-zol RNA MiniPrep Plus isolation kit (R2070, Zymo Research, Irvine, CA, USA). To eliminate genomic DNA contamination, isolated RNA was treated with DNase I (Rnase-Free Dnase Set, 79256, Qiagen, Germantown, MD, USA). Total RNA (500 ng) from each sample was reverse transcribed using iScript RT Supermix (1708841, Bio-Rad, Hercules, CA, USA). Real-time PCR was performed using SsoAdvanced Universal SYBR Green Supermix and either Bio-Rad PrimePCR primers or Integrated DNA Technologies primers (Supplementary Table SIII). HPRT1 (hypoxanthine phosphoribosyltransferase 1) and RPLP0 (ribosomal protein lateral stalk subunit P0) were used as housekeeping genes for normalization of data. A one-way ANOVA was used to compare mRNA levels between treatment groups with a P-value of ≤0.05 considered significant.

RNA-seq transcriptome analysis

Quality and concentration of RNA extracted from hormone-treated organoids from each donor were determined using a Fragment Analyzer (Advanced Analytical Technologies, Ankeny, IA, USA). Libraries were prepared by the University of Missouri Genomics Technology Core Facility using an Illumina TruSeq mRNA kit (Illumina, San Diego, CA, USA) and sequenced (2 × 100 bp paired-end to a depth of 40–50 million reads) using an Illumina NextSeq 500. FastQC (v 0.11.9) and MultiQC (v 1.10.1) were used to evaluate the quality of the sequence. Trimmomatic (v 1.10.1) was used to remove adapters and low-quality bases from the reads. Trimmed reads were aligned to the human genome (GRCh38) with STAR aligner (v 2.7.9a) (average unique mapping rate of 89.21% and read length of 200 bp). Gene quantification was calculated by using featureCounts from Subread package (v 2.0.3). Counts extracted from featureCounts were used to perform differential gene analysis in R (version 4.0.2) using DESeq2 (v 1.30.1). ClusterProfiler software (version 4.0.5) was used to perform Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) Pathway analysis on differentially expressed genes (DEGs) with absolute log2 fold change >1 and adjusted P-value ≤ 0.05. Design formula (donor – condition) was used as a factor to account for variation in the data. Increased and decreased genes were each subjected to a separate KEGG pathway analysis. Transcriptome data are available in the Gene Expression Omnibus (GSE205084).

Mass spectrometry analysis of basolateral secretion of organoid proteins

Mass spectrometry analysis was conducted by the Charles W. Gehrke Proteomics Center at the University of Missouri. To analyze the secreted proteome, media were collected from four wells per treatment group and pooled (three donors per treatment). Samples were mixed with four volumes of 5% trichloroacetic acid in acetone and incubated at −20°C overnight. Following centrifugation (20 000×g for 10 min at 4°C), samples were washed twice with 80% acetone and pellets were resuspended in 6 M urea, 2 M thiourea, and 100 mM ammonium bicarbonate. Protein was quantified using the Pierce 660 nM Protein Assay method (Pierce Chemical, Dallas, TX, USA). Thirty micrograms of protein from each sample was reduced and alkylated, digested with lysC at an enzyme to protein ratio of 1:70 and incubated for 3 h at 37°C. Trypsin was added to the samples at a trypsin to protein w/w ratio of 1:50 and the samples were digested overnight at 37°C. Digested peptides were purified using Pierce C18 tips. To generate a spectra library, 5 µg purified peptides from each sample were combined, lyophilized, and resuspended in 5% acetonitrile, 0.1% formic acid. Samples were acquired on the Bruker timsTOFPRO with the DIA acquisition method using the 30 min liquid chromatography gradient. The spectral library was built by using data-dependent acquisition data by Pulsar in Spectronaut, filtered with a PSM/peptide/protein false discovery rate (FDR) of 1%. The Uniprot-Human database was used (20 381 items). Data were searched with tryspin as enzyme, two missed cleavages allowed; carbamidomethyl cysteine as a fixed modification; acetylation on protein N-terminus, oxidation (M) as variable modifications. Data independent acquisition (DIA) data were searched using Spectronaut with the spectra library generated by Pulsar. The precursor/protein Q value cutoff was 0.01. Quantitation type of MS2 area was chosen and data filtering was set to Q value sparse. The DIA data were filtered with precursor FDR and protein group at 1%. A protein was considered present if identified in two samples per treatment group and a protein intensity of >6 in at least one sample. One-way ANOVA with a post hoc Tukey’s honestly significant difference test was used to compare treatment groups. A P-value of ≤0.05 was considered significant. The data were deposited in the proteomics identifications database (PRIDE, www.ebi.ac.uk/pride/ submission reference 1-20220602-7305).

Culture and treatment of primary human endometrial stromal cells

Primary endometrial stromal cells were grown to 80% confluence in 10% CSFBS DMEM/F-12 and then treated with either vehicle as a control or 10 nM E2 and 1 µM MPA (E2 + MPA) in 0.5% CSFBS DMEM/F-12 for 6 days. Next, stromal cells were treated with either vehicle control, E2 and MPA, E2 + MPA + 0.5 mM cAMP (2′-O-dibutyryladenosine 3′, 5′-cyclic monophosphate sodium salt; Sigma, D0627), E2 + MPA + 1 µM PGE2 (Prostaglandin E2, Sigma, P0409), and E2 + MPA with either cystatin-C (CST3, 1196-PI-010, R&D Systems, Minneapolis, MN, USA), stanniocalcin-1 (STC1, R&D Systems, 9400-SO-050), or alpha-1-antichymotrypsin (SERPINA3, R&D Systems, 1295-PI-010) at 10, 100, and 1000 ng/ml for an additional 4 days (three wells/treatment). To determine whether CST3 influences PGE2-induced stromal cell decidualization, primary endometrial stromal cells were treated with either vehicle control or E2 + MPA in 0.5% CSFBS DMEM/F-12 for 6 days. Stromal cells were then treated with either vehicle control, E2 + MPA, E2 + MPA + PGE2, or E2 + MPA + PGE2 and CST3 at 10, 100, and 1000 ng/ml for an additional 4 days (three wells/treatment).

Results

Development of human organoids in base media without WNT-activating factors

During the 12-day experiment (Fig. 1A), all human endometrial epithelial organoids grew substantially larger in diameter, with no obvious differences in growth or morphology between the treatment groups (Fig. 1B). Forkhead box A2 (FOXA2) is a transcription factor that is expressed specifically in the GE of the endometrium of the human uterus (Kelleher et al., 2019a). The immunofluorescence signal intensity for FOXA2 was similar in the nuclei of most cells in the organoid regardless of hormone treatment (Fig. 1C). The organoids contained a mixed population of GE cells, including non-ciliated and ciliated cells based on immunofluorescence analysis of acetylated tubulin (AcTUB), a marker of ciliated cells (Fig. 1C). No differences in growth, FOXA2 or ciliated cells were observed among organoids from the three donors (data not shown).

Steroid hormones and PGE2 alter the organoid transcriptome

Bulk RNA-seq analysis of hormone-treated organoids derived from three donors revealed significant changes in their transcriptomes in response to hormone treatment (Fig. 2A, Supplementary Data SI). Changes in mRNA abundance between control and hormone treatment groups were assessed using several parameters (log2 fold change ≥ 1.0, P 0.05, and fragments per kilobase of exon per million mapped fragments ≥ 1.0 in one group). The effects of hormone treatment and PGE2 on the organoid transcriptome are summarized in Fig. 2.

Transcriptome analysis of hormone- and PGE2-treated human endometrial epithelial organoids. Organoids were seeded in Matrigel and grown for 4 days in expansion media (ExM), followed by treatment with vehicle as a control or estrogen (E2) for 2 days. Next, the ExM was replaced with base media, and organoids were treated for 6 days with either vehicle as a control, E2, E2 + medroxyprogesterone acetate (MPA), MPA alone, or MPA + prostaglandin E2 (PGE2) for 6 days. On Day 12, RNA was extracted, and the transcriptome determined by RNA sequencing. (A) Volcano plots of differentially expressed genes based on organoid response to hormone treatments. (B) Venn diagrams comparing genes that were differentially increased or decreased in all treatments compared to control. (C) Venn diagrams comparing genes that were differentially increased or decreased in E2 and E2 + MPA treatments compared to control. (D) Venn diagrams comparing genes that were differentially increased or decreased in MPA and MPA + PGE2 treatments compared to control.
Figure 2.

Transcriptome analysis of hormone- and PGE2-treated human endometrial epithelial organoids. Organoids were seeded in Matrigel and grown for 4 days in expansion media (ExM), followed by treatment with vehicle as a control or estrogen (E2) for 2 days. Next, the ExM was replaced with base media, and organoids were treated for 6 days with either vehicle as a control, E2, E2 + medroxyprogesterone acetate (MPA), MPA alone, or MPA + prostaglandin E2 (PGE2) for 6 days. On Day 12, RNA was extracted, and the transcriptome determined by RNA sequencing. (A) Volcano plots of differentially expressed genes based on organoid response to hormone treatments. (B) Venn diagrams comparing genes that were differentially increased or decreased in all treatments compared to control. (C) Venn diagrams comparing genes that were differentially increased or decreased in E2 and E2 + MPA treatments compared to control. (D) Venn diagrams comparing genes that were differentially increased or decreased in MPA and MPA + PGE2 treatments compared to control.

COL1A2 (collagen type I alpha 2 Chain) showed the greatest increase in mRNA level, and PGR was among the top 10 genes impacted by E2. Treatment with MPA alone or in combination with E2 or PGE2 altered expression of the greatest number of genes compared to the control (Fig. 2A and B). ENPP3 (ectonucleotide pyrophosphatase/phosphodiesterase family member 3) and LEFTY1 (left-right determination factor 1) were among the top 10 genes stimulated by MPA compared to control. Treatment with MPA + PGE2 compared to the control or MPA alone revealed several PGE2-regulated genes, particularly an increase in metallothionein genes (MT1G, MT1H, MT1M). MMP10 (matrix meallopeptidase 10) showed the greatest decrease in expression in E2 + MPA and MPA treatments compared to control.

GO enrichment analysis of DEGs in hormone-treated organoids found enrichment (P <0.05) for specific biological processes based on hormone treatment (Supplementary Fig. S1). Biological processes enriched by E2 treatment of organoids included nucleus localization, and cilia assembly and movement. Genes decreased by E2 treatment were enriched for biological processes related to immune and stress responses. Genes increased by E2 + MPA treatment compared to control were also enriched for cilium assembly and movement as well as extracellular transport. Indeed, an increase in ciliated cells in the organoids was observed with E2 and MPA treatment (Fig. 1C). Processes decreased by E2 + MPA compared to control included angiogenesis and tube morphogenesis. Biological processes enriched in genes increased by MPA + PGE2 treatment included responses to and regulation of metal ions copper and zinc. Genes decreased by MPA + PGE2 were enriched for cell motility, migration, and locomotion. No biological processes were identified as enriched by treatment with MPA alone.

ENPP3 was one of the top five genes increased by MPA, E2 + MPA, and MPA + PGE2, indicating P4 responsivity (Fig. 2A–C). Immunostaining revealed that ENPP3 was substantially increased in the organoids by treatment with MPA, MPA + E2, or MPA + PGE2 as compared to E2 alone or the control (Fig. 1C). ENPP3 was observed at the apical surface of the organoid cells, which is reflective of the in vivo secretion pattern of ENPP3 and presence in the uterine lumen (Boggavarapu et al., 2016; Chen et al., 2018).

Identification of proteins secreted by the organoids

The secreted proteome of organoids treated with hormones and PGE2 was determined by mass spectrometry. This analysis identified ∼124 proteins in the basolateral organoid secretome (Supplementary Data SII). Biological processes enriched for all proteins identified in the organoid secretome included extracellular matrix organization, regulation of peptidase activity, wound healing, secretion, and immune response (Fig. 3A).

Mass spectrometry analysis of proteins secreted basolaterally by hormone- and PGE2-treated human endometrial epithelial organoids. Organoids were seeded in Matrigel and grown for 4 days in expansion media, followed by treatment with vehicle as a control or estrogen (E2) for 2 days. Next, the ExM was replaced with base media, and organoids were treated for 6 days with either vehicle as a control, E2, E2 + medroxyprogesterone acetate (MPA), MPA alone, or MPA + prostaglandin E2 (PGE2) for 6 days. On Day 12, proteins in the base media surrounding the organoid were determined by mass spectrometry analysis. (A) ShinyGO visualization of enriched biological processes for genes identified in organoid conditioned media. (B) Integration of proteins that were increased or decreased for all treatments compared to control. (C) REVIGO visualization of enriched biological processes for proteins increased or decreased by E2 and E2 + MPA treatment compared to control.
Figure 3.

Mass spectrometry analysis of proteins secreted basolaterally by hormone- and PGE2-treated human endometrial epithelial organoids. Organoids were seeded in Matrigel and grown for 4 days in expansion media, followed by treatment with vehicle as a control or estrogen (E2) for 2 days. Next, the ExM was replaced with base media, and organoids were treated for 6 days with either vehicle as a control, E2, E2 + medroxyprogesterone acetate (MPA), MPA alone, or MPA + prostaglandin E2 (PGE2) for 6 days. On Day 12, proteins in the base media surrounding the organoid were determined by mass spectrometry analysis. (A) ShinyGO visualization of enriched biological processes for genes identified in organoid conditioned media. (B) Integration of proteins that were increased or decreased for all treatments compared to control. (C) REVIGO visualization of enriched biological processes for proteins increased or decreased by E2 and E2 + MPA treatment compared to control.

Changes in secreted protein abundance between control and hormone treatment groups were assessed, according to statistical differences (P <0.05) and fold change (≥1.5-fold) between the control and treatment groups (Fig. 3B, Tables I, II, III, and IV, Supplementary Data SII). Compared to the control, E2 alone increased 22 proteins including SERPINA3, COL1A2, and CST3, and decreased 43 proteins including ADAMTS1, PROM1, and MMP10. Treatment with E2 + MPA increased 23 proteins including COL1A2 and SERPINA3, and decreased 39 proteins including IGFBP3, ADAMTS1, and MMP10. MPA alone increased 18 proteins including CST3, COL1A2, and SERPINA3, and decreased 33 proteins including APOA1, ADAMTS1, and MMP10. Finally, treatment with MPA + PGE2 increased the abundance of 19 proteins including CST3, SERPINA3, and B2M, and decreased 71 proteins including ADAMTS1, APOA2, and FN1. Of note, five proteins were uniquely increased by E2 + MPA treatment, while all other increased proteins were common amongst two or more treatment groups (Fig. 3B). Moreover, 27 proteins were uniquely decreased by MPA + PGE2 treatment, while all other decreased proteins were common among two or more treatment groups (Fig. 3B). Integration of the organoid transcriptome and basolateral secreted proteome data is presented in Supplementary Fig. S2. Note that relatively few genes and encoded proteins were coordinately increased or decreased by hormone treatment. One caveat of the present study is that the intraorganoid, apical secretome (i.e. proteins present within the lumen of the organoid itself) was not determined. Indeed, substantial differences exist in the apical (intraorganoid) as compared to basolateral (extraorganoid) secretome (Simintiras et al., 2021).

Table I

Top 10 proteins in the human organoid secretome responding to E2 treatment.a

Increasedb
Decreasedb
SymbolNameFold changeSymbolNameFold change
SERPINA3Alpha-1-antichymotrypsin9.1MMP10Stromelysin-2−6.5
COL1A2Collagen alpha-2(I) chain5.5ADAMTS1A disintegrin and metalloproteinase with thrombospondin motifs 1−4.6
B2MBeta-2-microglobulin4.9PROM1Prominin-1−3.6
TFPI2Tissue factor pathway inhibitor 24.0CLIC1Chloride intracellular channel protein 1−3.3
CST3Cystatin-C3.3APOA1Apolipoprotein A-I−2.8
ALPPAlkaline phosphatase, placental type2.5SERPINF2Alpha-2-antiplasmin−2.8
RBP4Retinol-binding protein 42.3IGFBP3Insulin-like growth factor-binding protein 3−2.5
SERPINA5Plasma serine protease inhibitor2.1HABP2Hyaluronan-binding protein 2−2.4
LEFTY2Left-right determination factor 22.1CPXM2Inactive carboxypeptidase-like protein X2−2.3
SEMA3BSemaphorin-3B2.1CFBComplement factor B−2.3
Increasedb
Decreasedb
SymbolNameFold changeSymbolNameFold change
SERPINA3Alpha-1-antichymotrypsin9.1MMP10Stromelysin-2−6.5
COL1A2Collagen alpha-2(I) chain5.5ADAMTS1A disintegrin and metalloproteinase with thrombospondin motifs 1−4.6
B2MBeta-2-microglobulin4.9PROM1Prominin-1−3.6
TFPI2Tissue factor pathway inhibitor 24.0CLIC1Chloride intracellular channel protein 1−3.3
CST3Cystatin-C3.3APOA1Apolipoprotein A-I−2.8
ALPPAlkaline phosphatase, placental type2.5SERPINF2Alpha-2-antiplasmin−2.8
RBP4Retinol-binding protein 42.3IGFBP3Insulin-like growth factor-binding protein 3−2.5
SERPINA5Plasma serine protease inhibitor2.1HABP2Hyaluronan-binding protein 2−2.4
LEFTY2Left-right determination factor 22.1CPXM2Inactive carboxypeptidase-like protein X2−2.3
SEMA3BSemaphorin-3B2.1CFBComplement factor B−2.3
a

E2 alone versus control.

b

Differences (P ≤ 0.05) between the estrogen (E2) alone and control treatment groups was determined by one-way ANOVA with a post hoc Tukey’s honestly significant difference test.

Table I

Top 10 proteins in the human organoid secretome responding to E2 treatment.a

Increasedb
Decreasedb
SymbolNameFold changeSymbolNameFold change
SERPINA3Alpha-1-antichymotrypsin9.1MMP10Stromelysin-2−6.5
COL1A2Collagen alpha-2(I) chain5.5ADAMTS1A disintegrin and metalloproteinase with thrombospondin motifs 1−4.6
B2MBeta-2-microglobulin4.9PROM1Prominin-1−3.6
TFPI2Tissue factor pathway inhibitor 24.0CLIC1Chloride intracellular channel protein 1−3.3
CST3Cystatin-C3.3APOA1Apolipoprotein A-I−2.8
ALPPAlkaline phosphatase, placental type2.5SERPINF2Alpha-2-antiplasmin−2.8
RBP4Retinol-binding protein 42.3IGFBP3Insulin-like growth factor-binding protein 3−2.5
SERPINA5Plasma serine protease inhibitor2.1HABP2Hyaluronan-binding protein 2−2.4
LEFTY2Left-right determination factor 22.1CPXM2Inactive carboxypeptidase-like protein X2−2.3
SEMA3BSemaphorin-3B2.1CFBComplement factor B−2.3
Increasedb
Decreasedb
SymbolNameFold changeSymbolNameFold change
SERPINA3Alpha-1-antichymotrypsin9.1MMP10Stromelysin-2−6.5
COL1A2Collagen alpha-2(I) chain5.5ADAMTS1A disintegrin and metalloproteinase with thrombospondin motifs 1−4.6
B2MBeta-2-microglobulin4.9PROM1Prominin-1−3.6
TFPI2Tissue factor pathway inhibitor 24.0CLIC1Chloride intracellular channel protein 1−3.3
CST3Cystatin-C3.3APOA1Apolipoprotein A-I−2.8
ALPPAlkaline phosphatase, placental type2.5SERPINF2Alpha-2-antiplasmin−2.8
RBP4Retinol-binding protein 42.3IGFBP3Insulin-like growth factor-binding protein 3−2.5
SERPINA5Plasma serine protease inhibitor2.1HABP2Hyaluronan-binding protein 2−2.4
LEFTY2Left-right determination factor 22.1CPXM2Inactive carboxypeptidase-like protein X2−2.3
SEMA3BSemaphorin-3B2.1CFBComplement factor B−2.3
a

E2 alone versus control.

b

Differences (P ≤ 0.05) between the estrogen (E2) alone and control treatment groups was determined by one-way ANOVA with a post hoc Tukey’s honestly significant difference test.

Table II

Top 10 proteins in the human organoid secretome responding to treatment with E2+MPA.a

Increasedb
Decreasedb
SymbolNameFold changeSymbolNameFold change
SERPINA3Alpha-1-antichymotrypsin17.2MMP10Stromelysin-2−14.8
COL1A2Collagen alpha-2(I) chain10.4ADAMTS1A disintegrin and metalloproteinase with thrombospondin motifs 1−6.7
TFPI2Tissue factor pathway inhibitor 25.6IGFBP3Insulin-like growth factor-binding protein 3−4.5
B2MBeta-2-microglobulin3.7PROM1Prominin-1−4.2
LEFTY2Left-right determination factor 23.4SERPINF2Alpha-2-antiplasmin−4.1
SERPINA5Plasma serine protease inhibitor3.2APOA1Apolipoprotein A-I−3.7
MIFMacrophage migration inhibitory factor2.7HABP2Hyaluronan-binding protein 2−2.9
ALPPAlkaline phosphatase, placental type2.6CPXM2Inactive carboxypeptidase-like protein X2−2.7
CST3Cystatin-C2.5MUC5BMucin-5B−2.5
SEMA3BSemaphorin-3B2.4CFBComplement factor B−2.4
Increasedb
Decreasedb
SymbolNameFold changeSymbolNameFold change
SERPINA3Alpha-1-antichymotrypsin17.2MMP10Stromelysin-2−14.8
COL1A2Collagen alpha-2(I) chain10.4ADAMTS1A disintegrin and metalloproteinase with thrombospondin motifs 1−6.7
TFPI2Tissue factor pathway inhibitor 25.6IGFBP3Insulin-like growth factor-binding protein 3−4.5
B2MBeta-2-microglobulin3.7PROM1Prominin-1−4.2
LEFTY2Left-right determination factor 23.4SERPINF2Alpha-2-antiplasmin−4.1
SERPINA5Plasma serine protease inhibitor3.2APOA1Apolipoprotein A-I−3.7
MIFMacrophage migration inhibitory factor2.7HABP2Hyaluronan-binding protein 2−2.9
ALPPAlkaline phosphatase, placental type2.6CPXM2Inactive carboxypeptidase-like protein X2−2.7
CST3Cystatin-C2.5MUC5BMucin-5B−2.5
SEMA3BSemaphorin-3B2.4CFBComplement factor B−2.4
a

E2 + MPA (estrogen + medroxyprogesterone acetate) alone versus control.

b

Differences (P ≤ 0.05) between the E2 + MPA and control treatment groups was determined by one-way ANOVA with a post hoc Tukey’s honestly significant difference test.

Table II

Top 10 proteins in the human organoid secretome responding to treatment with E2+MPA.a

Increasedb
Decreasedb
SymbolNameFold changeSymbolNameFold change
SERPINA3Alpha-1-antichymotrypsin17.2MMP10Stromelysin-2−14.8
COL1A2Collagen alpha-2(I) chain10.4ADAMTS1A disintegrin and metalloproteinase with thrombospondin motifs 1−6.7
TFPI2Tissue factor pathway inhibitor 25.6IGFBP3Insulin-like growth factor-binding protein 3−4.5
B2MBeta-2-microglobulin3.7PROM1Prominin-1−4.2
LEFTY2Left-right determination factor 23.4SERPINF2Alpha-2-antiplasmin−4.1
SERPINA5Plasma serine protease inhibitor3.2APOA1Apolipoprotein A-I−3.7
MIFMacrophage migration inhibitory factor2.7HABP2Hyaluronan-binding protein 2−2.9
ALPPAlkaline phosphatase, placental type2.6CPXM2Inactive carboxypeptidase-like protein X2−2.7
CST3Cystatin-C2.5MUC5BMucin-5B−2.5
SEMA3BSemaphorin-3B2.4CFBComplement factor B−2.4
Increasedb
Decreasedb
SymbolNameFold changeSymbolNameFold change
SERPINA3Alpha-1-antichymotrypsin17.2MMP10Stromelysin-2−14.8
COL1A2Collagen alpha-2(I) chain10.4ADAMTS1A disintegrin and metalloproteinase with thrombospondin motifs 1−6.7
TFPI2Tissue factor pathway inhibitor 25.6IGFBP3Insulin-like growth factor-binding protein 3−4.5
B2MBeta-2-microglobulin3.7PROM1Prominin-1−4.2
LEFTY2Left-right determination factor 23.4SERPINF2Alpha-2-antiplasmin−4.1
SERPINA5Plasma serine protease inhibitor3.2APOA1Apolipoprotein A-I−3.7
MIFMacrophage migration inhibitory factor2.7HABP2Hyaluronan-binding protein 2−2.9
ALPPAlkaline phosphatase, placental type2.6CPXM2Inactive carboxypeptidase-like protein X2−2.7
CST3Cystatin-C2.5MUC5BMucin-5B−2.5
SEMA3BSemaphorin-3B2.4CFBComplement factor B−2.4
a

E2 + MPA (estrogen + medroxyprogesterone acetate) alone versus control.

b

Differences (P ≤ 0.05) between the E2 + MPA and control treatment groups was determined by one-way ANOVA with a post hoc Tukey’s honestly significant difference test.

Table III

Top 10 proteins in the human organoid secretome responding to treatment with MPA alone.a

Increasedb
Decreasedb
SymbolNameFold changeSymbolNameFold change
SERPINA3Alpha-1-antichymotrypsin15.7MMP10Stromelysin-2−5.7
COL1A2Collagen alpha-2(I) chain6.7ADAMTS1A disintegrin and metalloproteinase with thrombospondin motifs 1−4.3
B2MBeta-2-microglobulin4.5APOA1Apolipoprotein A-I−3.6
ALPPAlkaline phosphatase, placental type4.2PROM1Prominin-1−2.8
TFPI2Tissue factor pathway inhibitor 23.9FN1Fibronectin−2.6
SERPINA5Plasma serine protease inhibitor3.3CLIC1Chloride intracellular channel protein 1−2.4
CST3Cystatin-C3.1CPXM2Inactive carboxypeptidase-like protein X2−2.3
LEFTY2Left-right determination factor 22.8TIMP1Metalloproteinase inhibitor 1−2.2
LTFLactotransferrin2.5IGFBP3Insulin-like growth factor-binding protein 3−2.2
RBP4Retinol-binding protein 42.1NPC2NPC intracellular cholesterol transporter 2−2.2
Increasedb
Decreasedb
SymbolNameFold changeSymbolNameFold change
SERPINA3Alpha-1-antichymotrypsin15.7MMP10Stromelysin-2−5.7
COL1A2Collagen alpha-2(I) chain6.7ADAMTS1A disintegrin and metalloproteinase with thrombospondin motifs 1−4.3
B2MBeta-2-microglobulin4.5APOA1Apolipoprotein A-I−3.6
ALPPAlkaline phosphatase, placental type4.2PROM1Prominin-1−2.8
TFPI2Tissue factor pathway inhibitor 23.9FN1Fibronectin−2.6
SERPINA5Plasma serine protease inhibitor3.3CLIC1Chloride intracellular channel protein 1−2.4
CST3Cystatin-C3.1CPXM2Inactive carboxypeptidase-like protein X2−2.3
LEFTY2Left-right determination factor 22.8TIMP1Metalloproteinase inhibitor 1−2.2
LTFLactotransferrin2.5IGFBP3Insulin-like growth factor-binding protein 3−2.2
RBP4Retinol-binding protein 42.1NPC2NPC intracellular cholesterol transporter 2−2.2
a

MPA (medroxyprogesterone acetate) alone versus control.

b

Differences (P ≤ 0.05) between the MPA and control treatment groups was determined by one-way ANOVA with a post hoc Tukey’s honestly significant difference test.

Table III

Top 10 proteins in the human organoid secretome responding to treatment with MPA alone.a

Increasedb
Decreasedb
SymbolNameFold changeSymbolNameFold change
SERPINA3Alpha-1-antichymotrypsin15.7MMP10Stromelysin-2−5.7
COL1A2Collagen alpha-2(I) chain6.7ADAMTS1A disintegrin and metalloproteinase with thrombospondin motifs 1−4.3
B2MBeta-2-microglobulin4.5APOA1Apolipoprotein A-I−3.6
ALPPAlkaline phosphatase, placental type4.2PROM1Prominin-1−2.8
TFPI2Tissue factor pathway inhibitor 23.9FN1Fibronectin−2.6
SERPINA5Plasma serine protease inhibitor3.3CLIC1Chloride intracellular channel protein 1−2.4
CST3Cystatin-C3.1CPXM2Inactive carboxypeptidase-like protein X2−2.3
LEFTY2Left-right determination factor 22.8TIMP1Metalloproteinase inhibitor 1−2.2
LTFLactotransferrin2.5IGFBP3Insulin-like growth factor-binding protein 3−2.2
RBP4Retinol-binding protein 42.1NPC2NPC intracellular cholesterol transporter 2−2.2
Increasedb
Decreasedb
SymbolNameFold changeSymbolNameFold change
SERPINA3Alpha-1-antichymotrypsin15.7MMP10Stromelysin-2−5.7
COL1A2Collagen alpha-2(I) chain6.7ADAMTS1A disintegrin and metalloproteinase with thrombospondin motifs 1−4.3
B2MBeta-2-microglobulin4.5APOA1Apolipoprotein A-I−3.6
ALPPAlkaline phosphatase, placental type4.2PROM1Prominin-1−2.8
TFPI2Tissue factor pathway inhibitor 23.9FN1Fibronectin−2.6
SERPINA5Plasma serine protease inhibitor3.3CLIC1Chloride intracellular channel protein 1−2.4
CST3Cystatin-C3.1CPXM2Inactive carboxypeptidase-like protein X2−2.3
LEFTY2Left-right determination factor 22.8TIMP1Metalloproteinase inhibitor 1−2.2
LTFLactotransferrin2.5IGFBP3Insulin-like growth factor-binding protein 3−2.2
RBP4Retinol-binding protein 42.1NPC2NPC intracellular cholesterol transporter 2−2.2
a

MPA (medroxyprogesterone acetate) alone versus control.

b

Differences (P ≤ 0.05) between the MPA and control treatment groups was determined by one-way ANOVA with a post hoc Tukey’s honestly significant difference test.

Table IV

Top 10 proteins in the human organoid secretome responding to treatment with MPA+PGE2.a

Increasedb
Decreasedb
SymbolNameFold changeSymbolNameFold change
B2MBeta-2-microglobulin7.0KRT8Keratin, type II cytoskeletal 8−67.5
SERPINA3Alpha-1-antichymotrypsin5.9FN1Fibronectin−4.8
CST3Cystatin-C5.0ITIH5Inter-alpha-trypsin inhibitor heavy chain H5−4.4
RBP4Retinol-binding protein 42.8APOA1Apolipoprotein A-I−3.8
COL1A2Collagen alpha-2(I) chain2.6ADAMTS1A disintegrin and metalloproteinase with thrombospondin motifs 1−3.4
LTFLactotransferrin2.3B4GALT1Beta-1,4-galactosyltransferase 1−3.3
APOC3Apolipoprotein C-III2.3MMP10Stromelysin-2−3.3
SULT1C2Sulfotransferase 1C22.3CLIC1Chloride intracellular channel protein 1−3.1
SERPINA5Plasma serine protease inhibitor2.2PROM1Prominin-1−3.1
ALPPAlkaline phosphatase, placental type2.1NPC2NPC intracellular cholesterol transporter 2−3.0
Increasedb
Decreasedb
SymbolNameFold changeSymbolNameFold change
B2MBeta-2-microglobulin7.0KRT8Keratin, type II cytoskeletal 8−67.5
SERPINA3Alpha-1-antichymotrypsin5.9FN1Fibronectin−4.8
CST3Cystatin-C5.0ITIH5Inter-alpha-trypsin inhibitor heavy chain H5−4.4
RBP4Retinol-binding protein 42.8APOA1Apolipoprotein A-I−3.8
COL1A2Collagen alpha-2(I) chain2.6ADAMTS1A disintegrin and metalloproteinase with thrombospondin motifs 1−3.4
LTFLactotransferrin2.3B4GALT1Beta-1,4-galactosyltransferase 1−3.3
APOC3Apolipoprotein C-III2.3MMP10Stromelysin-2−3.3
SULT1C2Sulfotransferase 1C22.3CLIC1Chloride intracellular channel protein 1−3.1
SERPINA5Plasma serine protease inhibitor2.2PROM1Prominin-1−3.1
ALPPAlkaline phosphatase, placental type2.1NPC2NPC intracellular cholesterol transporter 2−3.0
a

MPA + PGE2 (medroxyprogesterone acetate + prostaglandin E2) versus control.

b

Differences (P ≤ 0.05) between the MPA+PGE2 and control treatment groups was determined by one-way ANOVA with a post hoc Tukey’s honestly significant difference test.

Table IV

Top 10 proteins in the human organoid secretome responding to treatment with MPA+PGE2.a

Increasedb
Decreasedb
SymbolNameFold changeSymbolNameFold change
B2MBeta-2-microglobulin7.0KRT8Keratin, type II cytoskeletal 8−67.5
SERPINA3Alpha-1-antichymotrypsin5.9FN1Fibronectin−4.8
CST3Cystatin-C5.0ITIH5Inter-alpha-trypsin inhibitor heavy chain H5−4.4
RBP4Retinol-binding protein 42.8APOA1Apolipoprotein A-I−3.8
COL1A2Collagen alpha-2(I) chain2.6ADAMTS1A disintegrin and metalloproteinase with thrombospondin motifs 1−3.4
LTFLactotransferrin2.3B4GALT1Beta-1,4-galactosyltransferase 1−3.3
APOC3Apolipoprotein C-III2.3MMP10Stromelysin-2−3.3
SULT1C2Sulfotransferase 1C22.3CLIC1Chloride intracellular channel protein 1−3.1
SERPINA5Plasma serine protease inhibitor2.2PROM1Prominin-1−3.1
ALPPAlkaline phosphatase, placental type2.1NPC2NPC intracellular cholesterol transporter 2−3.0
Increasedb
Decreasedb
SymbolNameFold changeSymbolNameFold change
B2MBeta-2-microglobulin7.0KRT8Keratin, type II cytoskeletal 8−67.5
SERPINA3Alpha-1-antichymotrypsin5.9FN1Fibronectin−4.8
CST3Cystatin-C5.0ITIH5Inter-alpha-trypsin inhibitor heavy chain H5−4.4
RBP4Retinol-binding protein 42.8APOA1Apolipoprotein A-I−3.8
COL1A2Collagen alpha-2(I) chain2.6ADAMTS1A disintegrin and metalloproteinase with thrombospondin motifs 1−3.4
LTFLactotransferrin2.3B4GALT1Beta-1,4-galactosyltransferase 1−3.3
APOC3Apolipoprotein C-III2.3MMP10Stromelysin-2−3.3
SULT1C2Sulfotransferase 1C22.3CLIC1Chloride intracellular channel protein 1−3.1
SERPINA5Plasma serine protease inhibitor2.2PROM1Prominin-1−3.1
ALPPAlkaline phosphatase, placental type2.1NPC2NPC intracellular cholesterol transporter 2−3.0
a

MPA + PGE2 (medroxyprogesterone acetate + prostaglandin E2) versus control.

b

Differences (P ≤ 0.05) between the MPA+PGE2 and control treatment groups was determined by one-way ANOVA with a post hoc Tukey’s honestly significant difference test.

Several biological processes were enriched in secreted proteins that increased in response to E2 alone, including acute inflammatory response, secretion, cell proliferation, and regulation of proteolysis (Fig. 3C). Similarly, secreted proteins increased by E2 + MPA treatment were enriched for biological processes such as acute inflammatory response, regulation of hydrolase activity, neutrophil activation, and negative regulation of developmental processes. Collectively, these results support the idea that E2, MPA, and PGE2 have distinct and combinatorial effects on proteins secreted from the basolateral aspect of the organoid, with potential paracrine effects on surrounding endometrial stromal cells.

Impact of selected organoid secreted proteins on stromal cell decidualization

To determine the impact of organoid secreted proteins on stromal cell decidualization (Fig. 4A), three proteins were selected (CST3, STC1, SERPINA3) as they were increased in the media of organoids treated with MPA, MPA + E2, and MPA + PGE2 as compared to the control (Fig. 4, Tables I, II, III, and IV, Supplementary Data SII).

Effects of PGE2 and proteins secreted by human endometrial epithelial organoid on stromal cell decidualization. (A) Primary endometrial stromal cells were seeded, grown to 80% confluence in 10% charcoal stripped FBS (CSFBS) for 2 days, changed to 0.5% CSFBS media, and then treated with either vehicle as a control or estrogen and medroxyprogesterone acetate (E2 + MPA) for 6 days. Next, stromal cells were treated with vehicle as a control (CTRL), and E2 + MPA, E2 + MPA with cyclic adenosine monophosphate (E2 + MPA + cAMP), E2 + MPA with prostaglandin E2 (E2 + MPA + PGE2), or E2 + MPA with increasing amounts (10, 100, or 1000 ng/ml) of recombinant cystatin C (CST3), stanniocalcin 1 (STC1), or serpin family A member 3 (SERPINA3) for 4 days. On Day 10, RNA was harvested from cells, and mRNA levels assessed by quantitative RT-PCR. Each treatment was performed in triplicate, and the entire study contained three biological replicates. (B) Established markers of stromal cell decidualization including prolactin (PRL), insulin-like growth factor binding protein 1 (IGFBP1), interleukin 15 (IL15), and somatostatin (SMST). Solid dots show the means for the individual biological replicates. (C) Markers of active decidual cells including crystallin alpha B (CRYAB), hydroxysteroid 11-beta dehydrogenase 1 (HSD11B1), and scavenger receptor class A member 5 (SCARA5). (D) Markers of senescent decidual cells including ABI family member 3 binding protein (ABI3BP), clusterin (CLU), and iodothyronine deiodinase 2 (DIO2). Data are presented as mean ± SEM for each of the n = 3 biological replicates. One-way ANOVA was performed to compare treatment of cells with cAMP, PGE2 or recombinant proteins to E2 + MPA only. Significant effects of treatment (P ≤ 0.05) are denoted by an asterisk (*).
Figure 4.

Effects of PGE2 and proteins secreted by human endometrial epithelial organoid on stromal cell decidualization. (A) Primary endometrial stromal cells were seeded, grown to 80% confluence in 10% charcoal stripped FBS (CSFBS) for 2 days, changed to 0.5% CSFBS media, and then treated with either vehicle as a control or estrogen and medroxyprogesterone acetate (E2 + MPA) for 6 days. Next, stromal cells were treated with vehicle as a control (CTRL), and E2 + MPA, E2 + MPA with cyclic adenosine monophosphate (E2 + MPA + cAMP), E2 + MPA with prostaglandin E2 (E2 + MPA + PGE2), or E2 + MPA with increasing amounts (10, 100, or 1000 ng/ml) of recombinant cystatin C (CST3), stanniocalcin 1 (STC1), or serpin family A member 3 (SERPINA3) for 4 days. On Day 10, RNA was harvested from cells, and mRNA levels assessed by quantitative RT-PCR. Each treatment was performed in triplicate, and the entire study contained three biological replicates. (B) Established markers of stromal cell decidualization including prolactin (PRL), insulin-like growth factor binding protein 1 (IGFBP1), interleukin 15 (IL15), and somatostatin (SMST). Solid dots show the means for the individual biological replicates. (C) Markers of active decidual cells including crystallin alpha B (CRYAB), hydroxysteroid 11-beta dehydrogenase 1 (HSD11B1), and scavenger receptor class A member 5 (SCARA5). (D) Markers of senescent decidual cells including ABI family member 3 binding protein (ABI3BP), clusterin (CLU), and iodothyronine deiodinase 2 (DIO2). Data are presented as mean ± SEM for each of the n = 3 biological replicates. One-way ANOVA was performed to compare treatment of cells with cAMP, PGE2 or recombinant proteins to E2 + MPA only. Significant effects of treatment (P ≤ 0.05) are denoted by an asterisk (*).

As illustrated in Fig. 4A, primary cultures of human endometrial stromal cells were seeded and grown to 80% confluence, treated with E2 + MPA, E2 + MPA + cAMP, or E2 + MPA + PGE2, and increasing amounts of recombinant CST3, STC1, or SERPINA3 in the presence of E2 + MPA alone (Fig. 4A). Of note, cAMP has been used for decades to induce endometrial stromal cells to decidualize in vitro, but PGE2 may be a more natural and conserved inducer of decidualization (Stadtmauer and Wagner, 2022). Treatment of stromal cells with cAMP increased (P ≤ 0.01) expression of genes that are established markers of decidualization including PRL (prolactin), IGFBP1 (insulin-like growth factor binding protein 1), IL15 (interleukin 15), and SMST (somatostatin) as compared to E2 + MPA only (Fig. 4B). Similarly, PGE2 also elicited decidualization based on increased (P < 0.01) levels of PRL, IGFBP1, IL15, and SMST mRNA compared to E2 + MPA only (Fig. 4B). In the presence of E2 + MPA, treatment of stromal cells with CST3, SERPINA3, and STC1 did not (P > 0.10) stimulate decidualization (Fig. 4B). Compared to E2 + MPA only, CST3 at the 1000 ng per ml concentration decreased (P < 0.05) PRL and IGFBP1 mRNA levels compared to E2 + MPA only but had no effect (P > 0.10) on IL15 or SMST mRNA levels (Fig. 4B).

A recent single-cell transcriptome analysis of human mid-secretory phase endometrium and in vitro endometrial stroma cultures defined genes involved in differentiation of stromal cells into active or senescent DCs (Lucas et al., 2020). CRYAB (crystallin alpha B), HSD11B1 (hydroxysteroid 11-beta dehydrogenase 1), and SCARA5 (scavenger receptor class A member 5) marked active DCs, while ABI3BP (ABI gene family member 3-binding protein), CLU (clusterin), and DIO2 (iodothyronine deiodinase 2) marked senescent DCs. In the present study, cAMP treatment induced the differentiation of endometrial stromal cells into active DCs (Fig. 4C) as well as senescent DCs (Fig. 4D) based on marker gene expression. In contrast, treatment of stromal cells with PGE2 increased (P < 0.01) expression of genes marking active DCs (Fig. 4C), but not (P > 0.10) senescent DCs (Fig. 4D). Addition of increasing amounts of CST3, SERPINA3, or STC1 in the presence of E2 + MPA only had no effects on expression of those marker genes (Fig. 4C and D).

In the next experiment, the effect of CST3 on in vitro stromal cell decidualization was tested by adding increasing amounts of CST3 in the presence of E2 + MPA with PGE2 (Fig. 5A). Inclusion of CST3 at a concentration of 1000 ng per ml decreased PGE2-induced stromal decidualization based on PRL and IGFBP1 mRNA levels (Fig. 5B).

Effects of CST3 on PGE2-induced stromal cell decidualization. (A) Primary endometrial stromal cells were seeded, grown to 80% confluence in 10% charcoal stripped FBS (CSFBS) for 2 days, changed to 0.5% CSFBS media, and then treated with either vehicle as a control or estrogen and medroxyprogesterone acetate (E2 + MPA) for 6 days. Next, stromal cells were treated with vehicle as a control (CTRL), E2 + MPA with prostaglandin E2 (E2 + MPA + PGE2), or E2 + MPA + PGE2 with increasing amounts (10, 100, or 1000 ng/ml) of recombinant cystatin C (CST3), stanniocalcin 1 (STC1), or serpin family A member 3 (SERPINA3) for 4 days. On Day 10, RNA was harvested from cells, and mRNA levels assessed by quantitative RT-PCR. Each treatment was performed in triplicate, and the entire study contained three biological replicates. (B) Established markers of stromal cell decidualization including prolactin (PRL) and insulin-like growth factor binding protein 1 (IGFBP1). Data are presented as mean ± SEM for each of the n = 3 biological replicates. One-way ANOVA was performed to compare treatment of cells with recombinant proteins to E2 + MPA + PGE2 only. Solid dots show the means for the individual biological replicates. Significant effects of treatment (P ≤ 0.05) are denoted by an asterisk (*).
Figure 5.

Effects of CST3 on PGE2-induced stromal cell decidualization. (A) Primary endometrial stromal cells were seeded, grown to 80% confluence in 10% charcoal stripped FBS (CSFBS) for 2 days, changed to 0.5% CSFBS media, and then treated with either vehicle as a control or estrogen and medroxyprogesterone acetate (E2 + MPA) for 6 days. Next, stromal cells were treated with vehicle as a control (CTRL), E2 + MPA with prostaglandin E2 (E2 + MPA + PGE2), or E2 + MPA + PGE2 with increasing amounts (10, 100, or 1000 ng/ml) of recombinant cystatin C (CST3), stanniocalcin 1 (STC1), or serpin family A member 3 (SERPINA3) for 4 days. On Day 10, RNA was harvested from cells, and mRNA levels assessed by quantitative RT-PCR. Each treatment was performed in triplicate, and the entire study contained three biological replicates. (B) Established markers of stromal cell decidualization including prolactin (PRL) and insulin-like growth factor binding protein 1 (IGFBP1). Data are presented as mean ± SEM for each of the n = 3 biological replicates. One-way ANOVA was performed to compare treatment of cells with recombinant proteins to E2 + MPA + PGE2 only. Solid dots show the means for the individual biological replicates. Significant effects of treatment (P ≤ 0.05) are denoted by an asterisk (*).

Discussion

This study highlights the steroid hormone responsiveness of human endometrial epithelial organoids and the influence of those hormones on their transcriptome and secreted proteome. Culture conditions that mimic the steroidal milieu of the menstrual cycle resulted in transcriptional changes in organoids that are reflective of uterine glands in vivo, making them an attractive model for investigating P4-regulated gland products and their influence on stromal cell decidualization and early pregnancy. A comprehensive analysis of the basolateral organoid secretome revealed changes in protein abundance in response to P4. Moreover, one of the secreted proteins (CST3) impacted stromal cell decidualization by attenuating differentiation, supporting the role of gland-stromal crosstalk in regulating biological events important for the establishment of pregnancy.

The development of human organoids (Boretto et al., 2017; Turco et al., 2017) and comprehensive analysis of their cell composition and transcriptome (Fitzgerald et al., 2019) highlights the potential for this culture system as a model of uterine epithelial glands. Commonly, organoids are cultured in a defined medium, established from the study of other tissues and organs such as the intestine (Boretto et al., 2017; Turco et al., 2017). While the media components are necessary for the initial establishment of organoids and to maintain the stem progenitor-like nature of the cells during long-term culture, specific components in the ExM may inhibit the organoid from fully differentiating in response to E2 and P4. In the current study, several factors [N-2 supplement, Noggin, RSPO1 (roof plate-specific spondin-1), HGF, FGF10, EGF, N-acetyl-L-cysteine, Nicotinamide, A83-01] that were in the ExM were removed at the onset of hormone treatment to investigate hormone responsiveness of endometrial organoids. In these conditions, organoids maintained their morphology and exhibited sustained expression of the GE marker FOXA2 and upregulation of a known P4-regulated gene ENPP3. Of note, RSPO1 is a ligand for leucine-rich repeat-containing G-protein coupled receptors (LGR proteins) and positively regulates the WNT signaling pathway (Binnerts et al., 2007). Physiologically, the receptive mid-secretory phase endometrium in women is associated with decreased WNT signaling, as Dickkopf-1 that is a potent inhibitor of WNT signaling, is substantially upregulated in the stroma surrounding the endometrial glands in response to P4 during the transition from the proliferative to secretory phase of the cycle (Tulac et al., 2003, 2006). Thus, removal of WNT activating factors from the organoid culture medium before MPA treatment is likely more physiological.

In the present study, treatment of organoids with E2 and MPA induced substantial changes in the transcriptome, including both increased and decreased expression of genes. By the mid-secretory phase, the PGR is downregulated in and lost from the endometrial epithelial cells but not stromal cells (Lessey et al., 1996), and the loss of PGR is associated with the induction or upregulation of many genes in the endometrial glands. One of the most P4-responsive genes encodes SCGB1D4, a secretoglobin (SCGB) family member (Jackson et al., 2011). SCGB1D4 is stimulated by interferon-gamma and is postulated to have roles in the immune response and regulation of migration and invasion (Choi et al., 2004). OLFM4 (olfactomedin 4) was another P4-increased gene that is mainly expressed in the digestive system, prostate, and bone marrow, with aberrant expression in cancers. In endometrial adenocarcinoma, OLFM4 is positively associated with estrogen receptor 1 (ESR1) and PGR expression and correlated with tumor development (Duan et al., 2014). The relation of OLFM4 to ESR1 and regulation by E2 and EGF suggest a role for OLFM4 in tissue remodeling prior to the secretory phase (Dassen et al., 2010). Our previous study with human organoids identified ENPP3 as a P4-regulated gene (Fitzgerald et al., 2019). In the present study, ENPP3 was increased by E2 + MPA, MPA, and MPA + PGE2, but not E2 alone at the transcript and protein level. In the uterine lumen of women, ENPP3 was enriched in the mid-secretory as compared to the early secretory phase (Boggavarapu et al., 2016; Chen et al., 2018). Reflective of in vivo conditions, ENPP3 was localized apically in steroid hormone-treated organoids, suggesting secretion into the organoid lumen (Boggavarapu et al., 2016). The increase of ENPP3 during the mid-secretory phase of the menstrual cycle, its secretion into the uterine cavity, and regulation by P4 suggest a role for GE-derived ENPP3 in embryo implantation. In fact, overexpression of ENPP3 in an endometrial epithelial cell line increased rates of embryo attachment and expression of endometrial receptivity-related factors, leukemia inhibitory factor, and beta3-integrin (Chen et al., 2018). Of note, endometrial glands and their secretions have an unequivocal role in embryo growth and implantation in sheep (Spencer et al., 2019; Kelleher et al., 2019b). Future studies can leverage organoids as an in vitro model to understand how P4 acts on uterine glands to stimulate genes involved in endometrial receptivity, and the role of uterine glands in post-implantation development of the decidua and the placenta (Fitzgerald et al., 2021; Alzamil et al., 2021; Rawlings et al., 2021).

PGE2 was recently identified as a conserved factor inducing the cyclic AMP/protein kinase A (cAMP/PKA) pathway during stromal cell decidualization (Stadtmauer and Wagner, 2022). Expression of PGE synthase and synthesis of PGE2 was localized to GE and endothelial cells in both basalis and functionalis regions of the human endometrium (Milne et al., 2001). By contrast, stromal staining was predominantly localized in the functionalis layer. Given that PGE2 has a biological role in stromal cell decidualization, PGE2 could also have a role in secretory transformation of the endometrial glands. In the present study, PGE2 treatment had synergistic effects with MPA on the organoid to modulate the organoid transcriptome, and induced decidualization of stromal cells in vitro toward active DC. The effects of PGE2 are via cAMP signaling in rat endometrial stromal cells (Yee and Kennedy, 1991).

In the present study, PGE2 specifically increased expression of several metallothionein genes in the organoids, including MT1G, MT1M, MT1H, MT1X, and MT1E. A recent single-cell transcriptome study of the human endometrium found upregulation of MT genes in unciliated epithelia in the early secretory phase as compared to the proliferative phase (Wang et al., 2020). Metallothioneins are involved in zinc binding and detoxication and are upregulated in human endometrium during the window of implantation (Kao et al., 2002). They play a key role in defending against oxidative stress by scavenging free radicals and protecting against oxidative damage by heavy metals (Cai et al., 1999; Habeebu et al., 2000; Liang et al., 2015; Milnerowicz et al., 2015). Mitigation of oxidative stress during embryo implantation may be important, as oxidative stress is implicated in subfertility and infertility in humans (Agarwal et al., 2012).

The importance of uterine glands and their influence on stromal cell decidualization in animal models suggests a key reciprocal relationship between endometrial epithelia and stromal cells (Kelleher et al., 2017; 2018). While this has been inferred, a direct relationship between them has yet to be identified. Indeed, most studies have focused on proteins secreted apically, either from uterine glands or luminal epithelium, that would directly regulate embryo implantation as well as development of the placenta (Dhakal et al., 2020). In contrast, proteins secreted basolaterally have the potential to regulate the underlying stroma (Fahey et al., 2005). As such, the present study identified P4-regulated proteins secreted in a basolateral manner by the organoids and determined the influence of three proteins using an in vitro stromal cell decidualization assay.

Of the proteins selected for analysis (SERPINA3, STC1, CST3), CST3 was the only protein that impacted stromal cell decidualization. CST3 is a small secretory protein that inhibits cysteine proteases, such as cathepsins, and is present in the luminal and glandular epithelial cells of human endometrium (Abrahamson et al., 1990; Lee et al., 2018). Cathepsins CTSB and CTSL1 and their inhibitor CST3 are increased in the porcine endometrial epithelia during pregnancy and in response to P4 (Song et al., 2010). In the rat endometrium, CST3 peaks just prior to embryo implantation in the GE (Quinn et al., 2006). In the mouse, Cst3 mRNA is initially increased in the peripheral decidualizing cells and the protein localized to the primary decidual zone (Afonso et al., 1997). Of note, cathepsin B and CST3 are upregulated after the completion of decidualization on gestational day 9.5 in mice (Afonso et al., 1997). Therefore, it is possible that CST3 is involved in regulating the degree of decidual breakdown, apoptosis, and breakdown by cysteine proteinases. This idea is supported here by our findings that increasing concentrations of CST3 attenuated stromal cell differentiation into active DC. As such, CST3 may be secreted by endometrial epithelia to govern stromal cell decidualization during pregnancy establishment in women. Of note, autocrine PRL inhibits human endometrial stromal cell decidualization (Eyal et al., 2007). Thus, factors from the glands as well as the differentiated and developing DC may act in concert to locally regulate the complex process of endometrial decidualization during embryo implantation and pregnancy establishment in women. Indeed, early or late decidualization relative to embryo implantation can cause a number of issues including recurrent pregnancy loss and later pregnancy complications (Salker et al., 2010; Lucas et al., 2016; Garrido-Gomez et al., 2017; Sebastian-Leon et al., 2018). In summary, increased knowledge of how uterine glands secrete or produce factors in response to P4 is important to understand the paracrine regulation of stromal cell decidualization, embryo implantation, and placental development in women. Thus, human endometrial epithelial organoid models are useful to interrogate peri-implantation endometrial function, as this area of early pregnancy is difficult to impossible to obtain tissues for study in women.

Supplementary data

Supplementary data are available at Molecular Human Reproduction online.

Data availability

The transcriptome data underlying this article are available in the Gene Expression Omnibus (GSE205084) at https://www.ncbi.nlm.nih.gov/geo/. The proteomic data underlying this article are available in PRIDE (submission reference 1-20220602-7305) at www.ebi.ac.uk/pride/.

Acknowledgements

The authors are grateful to the Research Success Center of the Department of Obstetrics, Gynecology and Women’s Health at the University of Missouri for aid with procuring endometrial tissue. Further, they appreciate the able assistance of Hongyu Liu in transcriptome analysis, and Advanced Technology Core facilities at the University of Missouri.

Authors’ roles

H.C.F., A.M.K., D.J.S., and T.E.S. designed research; H.C.F. and C.R. performed research; H.C.F. contributed new reagents/analytic tools; H.C.F. analyzed data; and H.C.F., A.M.K., and T.E.S. wrote the paper. All authors reviewed the article.

Funding

This work was supported by the Eunice Kennedy Shriver National Institute of Child Health and Development (NIH R01 HD096266 and NIH U01 HD104482) and a Tier 2 award from the University of Missouri System Research and Creative Works Strategic Investment Program. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

Conflict of interest

All authors declare no conflict of interests.

References

Abrahamson
M
,
Olafsson
I
,
Palsdottir
A
,
Ulvsbäck
M
,
Lundwall
Å
,
Jensson
O
,
Grubb
A.
Structure and expression of the human cystatin C gene
.
Biochem J
1990
;
268
:
287
294
.

Afonso
S
,
Romagnano
L
,
Babiarz
B.
The expression and function of cystatin C and cathepsin B and cathepsin L during mouse embryo implantation and placentation
.
Development
1997
;
124
:
3415
3425
.

Agarwal
A
,
Aponte-Mellado
A
,
Premkumar
BJ
,
Shaman
A
,
Gupta
S.
The effects of oxidative stress on female reproduction: a review
.
Reprod Biol Endocrinol
2012
;
10
:
49
.

Aghajanova
L
,
Horcajadas
JA
,
Weeks
JL
,
Esteban
FJ
,
Nezhat
CN
,
Conti
M
,
Giudice
LC.
The protein kinase A pathway-regulated transcriptome of endometrial stromal fibroblasts reveals compromised differentiation and persistent proliferative potential in endometriosis
.
Endocrinology
2010
;
151
:
1341
1355
.

Alecozay
AA
,
Harper
MJK
,
Schenken
RS
,
Hanahan
DJ.
Paracrine interactions between platelet-activating factor and prostaglandins in hormonally-treated human luteal phase endometrium in vitro
.
J Reprod Fertil
1991
;
91
:
301
312
.

Al-Sabbagh
M
,
Lam
EW
,
Brosens
JJ.
Mechanisms of endometrial progesterone resistance
.
Mol Cell Endocrinol
2012
;
358
:
208
215
.

Alzamil
L
,
Nikolakopoulou
K
,
Turco
MY.
Organoid systems to study the human female reproductive tract and pregnancy
.
Cell Death Differ
2021
;
28
:
35
51
.

Binnerts
ME
,
Kim
KA
,
Bright
JM
,
Patel
SM
,
Tran
K
,
Zhou
M
,
Leung
JM
,
Liu
Y
,
Lomas
WE
,
Dixon
M
et al.
R-Spondin1 regulates Wnt signaling by inhibiting internalization of LRP6
.
Proc Natl Acad Sci USA
2007
;
104
:
14700
14705
.

Boggavarapu
NR
,
Lalitkumar
S
,
Joshua
V
,
Kasvandik
S
,
Salumets
A
,
Lalitkumar
PG
,
Gemzell-Danielsson
K.
Compartmentalized gene expression profiling of receptive endometrium reveals progesterone regulated ENPP3 is differentially expressed and secreted in glycosylated form
.
Sci Rep
2016
;
6
:
33811
.

Boretto
M
,
Cox
B
,
Noben
M
,
Hendriks
N
,
Fassbender
A
,
Roose
H
,
Amant
F
,
Timmerman
D
,
Tomassetti
C
,
Vanhie
A
et al.
Development of organoids from mouse and human endometrium showing endometrial epithelium physiology and long-term expandability
.
Development
2017
;
144
:
1775
1786
.

Burton
GJ
,
Watson
AL
,
Hempstock
J
,
Skepper
JN
,
Jauniaux
E.
Uterine glands provide histiotrophic nutrition for the human fetus during the first trimester of pregnancy
.
J Clin Endocrinol Metab
2002
;
87
:
2954
2959
.

Cai
L
,
Satoh
M
,
Tohyama
C
,
Cherian
MG.
Metallothionein in radiation exposure: its induction and protective role
.
Toxicology
1999
;
132
:
85
98
.

Chen
JC
,
Erikson
DW
,
Piltonen
TT
,
Meyer
MR
,
Barragan
F
,
McIntire
RH
,
Tamaresis
JS
,
Vo
KC
,
Giudice
LC
,
Irwin
JC.
Coculturing human endometrial epithelial cells and stromal fibroblasts alters cell-specific gene expression and cytokine production
.
Fertil Steril
2013
;
100
:
1132
1143
.

Chen
Q
,
Xin
A
,
Qu
R
,
Zhang
W
,
Li
L
,
Chen
J
,
Lu
X
,
Gu
Y
,
Li
J
,
Sun
X.
Expression of ENPP3 in human cyclic endometrium: a novel molecule involved in embryo implantation
.
Reprod Fertil Dev
2018
;
30
:
1277
1285
.

Choi
MS
,
Ray
R
,
Zhang
Z
,
Mukherjee
AB.
IFN-γ stimulates the expression of a novel secretoglobin that regulates chemotactic cell migration and invasion
.
J Immunol
2004
;
172
:
4245
4252
.

Cooke
P
,
Buchanan
D
,
Young
P
,
Setiawan
T
,
Brody
J
,
Korach
K
,
Taylor
J
,
Lubahn
D
,
Cunha
G.
Stromal estrogen receptors mediate mitogenic effects of estradiol on uterine epithelium
.
Proc Natl Acad Sci USA
1997
;
94
:
6535
6540
.

Dassen
H
,
Punyadeera
C
,
Delvoux
B
,
Schulkens
I
,
Marchetti
C
,
Kamps
R
,
Klomp
J
,
Dijcks
F
,
de Goeij
A
,
D'Hooghe
T
et al.
Olfactomedin-4 regulation by estrogen in the human endometrium requires epidermal growth factor signaling
.
Am J Pathol
2010
;
177
:
2495
2508
.

Dhakal
P
,
Kelleher
AM
,
Behura
SK
,
Spencer
TE.
Sexually dimorphic effects of forkhead box a2 (FOXA2) and uterine glands on decidualization and fetoplacental development
.
Proc Natl Acad Sci USA
2020
;
117
:
23952
23959
.

Duan
C
,
Liu
X
,
Liang
S
,
Yang
Z
,
Xia
M
,
Wang
L
,
Chen
S
,
Yu
L.
Oestrogen receptor-mediated expression of Olfactomedin 4 regulates the progression of endometrial adenocarcinoma
.
J Cell Mol Med
2014
;
18
:
863
874
.

Evans
J
,
Salamonsen
LA
,
Winship
A
,
Menkhorst
E
,
Nie
G
,
Gargett
CE
,
Dimitriadis
E.
Fertile ground: human endometrial programming and lessons in health and disease
.
Nat Rev Endocrinol
2016
;
12
:
654
667
.

Eyal
O
,
Jomain
JB
,
Kessler
C
,
Goffin
V
,
Handwerger
S.
Autocrine prolactin inhibits human uterine decidualization: a novel role for prolactin
.
Biol Reprod
2007
;
76
:
777
783
.

Fahey
JV
,
Schaefer
TM
,
Channon
JY
,
Wira
CR.
Secretion of cytokines and chemokines by polarized human epithelial cells from the female reproductive tract
.
Hum Reprod
2005
;
20
:
1439
1446
.

Filant
J
,
Spencer
TE.
Endometrial glands are essential for blastocyst implantation and decidualization in the mouse uterus
.
Biol Reprod
2013
;
88
:
93
.

Fitzgerald
HC
,
Dhakal
P
,
Behura
SK
,
Schust
DJ
,
Spencer
TE.
Self-renewing endometrial epithelial organoids of the human uterus
.
Proc Natl Acad Sci USA
2019
;
116
:
23132
23142
.

Fitzgerald
HC
,
Evans
J
,
Johnson
N
,
Infusini
G
,
Webb
A
,
Rombauts
LJR
,
Vollenhoven
BJ
,
Salamonsen
LA
,
Edgell
TA.
Idiopathic infertility in women is associated with distinct changes in proliferative phase uterine fluid proteins
.
Biol Reprod
2018
;
98
:
752
764
.

Fitzgerald
HC
,
Schust
DJ
,
Spencer
TE.
In vitro models of the human endometrium: evolution and application for women's health
.
Biol Reprod
2021
;
104
:
282
293
.

Frank
GR
,
Brar
AK
,
Cedars
MI
,
Handwerger
S.
Prostaglandin E2 enhances human endometrial stromal cell differentiation
.
Endocrinology
1994
;
134
:
258
263
.

Garrido-Gomez
T
,
Dominguez
F
,
Quiñonero
A
,
Diaz-Gimeno
P
,
Kapidzic
M
,
Gormley
M
,
Ona
K
,
Padilla-Iserte
P
,
McMaster
M
,
Genbacev
O
et al.
Defective decidualization during and after severe preeclampsia reveals a possible maternal contribution to the etiology
.
Proc Natl Acad Sci USA
2017
;
114
:
E8468
E8477
.

Gellersen
B
,
Brosens
J.
Cyclic AMP and progesterone receptor cross-talk in human endometrium: a decidualizing affair
.
J Endocrinol
2003
;
178
:
357
372
.

Habeebu
SS
,
Liu
J
,
Liu
Y
,
Klaassen
CD.
Metallothionein-null mice are more sensitive than wild-type mice to liver injury induced by repeated exposure to cadmium
.
Toxicol Sci
2000
;
55
:
223
232
.

Hempstock
J
,
Cindrova-Davies
T
,
Jauniaux
E
,
Burton
GJ.
Endometrial glands as a source of nutrients, growth factors and cytokines during the first trimester of human pregnancy: a morphological and immunohistochemical study
.
Reprod Biol Endocrinol
2004
;
2
:
58
.

Jackson
BC
,
Thompson
DC
,
Wright
MW
,
McAndrews
M
,
Bernard
A
,
Nebert
DW
,
Vasiliou
V.
Update of the human secretoglobin (SCGB) gene superfamily and an example of ‘evolutionary bloom’ of androgen-binding protein genes within the mouse Scgb gene superfamily
.
Hum Genomics
2011
;
5
:
691
702
.

Kao
LC
,
Tulac
S
,
Lobo
S
,
Imani
B
,
Yang
JP
,
Germeyer
A
,
Osteen
K
,
Taylor
RN
,
Lessey
BA
,
Giudice
LC.
Global gene profiling in human endometrium during the window of implantation
.
Endocrinology
2002
;
143
:
2119
2138
.

Kelleher
AM
,
Behura
SK
,
Burns
GW
,
Young
SL
,
DeMayo
FJ
,
Spencer
TE.
Integrative analysis of the forkhead box A2 (FOXA2) cistrome for the human endometrium
.
FASEB J
2019a
;
33
:
8543
8554
.

Kelleher
AM
,
DeMayo
FJ
,
Spencer
TE.
Uterine glands: developmental biology and functional roles in pregnancy
.
Endocr Rev
2019b
;
40
:
1424
1445
.

Kelleher
AM
,
DeMayo
FJ
,
Spencer
TE.
Uterine glands: developmental biology and functional roles in pregnancy
.
Endocr Rev
2019c
;
40
:
1424
1445
.

Kelleher
AM
,
Milano-Foster
J
,
Behura
SK
,
Spencer
TE.
Uterine glands coordinate on-time embryo implantation and impact endometrial decidualization for pregnancy success
.
Nat Commun
2018
;
9
:
2435
2435
.

Kelleher
AM
,
Peng
W
,
Pru
JK
,
Pru
CA
,
DeMayo
FJ
,
Spencer
TE.
Forkhead box a2 (FOXA2) is essential for uterine function and fertility
.
Proc Natl Acad Sci USA
2017
;
114
:
E1018
E1026
.

Klemmt
PA
,
Carver
JG
,
Kennedy
SH
,
Koninckx
PR
,
Mardon
HJ.
Stromal cells from endometriotic lesions and endometrium from women with endometriosis have reduced decidualization capacity
.
Fertil Steril
2006
;
85
:
564
572
.

Lédée-Bataille
N
,
Laprée-Delage
G
,
Taupin
J-L
,
Dubanchet
S
,
Frydman
R
,
Chaouat
G.
Concentration of leukaemia inhibitory factor (LIF) in uterine flushing fluid is highly predictive of embryo implantation
.
Hum Reprod
2002
;
17
:
213
218
.

Lee
RK-K
,
Tseng
H-C
,
Hwu
Y-M
,
Fan
C-C
,
Lin
M-H
,
Yu
J-J
,
Yeh
L-Y
,
Li
S-H.
Expression of cystatin C in the female reproductive tract and its effect on human sperm capacitation
.
Reprod Biol Endocrinol
2018
;
16
:
8
.

Lessey
BA
,
Yeh
I
,
Castelbaum
AJ
,
Fritz
MA
,
Ilesanmi
AO
,
Korzeniowski
P
,
Sun
J
,
Chwalisz
K.
Endometrial progesterone receptors and markers of uterine receptivity in the window of implantation
.
Fertil Steril
1996
;
65
:
477
483
.

Li
Q
,
Kannan
A
,
DeMayo
FJ
,
Lydon
JP
,
Cooke
PS
,
Yamagishi
H
,
Srivastava
D
,
Bagchi
MK
,
Bagchi
IC.
The antiproliferative action of progesterone in uterine epithelium is mediated by Hand2
.
Science
2011
;
331
:
912
916
.

Liang
T
,
Zhang
Q
,
Sun
W
,
Xin
Y
,
Zhang
Z
,
Tan
Y
,
Zhou
S
,
Zhang
C
,
Cai
L
,
Lu
X
et al.
Zinc treatment prevents type 1 diabetes-induced hepatic oxidative damage, endoplasmic reticulum stress, and cell death, and even prevents possible steatohepatitis in the OVE26 mouse model: Important role of metallothionein
.
Toxicol Lett
2015
;
233
:
114
124
.

Lucas
ES
,
Dyer
NP
,
Murakami
K
,
Lee
YH
,
Chan
YW
,
Grimaldi
G
,
Muter
J
,
Brighton
PJ
,
Moore
JD
,
Patel
G
et al.
Loss of endometrial plasticity in recurrent pregnancy loss
.
Stem Cells
2016
;
34
:
346
356
.

Lucas
ES
,
Vrljicak
P
,
Muter
J
,
Diniz-da-Costa
MM
,
Brighton
PJ
,
Kong
C-S
,
Lipecki
J
,
Fishwick
KJ
,
Odendaal
J
,
Ewington
LJ
et al.
Recurrent pregnancy loss is associated with a pro-senescent decidual response during the peri-implantation window
.
Commun Biol
2020
;
3
:
37
.

Macklon
NS
,
Brosens
JJ.
The human endometrium as a sensor of embryo quality
.
Biol Reprod
2014
;
91
:
1
8
.

Milne
SA
,
Perchick
GB
,
Boddy
SC
,
Jabbour
HN.
Expression, localization, and signaling of PGE2 and EP2/EP4 receptors in human nonpregnant endometrium across the menstrual cycle
.
J Clin Endocrinol Metab
2001
;
86
:
4453
4459
.

Milnerowicz
H
,
Ściskalska
M
,
Dul
M.
Pro-inflammatory effects of metals in persons and animals exposed to tobacco smoke
.
J Trace Elem Med Biol
2015
;
29
:
1
10
.

Peter Durairaj
RR
,
Aberkane
A
,
Polanski
L
,
Maruyama
Y
,
Baumgarten
M
,
Lucas
ES
,
Quenby
S
,
Chan
JKY
,
Raine-Fenning
N
,
Brosens
JJ
et al.
Deregulation of the endometrial stromal cell secretome precedes embryo implantation failure
.
Mol Hum Reprod
2017
;
23
:
478
487
.

Quinn
CE
,
Simmons
DG
,
Kennedy
TG.
Expression of Cystatin C in the rat endometrium during the peri-implantation period
.
Biochem Biophys Res Commun
2006
;
349
:
236
244
.

Rawlings
TM
,
Makwana
K
,
Tryfonos
M
,
Lucas
ES.
Organoids to model the endometrium: implantation and beyond
.
Reprod Fertil
2021
;
2
:
R85
R101
.

Salker
M
,
Teklenburg
G
,
Molokhia
M
,
Lavery
S
,
Trew
G
,
Aojanepong
T
,
Mardon
HJ
,
Lokugamage
AU
,
Rai
R
,
Landles
C
et al.
Natural selection of human embryos: impaired decidualization of endometrium disables embryo-maternal interactions and causes recurrent pregnancy loss
.
PLoS One
2010
;
5
:
e10287
.

Sebastian-Leon
P
,
Garrido
N
,
Remohi
J
,
Pellicer
A
,
Diaz-Gimeno
P.
Asynchronous and pathological windows of implantation: two causes of recurrent implantation failure
.
Hum Reprod
2018
;
33
:
626
635
.

Sherwin
JR
,
Hastings
JM
,
Jackson
KS
,
Mavrogianis
PA
,
Sharkey
AM
,
Fazleabas
AT.
The endometrial response to chorionic gonadotropin is blunted in a baboon model of endometriosis
.
Endocrinology
2010
;
151
:
4982
4993
.

Simintiras
CA
,
Dhakal
P
,
Ranjit
C
,
Fitzgerald
HC
,
Balboula
AZ
,
Spencer
TE.
Capture and metabolomic analysis of the human endometrial epithelial organoid secretome
.
Proc Natl Acad Sci USA
2021
;
118
:
e2026804118
.

Smith
SK
,
Kelly
RW.
The release of PGF2α and PGE2 from separated cells of human endometrium and decidua
.
Prostaglandins Leukot Essent Fatty Acids
1988
;
33
:
91
96
.

Song
G
,
Bailey
DW
,
Dunlap
KA
,
Burghardt
RC
,
Spencer
TE
,
Bazer
FW
,
Johnson
GA.
Cathepsin B, cathepsin L, and cystatin C in the porcine uterus and placenta: potential roles in endometrial/placental remodeling and in fluid-phase transport of proteins secreted by uterine epithelia across placental areolae
.
Biol Reprod
2010
;
82
:
854
864
.

Spencer
TE
,
Kelleher
AM
,
Bartol
FF.
Development and function of uterine glands in domestic animals
.
Annu Rev Anim Biosci
2019
;
7
:
125
147
.

Stadtmauer
DJ
,
Wagner
GP.
Single-cell analysis of prostaglandin E2-induced human decidual cell in vitro differentiation: a minimal ancestral deciduogenic signal
.
Biol Reprod
2022
;
106
:
155
172
.

Tulac
S
,
Nayak
NR
,
Kao
LC
,
Van Waes
M
,
Huang
J
,
Lobo
S
,
Germeyer
A
,
Lessey
BA
,
Taylor
RN
,
Suchanek
E
et al.
Identification, characterization, and regulation of the canonical Wnt signaling pathway in human endometrium
.
J Clin Endocrinol Metab
2003
;
88
:
3860
3866
.

Tulac
S
,
Overgaard
MT
,
Hamilton
AE
,
Jumbe
NL
,
Suchanek
E
,
Giudice
LC.
Dickkopf-1, an inhibitor of Wnt signaling, is regulated by progesterone in human endometrial stromal cells
.
J Clin Endocrinol Metab
2006
;
91
:
1453
1461
.

Turco
MY
,
Gardner
L
,
Hughes
J
,
Cindrova-Davies
T
,
Gomez
MJ
,
Farrell
L
,
Hollinshead
M
,
Marsh
SGE
,
Brosens
JJ
,
Critchley
HO
et al.
Long-term, hormone-responsive organoid cultures of human endometrium in a chemically defined medium
.
Nat Cell Biol
2017
;
19
:
568
577
.

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
:
4701
4712
.

Wang
W
,
Vilella
F
,
Alama
P
,
Moreno
I
,
Mignardi
M
,
Isakova
A
,
Pan
W
,
Simon
C
,
Quake
SR.
Single-cell transcriptomic atlas of the human endometrium during the menstrual cycle
.
Nat Med
2020
;
26
:
1644
1653
.

Yee
GM
,
Kennedy
TG.
Role of cyclic adenosine 3′,5′-monophosphate in mediating the effect of prostaglandin E2 on decidualization in vitro
.
Biol Reprod
1991
;
45
:
163
171
.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://dbpia.nl.go.kr/pages/standard-publication-reuse-rights)

Supplementary data