Skip to main content
Human Reproduction Open logoLink to Human Reproduction Open
. 2024 Aug 12;2024(3):hoae048. doi: 10.1093/hropen/hoae048

Aging promotes accumulation of senescent and multiciliated cells in human endometrial epithelium

Marina Loid 1,2, Darina Obukhova 3,4,5, Keiu Kask 6,7, Apostol Apostolov 8,9, Alvin Meltsov 10,11,12, Demis Tserpelis 13, Arthur van den Wijngaard 14, Signe Altmäe 15,16,17, Galina Yahubyan 18, Vesselin Baev 19, Merli Saare 20,21, Maire Peters 22,23, Ave Minajeva 24, Priit Adler 25, Ganesh Acharya 26, Kaarel Krjutškov 27,28, Maria Nikolova 29,30, Felipe Vilella 31, Carlos Simon 32,33,34,35, Masoud Zamani Esteki 36,37,38,, Andres Salumets 39,40,41,
PMCID: PMC11344589  PMID: 39185250

Abstract

STUDY QUESTION

What changes occur in the endometrium during aging, and do they impact fertility?

SUMMARY ANSWER

Both the transcriptome and cellular composition of endometrial samples from women of advanced maternal age (AMA) are significantly different from that of samples from young women, suggesting specific changes in epithelial cells that may affect endometrial receptivity.

WHAT IS KNOWN ALREADY

Aging is associated with the accumulation of senescent cells in aging tissues. Reproductive aging is mostly attributed to the decline in ovarian reserve and oocyte quality, whereas the endometrium is a unique complex tissue that is monthly renewed under hormonal regulation. Several clinical studies have reported lower implantation and pregnancy rates in oocyte recipients of AMA during IVF. Molecular studies have indicated the presence of specific mutations within the epithelial cells of AMA endometrium, along with altered gene expression of bulk endometrial tissue.

STUDY DESIGN, SIZE, DURATION

Endometrial transcriptome profiling was performed for 44 women undergoing HRT during the assessment of endometrial receptivity before IVF. Patients younger than 28 years were considered as the young maternal age (YMA) group (age 23–27 years) and women older than 45 years were considered as the AMA group (age 47–50 years). Endometrial biopsies were obtained on Day 5 of progesterone treatment and RNA was extracted. All endometrial samples were evaluated as being receptive based on the expression of 68 common endometrial receptivity markers. Endometrial samples from another 24 women classified into four age groups (YMA, intermediate age group 1 (IMA1, age 29–35), intermediate age group 2 (IMA2, age 36–44), and AMA) were obtained in the mid-secretory stage of a natural cycle (NC) and used for validation studies across the reproductive lifespan.

PARTICIPANTS/MATERIALS, SETTING, METHODS

A total of 24 HRT samples (12 YMA and 12 AMA) were subject to RNA sequencing (RNA-seq) and differential gene expression analysis, 20 samples (10 YMA and 10 AMA) were used for qPCR validation, and 24 NC samples (6 YMA, 6 IMA1, 6 IMA2 and 6AMA) were used for RNA-seq validation of AMA genes across the woman’s reproductive lifespan. Immunohistochemistry (IHC) was used to confirm some expression changes at the protein level. Computational deconvolution using six endometrial cell type-specific transcriptomic profiles was conducted to compare the cellular composition between the groups.

MAIN RESULTS AND THE ROLE OF CHANCE

Comparisons between YMA and AMA samples identified a lower proportion of receptive endometria in the AMA group (P = 0.007). Gene expression profiling identified 491 differentially expressed age-sensitive genes (P adj < 0.05) that revealed the effects of age on endometrial epithelial growth and receptivity, likely contributing to decreased reproductive performance. Our results indicate that changes in the expression of the cellular senescence marker p16INK4a and genes associated with metabolism, inflammation, and hormone response are involved in endometrial aging. Importantly, we demonstrate that the proportion of multi-ciliated cells, as discovered based on RNA-seq data deconvolution and tissue IHC results, is affected by endometrial aging, and propose a putative onset of age-related changes. Furthermore, we propose that aging has an impact on the transcriptomic profile of endometrial tissue in the context of endometrial receptivity.

LARGE SCALE DATA

The raw sequencing data reported in this article are deposited at the Gene Expression Omnibus under accession code GSE236128.

LIMITATIONS, REASONS FOR CAUTION

This retrospective study identified changes in the endometrium of patients undergoing hormonal replacement and validated these changes using samples obtained during a NC. However, future studies must clarify the importance of these findings on the clinical outcomes of assisted reproduction.

WIDER IMPLICATIONS OF THE FINDINGS

The findings reported in this study have important implications for devising future strategies aimed at improving fertility management in women of advanced reproductive age.

STUDY FUNDING/COMPETING INTEREST(S)

This research was funded by the Estonian Research Council (grant no. PRG1076), Horizon 2020 innovation grant (ERIN, grant no. EU952516), Enterprise Estonia (grant no. EU48695), MSCA-RISE-2020 project TRENDO (grant no. 101008193), EU 874867 project HUTER, the Horizon Europe NESTOR grant (grant no. 101120075) of the European Commission, the EVA specialty program (grant no. KP111513) of the Maastricht University Medical Center (MUMC+), MICIU/AEI/10.13039/501100011033 and FEDER, EU projects Endo-Map (grant no. PID2021-12728OB-100), ROSY (grant no. CNS2022-135999), and the National Science Fund of Bulgaria (grant no. KII-06 H31/2). The authors declare no competing interests.

Keywords: aging, endometrium, gene expression, endometrial receptivity, immunohistochemistry, assisted reproduction


WHAT DOES THIS MEAN FOR PATIENTS?

The twenties are considered the optimal age for female reproduction. However, in the modern era, many women delay their parenting plans, resulting in increased numbers of women seeking infertility treatment later in life. Although assisted reproduction techniques (ARTs), such as IVF, may alleviate the negative impact of advanced maternal age on oocyte quality, the likelihood of conception in the forties is still relatively low. While aging has been linked to endometrial health causing abnormal endometrial thickness, an increased inflammatory background, and altered hormonal responses, we still lack a full understanding of the actual changes in the endometrium on the molecular and cellular level, and how they impact embryo implantation. To explain the age-specific changes, we analysed the gene expression differences in endometrial samples between women of young and advanced ages undergoing HRT and compared the cell type composition of the samples between the two age groups. Our results introduce new knowledge that cellular aging processes may play an important role in the development and functioning of endometrial tissue, and these must be addressed when designing ART protocols for women of advanced reproductive age. Our findings also suggest that particular type of epithelial cells may have potential diagnostic significance for age-related changes. The observed very high proportions of ciliated cells (cells with slender hair-like projections on their surfaces) in some advanced-age patients emphasise the need for further clinical studies to estimate the optimal abundance of ciliated cells in the endometrium for implantation. It is also crucial to investigate whether an increased presence of these ciliated cells hinders embryo implantation or increases reproductive health risks, and therefore requires clinical attention.

Introduction

Nowadays, women often postpone having children due to socioeconomic reasons. In 2021, the mean maternal age in Europe was slightly above 31 years (European Statistical System, 2022). The fertility rate decreases substantially for women in their late thirties due to a decline in ovarian reserves, which may lead them to seek ARTs, such as IVF. For instance, IVF with donor oocytes has made procreation possible for women wishing to have children at older ages (Lambalk, 2022), and preimplantation genetic testing (PGT) of IVF embryos for age-associated genome aberrations has improved the chances of ART success when older women are using their own oocytes (Sanders et al., 2021).

However, advanced maternal age (AMA) remains a prevailing negative factor in the implantation rate for many women undergoing ART. This limitation mainly arises from the general fertility decline beginning at about the age of 35, which refers to the progressive decline of the ovarian follicle pool, accompanied by subsequent hormonal changes that affect cyclic endometrial maturation. The age-associated decline in ovarian function and altered hormonal regulation of adipose tissue may produce abnormally short or long menstrual cycles, resulting in either thin or hyperplastic endometria, both of which are associated with poor implantation rates (Djerassi et al., 1995; Burger et al., 1999; Onogi et al., 2020).

Early studies analysing women undergoing oocyte donation IVF cycles, in which HRT, also referred as artificial cycle, is often required for recipients, have provided controversial reports on the effect of patient age on implantation rate (Flamigni et al., 1993; Navot et al., 1994; Cano et al., 1995; Abdalla et al., 1997). Later studies using large sample sizes have found that the implantation rate drops significantly for women in their forties, even when using donor oocytes from young women (Toner et al., 2002; Soares et al., 2005; Sanders et al., 2021). Although nowadays, PGT for aneuploidies (PGT-A) prior to embryo transfer can prevent many implantation failures (Sacchi et al., 2019; Sanders et al., 2021), we hypothesized that eliminating genetically abnormal embryos via PGT alone is not optimal for ART in women of AMA, and there are specific age-related molecular processes in the endometrium that need to be taken into account. One study showing higher pregnancy rates in ART cycles with younger gestational carriers undergoing embryo transfer with and without PGT-A, compared to older participants (Perkins et al., 2016), supports our hypothesis. Moreover, genetic studies suggest that age increases the gene mutation burden in endometrial tissue, particularly in epithelial cells (Moore et al., 2020), which could potentially alter endometrial function with respect to receptivity and implantation in both natural and assisted conception, and increase the future risk of uterine neoplasms.

Our previous studies have shown that not only can gene expression modulate the receptivity of endometrial tissue, the proportion of different cell types in the endometrium can also affect the embryo–endometrial dialogue during implantation (Suhorutshenko et al., 2018). As characterizing cell types using different molecular techniques can be challenging due to costly and laborious cell-type isolation techniques or single-cell omics technologies, we introduced computational deconvolution as an accurate and cost-effective method to decompose endometrial tissue into individual cellular fractions (Suhorutshenko et al., 2018). In this study, we examine the molecular and cellular changes that occur in endometrial tissue as women age. To understand the extent of these changes, we compared the transcriptome landscape and tissue cellular composition between women of AMA (47–50 years old) and young maternal age (YMA, 20–27 years old) to estimate the effect of aging on endometrial regeneration and receptivity and validated these changes using endometrial samples from natural cycles (NC) across women’s reproductive lifespan (20–50 years old).

Materials and methods

Ethics statement, patients, and samples

Our research complies with all relevant ethical regulations. The research was approved by the Ethics Committee of the University of Tartu (Ethics Approval No. 340-12) and the Ethics Committee of the Faculty of Biology, Plovdiv University (Ethics Approval No. 3/02.09.2019). Informed consent was provided by all participants.

To investigate the association between endometrial receptivity and a woman’s age, we performed the comparison of 1249 anonymized transcriptomic profiles obtained from women undergoing HRT during the assessment of endometrial receptivity with the beREADY algorithm (Meltsov et al., 2023). The profiles were categorized according to their classified receptivity status and compared between age groups of patients aged 20–29, 30–39, 40–44, and 45 and over. To test whether the proportion of patients with any of the classified receptivity statuses changes linearly with age, we used the Chi-squared test for trend in proportions using the prop.trend.test() function in R.

For whole-transcriptome sequencing, endometrial biopsies were obtained from 44 anonymized women with an assessed endometrial receptivity status in HRT cycles, with all samples classified as receptive by the beREADY classifier. The study group consisted of 12 women aged 20–27 years in the YMA group, 12 women aged 47–50 years in the AMA group, and a validation group consisting of 10 YMA and 10 AMA samples. All women were infertility patients planning frozen embryo transfer (FET) in a HRT cycle following the study biopsy. The only inclusion criterion was the absence of reported uterine pathologies. In HRT, progesterone was administered orally, vaginally, or by injection. The first day of progesterone administration was considered Day 0 (P + 0). An endometrial biopsy for endometrial receptivity testing was taken on Day 5 of progesterone administration (P + 5). All endometrial tissue biopsies were collected using a Pipelle catheter (Laboratoire CCD, Paris, France) and immediately placed into RNAlater (Life Technologies, Waltham, MA, USA). An overview of the study design is presented in Fig. 1. All experiments were performed with blinding of their age group affiliation.

Figure 1.

Figure 1.

Schematic overview of the study design and workflow of the analysis. Endometrial biopsy samples were collected from 22 young maternal age (YMA) and 22 advanced maternal age (AMA) women on Day 5 of progesterone administration (P + 5, artificial cycle) undergoing endometrial receptivity testing using 68 transcriptomic markers and the beREADY algorithm. The study samples underwent mRNA sequencing (12 YMA and 12 AMA samples) and small RNA sequencing (11 YMA and 11 AMA samples), and validation samples (10 YMA and 10 AMA) were used for qPCR validation. Additionally, 24 women donated endometrial samples in the natural cycle. For the analyses, FFPE tissue blocks were prepared for all study samples and haematoxylin and eosin-stained slides were evaluated by the pathologist. Immunohistochemistry (IHC) analysis for p16INK4a, C2CD4B, AcTubA, and LhS28 proteins was performed in endometrial samples of both age groups. Sequencing data processing included quality control, adaptor trimming, and alignment to the human genome. Aligned reads were counted and analysed for differential expression of genes between the YMA and AMA groups. Computational deconvolution analysis was performed using single-cell mRNA sequencing data from six endometrial cell populations. The figure was created with BioRender.com.

mRNA sequencing of endometrial samples

Library preparation, sequencing, and initial data processing

Total RNA was extracted from all samples using the miRNeasy kit (Qiagen, Hilden, Germany), followed by DNase treatment with DNase Free (Life Technologies), according to the manufacturer’s protocol. RNA integrity was assessed with a Bioanalyzer (Agilent Technologies, Santa Clara, CA, USA) and concentration was measured with a Qubit RNA BR Assay kit (Thermo Fisher, Carlsbad, CA, USA). The cDNA libraries were randomly assigned to two sequencing batches and prepared using the TruSeq Stranded mRNA Sample Preparation Guide (Illumina, San Diego, CA, USA). The quality was assessed using a Bioanalyzer (Agilent). The libraries were pooled and single-read 75 bp sequencing was performed on NextSeq500 (Illumina).

Reads were then mapped to the human reference genome (UCSC release GRCh37/hg19), and quality control and alignment statistics were assessed using MultiQC (v. 1.12). The mapped reads were estimated at the gene level using the Ensembl database for annotation. The batch effect of the sequencing run was taken into account and differentially expressed genes (DEGs) between AMA and YMA groups were identified with a significance criterion of Benjamin and Hochberg adjusted P-value < 0.05, where the receptivity score was used as a covariate. The top 20 most significant genes, sorted by log2FC, were clustered hierarchically and plotted in a dendrogram. All bioinformatic analysis were performed in R. A detailed description of bioinformatic analyses is presented in Supplementary File S1.

Following the differential expression (DE) analyses, a power estimation for RNA sequencing (RNA-seq) experiments was used to determine the appropriate sample size required to achieve the desired level of statistical power. Using the PROspective Power Evaluation for RNA-seq (PROPER) R package (Wu et al., 2015), it was computed that given the sample size of 12, the significance level (P adj) of 0.05, and the effect size (log2FC) of 0.3, the power estimate was 85%, whereas for the genes with log2FC of at least 1, the power was 99%. Thus, the current study design was considered eligible, with well-powered sample sizes available of 12 YMA and 12 AMA women in the study groups.

To validate the results of RNA-seq analysis, total RNA was extracted from the endometrial samples of the validation group (YMA n = 10, AMA n = 10), then DNA depleted, and cDNA was synthesized using a Maxima First Strand cDNA synthesis kit (Thermo Fisher Scientific Baltics, Vilnius, Lithuania). qRT-PCR experiments were performed in duplicate as previously described (Suhorutshenko et al., 2018). Six loci (ALDH3A1, SPAG6, STC1, PPP1R1B, EML5, and C2CD4B) were selected based on their fold change between the groups and mean expression value in endometrial samples. Oligonucleotide primers are presented in Supplementary Table S1. Expression values were normalized to the endogenous control genes TBP and GAPDH. An unpaired two-sample Wilcoxon test was applied to estimate the significance level in R.

Functional analysis of DEGs

Enrichment analyses for gene ontology (GO) terms and biological pathways were conducted using g: Profiler (v. 0.2.1, updated 19 April 2023; Reimand et al., 2007) and Ingenuity pathway analysis (IPA, Qiagen) tools using all significant DEGs identified between the AMA and YMA groups. For graphical representation of functional analysis data, a GO chord plot was generated using the GOplot (v. 1.0.2) package in R. Networks of significant DEGs were then algorithmically generated based on their connectivity in IPA.

The DEGs were then compared with a set of endometrial receptivity genes differentially expressed in the window of implantation (WOI) of healthy women, as reported in previous publications involving biopsies from natural (Hu et al., 2014; Sigurgeirsson et al., 2017; Suhorutshenko et al., 2018) or HRT (Altmäe et al., 2016) cycles. The boxplots were generated with ggplot (v. 3.4.0). To explore the role of AMA DEGs in endometrial aging, the DEGs were compared with the lists of genes presented in the GenAge (de Magalhães and Toussaint, 2004), GTExAge (Ren and Kuan, 2020), and the Aging Atlas (Aging Atlas Consortium et al., 2021).

miRNA sequencing and analysis

For miRNA expression analysis, the miRNeasy advanced micro kit (Qiagen) was used to extract total RNA from the endometrial biopsies. The quality and quantity of the isolated RNA were assessed using RNA TapeStation (Agilent). For library preparation, 100 ng of total RNA was utilized, and subsequent libraries were generated using the NEXTflex Small RNA-seq Library Prep Kit v4 (Perkin Elmer, Waltham, MA, USA). The FASTQ files underwent quality assessment and adapter trimming using FastQC and Trim Galore. The miRGalaxy tool within Galaxy was employed to identify and classify isomiRs, i.e. miRNA sequences that have variations with respect to the reference sequence, based on their read offset relative to the reference miRs (RefSeq) and read copy number (Glogovitis et al., 2021). The IsoRead tool was used to identify miRs and isomiRs, followed by their classification into template and non-template isomiRs using the same module. For DE analysis, count matrices of miRs and isomiRs were analysed using DESeq2, and the expression threshold log2FC ≤ 0.5 and P-value < 0.05 were applied. The mRNA targets of miR/isomiRs were predicted using the miRDB database (Chen and Wang, 2020).

Validation using the NC samples of young, intermediate, and advanced age groups

To investigate whether expression changes found in the AMA group evolve over time, we performed coding RNA-seq of endometrial samples collected from individuals aged between 21 and 51 years. The patients were divided into four age groups: YMA (age 21–27, n = 6), intermediate maternal age group 1 (IMA1; age 28–36, n = 6), intermediate maternal age group 2 (IMA2; 37–44, n = 6) and AMA (45–51, n = 6). Endometrial biopsies were collected during the mid-secretory phase in NC, where no hormonal treatment was used for at least 3 months prior to sampling. There were no significant differences in blood levels of oestradiol and progesterone between the groups. mRNA was extracted from endometrial samples using the miRNeasy micro kit (Qiagen) with on-column DNase treatment (Qiagen), following the manufacturer’s instructions. RNA integrity and concentration were assessed with Qubit (Thermo Fisher) and sequencing libraries were synthesized using a TruSeq Exome RNA Library preparation kit (Illumina). Library quality and molarity were assessed using a TapeStation HS D1000 assay and sequencing was performed on a NextSeq 1000 platform (Illumina).

After the sequencing, read quality and alignment were processed as previously described. Genes with raw count values <4 in over 75% of samples were excluded from the analysis. To compare DEGs among four age groups in a pairwise manner, we used ‘∼Age_Group’ as the value for the ‘design’ argument in the DESeqDataSetFromMatrix() function. A detailed description of the analysis is presented in Supplementary File S1.

Deconvolution

Two single-cell transcriptome datasets of endometrial biopsies, the C1 dataset (library preparation method: Fluidigm; with 2148 cells analysed) and the 10× Genomics dataset (library preparation method: 10× Chromium Next GEM; with 71 032 cells analysed), and their respective cell-type labels were obtained from NCBI’s Gene Expression Omnibus (accession code GSE111976) (Wang et al., 2020). The dataset was chosen based on the criteria of completeness, annotation, sample size, and availability of supportive information. For the menstrual cycle phase, concordance with the bulk RNA-seq samples, single-cell data from the mid-secretory phase, cycle days 20–24 were only included. This resulted in 546-cell and 46 878-cell count matrices for the C1 and 10× Genomics datasets, respectively. These matrices of distinct endometrial cell types were used to calculate the proportions of each cell type in the bulk endometrial samples of the study group, using the dampened weighted least squares (DWLS) method from the DWLS R package (v. 0.1.0). To compute the significance of differences in the proportions of each endometrial cell type between the YMA and AMA samples, a non-parametric Mann–Whitney U-test (two sides) was used, and Bonferroni correction was applied. To visualize whether the proportion of ciliated epithelial cells correlated with a woman’s age, the samples were ordered according to age. Violin, bar, and scatter plots were produced using the R package ggplot2 (v. 3.4.0).

Histological evaluation and immunohistochemistry

For histological analysis of the study group samples, 24 microscope slides with tissue sections (4 μm, three sections per slide) were prepared at the Pathology Department of Tartu University Hospital (Tartu, Estonia) from formalin-fixed paraffin-embedded (FFPE) endometrial tissue sections using a standard haematoxylin and eosin staining protocol (Fischer et al., 2008). The slides were scanned at East Tallinn Central Hospital, using a 3DHistech Panoramic Flash III 250 scanner (3DHistech, Budapest, Hungary) at 20× magnification. The evaluation of cyclic endometrial changes was performed according to Noyes criteria (Noyes et al., 1950) by a pathologist.

To evaluate the expression of cellular senescence protein p16INK4a (p16, CDKN2A), immunohistochemistry (IHC) analysis was performed, using anti-p16INK4a, mouse monoclonal antibody against human CDKN2A (ready-to-use solution, Master Diagnostica, Valencia, Spain). Four samples from both the YMA and AMA groups were selected at random, with all samples considered receptive according to the beREADY algorithm. To ensure antibody specificity, tonsil tissue was used as a positive control for p16 staining, and healthy cervix tissue was used as a negative tissue control. FFPE tissue sections on slides were processed according to a standard protocol using the 3,3'-diaminobenzidine (DAB)-based Master Polymer Plus Detection System IHC Kit (Master Diagnostica). Incubation with the primary antibody was carried out overnight in a humidity chamber at 4°C. Further steps were carried out according to the manufacturer’s protocol. The chromogenic reaction was developed for 30 s and stopped thereafter. Cell nuclei were counterstained with Mayer’s haematoxylin solution for 15 s, and then the slides were washed for 10 min with tap water. The slides were dehydrated through graded ethanol and xylene solutions and mounted with a Leica CV mount (Leica Biosystems, Wetzlar, Germany).

For ciliated cell visualization, IHC staining of cilia basal bodies was performed as previously described using a mouse monoclonal anti-LhS28 antibody (Santa Cruz Biotechnology, Heidelberg, Germany) and anti-acetylated tubulin-α (AcTubA; Santa Cruz) was used to stain motile cilia. As a positive control for LhS28 staining, an additional endometrial biopsy was taken on Day 15 of the natural menstrual cycle from a fertile young woman aged 28 years. A fallopian tube biopsy from a 42-year-old woman was used as a positive control for AcTubA. For all negative controls, the primary antibody was omitted from the incubation. All information on the antibodies used in this study is listed in Supplementary Table S1.

Slides were scanned using a Leica SCN 400 slide scanner (Leica Biosystems) with a maximum magnification objective of 20× for IHC analysis and 40× for basal body (BB) and multiciliated cells (MCC) counting. Semi-quantitative analysis of three different areas of scanned sections was conducted with the ImageJ Fiji package (v. 1.52e) (Schindelin et al., 2012). Briefly, the intensity of the DAB signal was measured separately for the stromal, glandular epithelium, and surface epithelium of the endometrium. The relative DAB intensity was calculated using the formula: f = 255 − i, where ‘f’ is relative DAB intensity and ‘i’ is mean DAB intensity obtained from the software ranging from 0 (deep brown, highest expression) to 255 (total white). The Wilcoxon Mann–Whitney test was performed to determine statistical significance. Prism 6 (GraphPad Software, La Jolla, CA, USA) and ImageJ software (Fiji package) were used for data quantification and analysis. BB and MCC counting were performed with blinding by two researchers using anti-LhS28-stained and anti-AcTubA-stained endometrial tissue slides, respectively. The average amounts of ciliated cells exhibiting positive staining between the two measurements were estimated and divided by the area of luminal epithelium (LE). An unpaired sample Student’s t-test was applied to test the statistical significance of the differences between the two groups.

Identification of cilia-associated genes among AMA DEGs

To understand the impact of endometrial aging on cilial cells’ structure and function, we first compared 491 AMA DEGs with the 535 genes and proteins presented in the Human Protein Atlas (https://www.proteinatlas.org), which are highly expressed in the ciliated cells of human fallopian tube and endometrial tissue. The intersecting genes were then searched manually in PubMed (https://pubmed.ncbi.nlm.nih.gov) for evidence of their associations with cilia structures and functions. The expression of cilia-associated AMA DEGs was then analysed pairwise among four age groups.

Results

Receptivity of AMA endometrium based on transcriptomic markers

To determine whether a woman’s age may be a factor that affects endometrial receptivity, we compared 1249 endometrial receptivity test reports. We identified that a significantly (P = 0.007) lower proportion of samples tested as ‘receptive’ in women over 45 years old compared to women younger than 30 years. The proportions of receptivity status are presented in Fig. 2. The patient group over 45 years exhibited slightly higher proportions of all results of receptivity status, except for receptive result (Fig. 2A), and these changes occurred gradually over the age groups (Fig. 2B).

Figure 2.

Figure 2.

The results of the analysis of 1249 endometrial receptivity reports based on the beREADY algorithm. (A) The percentage of samples classified as ‘pre-receptive’, ‘early receptive’, ‘receptive’, ‘late receptive’, and ‘post-receptive’ in four age groups, arranged by receptivity status. The proportion of ‘receptive’ samples was significantly lower in women older than 45 years, compared to women aged 20–29 years. Chi-squared test P-value = 0.007. (B) The proportions of receptivity status, arranged by age. ** Chi-squared test P-value < 0.01.

DEGs and their respective altered pathways

To analyse the differences in gene expression between the endometria of YMA and AMA women, total RNA samples from the endometrial tissue of 12 young and 12 older women were sequenced. The main characteristics of the study groups are presented in Table 1. In total, 19 357 mRNA transcripts were expressed in the analysed endometrial samples. Based on the gene expression profile, the study samples in each group clustered together. The dendrogram of all study group samples based on the top 50 DEGs is presented in Supplementary Fig. S1A.

Table 1.

Overview of the study participants.

YMA AMA P-value*
Mean (±SD) Mean (±SD)
Study group (N) 12 12
Age 25.2 (1.2) 47.9 (1.2)
BMI 23.9 (4.1) 22.9 (3.1) ns
Cycle type HRT: P + 5 HRT: P + 5
qPCR validation group (N) 10 10
Age 25.4 (5.1) 47.7 (3.7)
BMI 24.1 (5.1) 23.3 (3.8) ns
Cycle type HRT: P + 5 HRT: P + 5
YMA IMA1 IMA2 AMA P-value*
Mean (±SD) Mean (±SD) Mean (±SD) Mean (±SD)
RNA-seq validation group (N) 6 6 6 6
Age 22.8 (1.8) 32.2 (1.7) 37.7 (2.8) 47.3 (2.1)
BMI 21.6 (1.6) 22.3 (3.1) 24.4 (3.5) 23.8 (2.9) ns
Cycle type NC: LH + 7 NC: LH + 7 NC: LH + 7 NC: LH + 7

YMA, young maternal age group; AMA, advanced maternal age group; IMA1 and IMA2, intermediate maternal age groups 1 and 2, respectively; N, number of participants in each group; HRT, HRT in artificial menstrual cycle; P + 5, Day 5 of progesterone administration; NC, natural menstrual cycle; LH, LH surge day; ns, non-significant.

*

Unpaired Student t-test P-value for pairwise comparisons.

After multiple testing corrections, 491 significant DEGs, 271 upregulated and 220 downregulated, were identified in the endometrium of the AMA group compared to the YMA group (Supplementary Table S2). The changes in the expression rate ranged from a –7 to a 7-fold change. No differences were observed in the expression of steroid hormone receptors between the groups. The genes with the highest expression fold change were GAST, TBX15, RBP4, GP2, ADGRF1, STC1, S100P, HPSE, DEPP1, KLK3, CXCL14, and CTNND2. The most statistically significant DEGs were ZNF229, C2CD4B, TBX15, SPAG6, OR1J4, TOR4A, CLDN4, GRB7, C2CD4A, RBP4, PPP1R1B, and RIMKLB (Fig. 3A, Supplementary Table S2). Real-time quantitative PCR analyses using validation group samples confirmed the expression differences of several DEGs, including ALDH3A1, EML5, SPAG6, STC1, PPP1R1B, and TBX15 (Supplementary Fig. S1B) in AMA endometrial samples compared to YMA samples.

Figure 3.

Figure 3.

Genes dysregulated in the endometrium of women of advanced maternal age. (A) The top differentially expressed (DE) genes and their biological functions, FC > 1, Benjamin–Hochberg adjusted P-value < 0.05. (B) Molecular interaction network generated using Ingenuity pathway analysis (IPA). One of the top significant networks (score 35) suggests the interaction between the p16 (CDKN2A) senescence marker with Notch, ERK and Hedgehog signalling. (C) Functional analysis of DE genes in advanced maternal age (AMA) samples represent gene ontology (GO) terms significantly enriched in the endometrium of AMA samples compared to young maternal age (YMA) samples. The most significantly enriched terms were cilium (GO: 0005929, P = 2.42×10−20), cilium movement (GO: 0003341, P = 4.74×10−17), axoneme (GO: 0005930, P = 2.00×10−17), and motile cilium (GO: 0031514, P = 1.44×10−17). BP, biological processes; CC, cellular component; HP, human phenotype; MF, molecular function; REAC, reactome; TF, transcription factor; HPA, Human Protein Atlas; WP, WikiPathways. GO terms marked with numbers are described in Supplementary Table S3A. (D and E) Immunohistochemistry (IHC) analysis of p16 (p16INK4a) senescence protein in YMA and AMA samples, respectively. (F) Negative control staining for p16, in which the primary antibody was omitted. (G) p16INK4a protein expression in endometrial cellular compartments, identified by IHC from YMA patients and AMA patients. p16 exhibited significantly higher expression in luminal epithelial cells (LE) and glandular epithelial cells (GE) (Wilcoxon rank-sum test P < 0.05).

Functional analysis of AMA DEGs showed that the most enriched GO terms were general cellular and organismal developmental processes. Significantly enriched cellular components were cilium (GO: 0005929; P adj = 2.42×10−20), axoneme (GO: 0005930, P = 2.00×10−17) and motile cilium (GO: 0031514; P adj = 1.44×10−17), as well as the biological process cilium movement (GO: 0003341, P adj = 4.74×10−17), as shown in Fig. 3C. Thus, the enrichment analysis identified GO terms associated with cilial development and functioning (Fig. 3C). A full list of significantly enriched terms is presented in Supplementary Table S3A. IPA analysis indicated nine significant molecular networks, including the networks associated with cell cycle progression and cancer, cellular movement (including immune cell movement), endocrine system disorders (including metabolism disorders), cell morphology, and development (Supplementary Table S3B, IPA score > 21, Benjamini and Hochberg P-value < 0.05). According to the IPA, DEGs in these networks are directly or indirectly associated with the cell cycle regulator molecule cyclin-dependent kinase 2A (CDKN2A), better known as the p16INK4a/ARF locus, which was upregulated in the AMA group (Fig. 3B, Supplementary Table S2). It encodes a cellular senescence marker, which interacts with Sonic hedgehog and Notch pathways (Fig. 3B, Supplementary Table S3B). CDKN2A was also the only AMA gene identified by all three databases for age-associated genes.

When examining DEGs for their putative role in endometrial functioning, we found that many genes upregulated in AMA were associated with cell cycle control (CDKN2A), insulin signalling (TMED6), and endometrial receptivity (GALNT12, TMED6, CLDN4, CD55), while downregulated genes were linked with sugar metabolism and inflammation (C2CD4A, C2CD4B, NFKB), cellular movement and invasion (SPAG6, HPSE, GRB7), and ovarian hormone signalling (STC1, GRB7, ALDH3A1). When compared to previously reported genes associated with endometrial receptivity (Hu et al., 2014; Altmäe et al., 2016; Sigurgeirsson et al., 2017; Suhorutshenko et al., 2018), 26 out of 73 transcriptomic endometrial dating genes (marked with an asterisk in Supplementary Fig. S2A) were dysregulated in AMA samples and 18 AMA genes (Supplementary Fig. S2B) were associated with HRT-specific changes in the endometrium.

The expression of p16INK4a in endometrial tissue

To localize the expression of the cellular aging marker p16INK4a (p16) in AMA/YMA endometria, IHC analysis was performed in four YMA and four AMA endometrial samples. The analysis of protein staining intensity showed that p16 was significantly higher in the luminal and glandular epithelial cells of the AMA samples (P = 0.049 and 0.026, respectively, Wilcoxon rank-sum test, two-sided), when compared to the YMA group (Fig. 3D–G). p16 was localized in both the cytoplasm and nuclei of epithelial and stromal cells, which was considered as positive staining. No differences in the cellular localization of p16 were observed between the groups. The staining intensity showed a higher degree of variation in the AMA group than in the YMA group, indicating possible age-related endometrial tissue heterogeneity.

The expression of AMA genes across the woman’s reproductive lifespan

The transcriptomes of 24 endometrial samples from women aged between 20 and 50 years obtained in NC were analysed in four age groups (YMA, IMA1, IMA2, and AMA). The expression of AMA genes was pairwise compared among four age groups across a woman’s reproductive lifespan (between 20 and 50 years old). As a result, several AMA genes changed significantly at different ages (Bonferroni adjusted P < 0.05). We present these genes in Fig. 4. Most of the age-associated changes occurred in the late thirties and early forties (Fig. 4A). However, the AMA genes ERICH3, SNTN, and WDR38 were significant only between the twenties and early thirties (IMA1 vs YMA). The expression of the TBX15 gene, the most highly differentially expressed gene in the AMA group, changed significantly at several ages and correlated positively with a woman’s age (R = 0.64, P = 0.00071) (Fig. 4B). The steroid hormone receptor genes showed no significant changes between groups. The full list of significant DEGs among the four age groups is presented in Supplementary Table S4.

Figure 4.

Figure 4.

The expression of advanced age-associated genes in the natural cycles across woman’s reproductive lifespan. (A) The pairwise comparisons among four age groups (young maternal age (YMA), intermediate maternal age groups 1 and 2 (IMA1, IMA2), and advanced maternal age (AMA)) revealed the possible onset of specific AMA-related gene expression changes (Bonferroni adj P-value < 0.05). The upregulated genes in groups with higher age are presented in white and the down-regulated genes are presented in black, which corresponded to the direction of gene expression changes in the study group. (B) Gene TBX15, which participates in the epithelial-to-mesenchymal transition of epithelial cells, showed a positive correlation with age of the women (R = 0.64, P-value = 0.00071).

Endometrial cell population proportions in YMA and AMA groups

Histological evaluation of the study group samples according to the Noyes criteria (Noyes et al., 1950) confirmed that all endometrial samples were in the secretory phase and corresponded to the receptive endometrium according to the beREADY algorithm (Meltsov et al., 2023). The results based on the Fluidigm C1 single-cell dataset of six endometrial cell types (Wang et al., 2020) showed a significantly higher proportion of ciliated cells (epithelial MCCs) in AMA samples than in YMA samples (proportion, 0.01 ± 0.005 vs 0.004 ± 0.002; P < 0.05, Wilcoxon rank-sum test, two-sided; Fig. 5A and B). The same analysis was also performed with the 10× Genomics single-cell dataset of eight endometrial cell types (Wang et al., 2020). It also revealed that MCCs were more abundant in the AMA samples than in their YMA counterparts (proportion, 0.006 ± 0.009 vs 0.0009 ± 0.002), although the difference was not statistically significant. This could be due to the different sample preparation and analysis techniques used to establish these two single-cell RNA-seq datasets. When the samples were ordered by the age of the women, it became clear that the AMA group samples had a higher, but more variable proportion of the MCCs than the YMA group (Fig. 5C).

Figure 5.

Figure 5.

Cell type proportions per age group determined by deconvolution analysis. All cell types have an associated colour, shown in the key. (A) Violin plots show the changes in cellular proportions between young and advanced maternal age groups for six cell types based on the Fluidigm C1 single-cell RNA-sequencing dataset. The difference in epithelial multiciliated cells (MCCs) between young and advanced maternal age groups was statistically significant, with increased numbers of MCCs in advanced maternal age (AMA) samples, according to two-sided Wilcoxon test with Bonferroni correction, P = 0.007. The boxplots show the interquartile range (box limits) and median (centre black line) of cellular proportion levels. A significantly higher proportion of MCCs was observed in the AMA group. (B) Stacked bar plots depict percentages of six cell types in young and advanced maternal age groups based on the Fluidigm C1 single-cell RNA-sequencing dataset. (C) Proportions of endometrial MCCs aligned according to age of the women.

IHC analysis of MCCs in the endometrium

IHC analyses of the cilia BBs and motile cilia, using anti-LhS28 and anti-AcTubA antibodies, respectively, were also conducted to determine whether the AMA samples had more ciliated cells than the YMA samples. The stained basal bodies of the ciliated cells were observed as crescent-shaped organelles near the surface of the epithelial cells with motile cilia visible on the surface (Fig. 6A–D). The number of positively stained BBs was counted in image analysis per area of the LE, i.e. BB counts per LE, in both the YMA and AMA groups. The average BB count per LE area was higher in the AMA group than in the YMA group (3.6 × 10−3 (SD ± 8 × 10−4) vs 2.0×10−3 (SD ± 9×10−4), respectively; Unpaired samples Student’s t-test, P = 0.02) (Fig. 6E). AcTubA-stained cilial structures were seen on the top of MCCs (Fig. 6F–I). The number of MCCs per LE area was higher also in AMA samples (2.16 × 10−3 (SD ± 7 × 10−4)) than in the YMA group (1.4 × 10−3 (SD ± 4 × 10−4)) (Unpaired samples Student’s t-test, P = 0.03) (Fig. 6F–J), which also supported the results of the deconvolution analysis.

Figure 6.

Figure 6.

Analysis of multiciliated cells in endometrial samples from young and advanced maternal age group women. (AD) Basal body staining using LhS28 antibody in endometrial tissue: young maternal age (YMA), advanced maternal age (AMA), Day 16 of natural cycle (positive control of endometrial tissue sample from a fertile YMA woman) and a negative control, respectively. (E) Relative basal body (BB) count per luminal epithelium (LE) area, calculated based on LhS28 staining in human endometrium, showed significantly higher numbers of BBs in the AMA group, according to independent sample Student’s t-test, P = 0.02. (FI) Motile cilia staining using an acetylated α-tubulin (AcTubA) antibody in endometrial tissue: YMA, AMA, a fallopian tube sample as a positive control, and a negative control, respectively. Primary antibodies were omitted from negative control experiments. (J) Relative multiciliated cells (MCC) count per luminal epithelium (LE) area, calculated based on AcTubA staining in human endometrium, showed significantly higher numbers of MCCs in the AMA group, according to independent sample Student’s t-test, P = 0.03. (K) Genomic localization of differentially expressed miRNAs associated with cilia and the target genes. miR-34b and miR-34c are localized to a single cluster that resides in the host gene MIR34BHG on chromosome 11. The target genes for miR-34b and miR-34c and their isomiRs are predicted with the miRDB database according to their sequence, which is depicted in colour. (L) Structure of endometrial multiciliated cells (MCCs) and functions of motile cilia affected by AMA. The gene names represent differentially expressed genes in the AMA endometrium (Benjamin and Hochberg (BH) adjusted P-value < 0.05) and are associated with motile cilia. baseMean, average expression in analysed samples; Log2FC, logarithmic expression fold change; padj, BH-adjusted p-value; PMID, PubMed identification number. The figure was created using BioRender.com.

DE of miRNAs in AMA vs YMA endometrium

To provide further evidence for AMA gene involvement in ciliogenesis via epigenetic miRNA interaction, we analysed the profiles of small non-coding miRNAs in 11 YMA and 11 AMA endometrial biopsies using small-RNA-seq performed by a size-based enrichment step with magnetic beads. A total of 220 differentially expressed miR/isomiRs were identified in the AMA group, 91 upregulated and 129 downregulated. The top 30 differentially expressed miRNAs, templated and non-templated isomiRs are presented in Supplementary Fig. S3 A–C. Of these, the cilia-associated miR34 family was represented by two RefSeqs (miR-34b-5p and miR-34c-5p) and 11 isomiRs, which had the highest log2FC values (Supplementary Fig. S3C). The miR-34b-5p isoform was most abundantly expressed within the family in AMA endometria. Notably, miR-34b and miR-34c were localized to a single cluster that resides in the host gene MIR34BHG on chromosome 11, as shown in Fig. 6K. Most of the isomiRs had a 5' shift of one nucleotide relative to the corresponding canonical microRNA. This leads to a change in the ‘seed’ regions, while the ‘seed’ of miR-34b matches those of the miR-34c isomiRs and vice versa. Among the predicted target genes, four were differentially expressed in AMA: MET, DAAM1, EML5 (targets of miR-34c-5p and the isomiRs of miR-34b-5p), and CTNND2, which is a target of all miR34 family members (Fig. 6K, Supplementary Table S2), which regulate motile cilia development (Song et al., 2014; Bonser et al., 2021).

Cilia-associated genes among AMA DEGs

To understand the impact of endometrial aging on cilial cell structure and function, we compared our DEG list with publicly available datasets. A total of 93 AMA DEGs were identified as highly expressed in the ciliated cells of human fallopian tube and endometrial tissue, based on the Human Protein Atlas (https://www.proteinatlas.org). These genes were then searched manually in PubMed (https://pubmed.ncbi.nlm.nih.gov) for associations with cilia structures and/or functions. We found 62 publications that provided evidence of cilia association for 84 AMA genes. The identified cilia-associated AMA DEGs are presented in Supplementary Table S5, and their association with cilial structures is shown in Fig. 6L.

In the NC validation group, most of the cilia-associated AMA DEGs were significant only between the YMA and AMA groups (Supplementary Fig. S4, Supplementary Table S4), confirming the results from the main study groups (Supplementary Table S5). The scatter plot illustrates the expression of cilia-associated genes at different ages, where the dots represent the samples. The results suggest a lower expression of cilia genes, except for SPAG6 gene, in the YMA, IMA1, and IMA2 groups, and a higher expression in the AMA group.

Discussion

This retrospective study delves into the phenomenon of endometrial aging with the goal of comprehending its mechanisms, pinpointing its onset, and uncovering its potential influence on endometrial function. Here, we compared the transcriptome profiles of endometrial biopsies taken during the WOI period from women of young and advanced reproductive ages. Compared to other endometrial aging studies, we analysed participants who followed a similar endometrial preparation protocol that minimizes inter-individual differences due to hormonal status bias and validated our findings using samples from NC. As HRT is often administered to AMA patients preparing for FET, our study provides considerable evidence of the impact of age on the development of endometrial tissue, as well as its molecular profile and cellular composition in women undergoing ART.

To begin with, the analysis of endometrial receptivity reports of women undergoing HRT showed that the endometrium of women over the age of 45 has a lower receptivity potential than that of women in their twenties (Fig. 2), which prompted us to investigate this phenomenon further. The comparison of gene expression profiles of YMA and AMA endometria revealed a set of genes that were dysregulated in the older women (Supplementary Table S2). The pathways most affected by age were cellular remodelling, cilial movement and cell migration, and immune response, whereas most enriched GO terms were associated with motile cilia (Fig. 3C, Supplementary Table S3), arising from a high proportion of cilia-associated genes (17.1%; 84 out of 491) (Supplementary Table S5), which aligns with previous work by the Díaz-Gimeno group (Devesa-Peiro et al., 2022). Although most genes change significantly during the forties, early alterations in SNTN and WDR38 in epithelial cells may signify the early onset of changes in the MCCs (Fig. 4A, Supplementary Fig. S4, Supplementary Table S4). The deconvolution analysis using cell type-specific gene expression profiles showed a significantly higher proportion of MCCs in the AMA group, confirmed by IHC (Figs 5A–C and 6). Previous studies have shown that MCCs are a distinct cell subtype within the endometrial epithelium, characterized by a specific transcriptomic profile (Wang et al., 2020; Garcia-Alonso et al., 2021). Motile cilia are microtubule-based cellular structures having a 9 + 2 axonemal organization and accessory structures that enable cilia movement. In the female reproductive tract, MCCs guarantee the movement of mucus and the embryo along the epithelial surface. Ciliogenesis is driven, although not exclusively, by oestrogen and through the inhibition of Notch signalling (Haider et al., 2019), and regulated by miR34/449 family microRNAs (Marcet et al., 2011; Song et al., 2014; Chevalier et al., 2015). Our miRNA analysis also confirmed the upregulation of cilia-regulating miRNAs and their target genes in AMA women (Fig. 6K, Supplementary Fig. S3).

During the menstrual cycle, rising oestrogen levels typically promote MCC proliferation and differentiation from epithelial progenitor cells, peaking around cycle Days 15–16. At this point, progesterone produced by the corpus luteum opposes oestrogen action, reducing the number of MCCs by the time of potential implantation (Masterton et al., 1975; More and Masterton, 1976). These cyclic events highlight the importance of appropriate cellular proportions of the MCCs in ensuing endometrial receptivity, including both sufficient proliferation and a programmed decrease in MCC number. The observed shift in MCC proportion may be caused by the increased production of ciliated cells or their insufficient decrease under the influence of secretory phase hormones in AMA women. This suggests that both an excess of oestrogen and a lack of progesterone activity may produce a larger amount of MCCs. Considering that MCCs are the major epithelial cell type in fallopian tubes (Lyons et al., 2006), their higher abundance might not favour embryo implantation in the uterus.

Exploring AMA DEGs, we identified a higher expression of CDKN2A (Fig. 3B and G, Supplementary Table S2), which encodes cyclin-dependent kinase (e.g. p16INK4a), a common biomarker of aging (Martin et al., 2014). Both the cytoplasmic and nuclear staining observed conformed with the endometrial cellular localization of p16 previously described by Parvanov et al. (2021). While nuclear overexpression is associated with pathological conditions driven by p16 activation, cytoplasmic p16 accumulation was suggested to be the sign of inactive p16 interacting with other proteins that prevent its activity (Martin et al., 2014; Mendaza et al., 2020; Parvanov et al., 2021). Although it has been suggested that p16-positive epithelial cells are required for a supportive environment at the WOI, it has also been shown that p16-staining in endometrial epithelial cells correlates positively with a woman’s age (Parvanov et al., 2021). Our molecular network analysis revealed the interaction of p16 with Notch signalling, which also regulates cell cycle progression (Fig. 3B). Normally, activated Notch induces apoptosis in MCCs in vertebrate animals (Tasca et al., 2021). In endometrium, senescent cells were shown to trigger overexpression of p16 by activating the mechanism of premature senescence (Vassilieva et al., 2018). Notably, the accumulation of putatively inactive p16 in the cytoplasm has also been associated with aging (Idda et al., 2020). Although it is unclear how p16 accumulation affects the human endometrial epithelium, it has been shown that p16 overexpression is associated with endometrial tubal metaplasia, characterized by a higher number of MCCs in the endometrium, and is considered a specific marker for dysplastic and neoplastic epithelial cells (Klaes et al., 2001; Horree et al., 2007). The detailed role of p16 in the development and function of endometrial MCCs remains largely understudied. This may be due to the fact that changes in CDKN2A/p16 may remain sub-significant in NC endometrium, as confirmed by the study of validation group samples (Supplementary Table S4).

Our results suggest that the expression of several genes may impact endometrial aging. For example, TBX15, the most highly upregulated gene in the AMA group, may play an important role in endometrial aging as its expression consistently increased during a woman’s reproductive lifespan (Figs 3B and 4A and B, Supplementary Table S2 and S4). This gene is expressed by epithelial cells and associated with epithelial-to-mesenchymal transition (EMT) (Niu et al., 2022). The upregulation of TBX15 reduces apoptosis through a nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB)-dependent mechanism (Arribas et al., 2016). The EMT is normally induced by embryonic signals, enabling endometrial luminal cells to acquire the characteristics of motile epithelial cells (Owusu-Akyaw et al., 2019); however, its strong activation in the absence of embryos observed in AMA samples may compromise normal epithelial function. We also identified a set of DEGs previously described in association with endometrial receptivity, such as CDA, CD55, CLDN4, GAST LAMB3, and STC1 (Haouzi et al., 2008; Díaz-Gimeno et al., 2011; Altmäe et al., 2017; Enciso et al., 2018) (Fig. 3A, Supplementary Table S2, Supplementary Fig. S2). The disturbance of oestrogen signalling in AMA is supported by the changes in direct oestrogen-responsive genes, such as GRB7, CD55, and CLDN4. Although synthetic progesterone is administered to activate progesterone receptors in the endometrium during HRT, and no differences in progesterone receptor expression were revealed between the groups, a higher proportion of MCCs together with the observed downregulation of some progesterone-regulated genes (e.g. CLDN4, ARID5B, STC1, and C2CD4B) might indicate suboptimal progesterone action in the AMA endometrium (Supplementary Fig. S2). It is unclear whether aging affects the way progesterone acts in the human endometrium, but animal studies suggest the possible effect of aging on progesterone sensitivity (Li et al., 2017).

We have to acknowledge some limitations of our study. While it provides strong evidence that age affects MCC differentiation in the AMA endometrium, this is a retrospective study, and our clinical data are limited. Blood hormone levels were not measured for all participants, and the endometrial pathology data may not be entirely comprehensive. Nevertheless, our study provides considerable evidence that age affects the development of the endometrial epithelium and its functions. The current study is the first to show that older women have a significantly higher proportion of ciliated epithelial cells in their endometrium. The development of epithelial MCCs is strongly associated with cell cycle regulation and can be altered by cellular senescence. Here, we propose that the accumulation of senescent epithelial cells might lead to a shift in the proportions of epithelial cell subpopulations, indicating an increased age-conferred risk for metaplasia in AMA women. However, it remains unclear whether this shift alone can cause implantation failure or whether it exacerbates other factors that compromise endometrial receptivity. Our study also provides evidence that aging does not occur at a certain time point and is specific to every individual. Therefore, a personalized strategy is required to address endometrial aging in clinical practice and the findings of this study contribute significantly to the advancement of fundamental knowledge in this area.

Supplementary Material

hoae048_Supplementary_Data

Acknowledgements

We express gratitude to Dr Nadezhda Milova and Dr Antoan Milov from the Center for Woman’s Health, Bulgaria for their help with sample collection, and Kadi Tilk and Arina Laanemets from the University of Tartu, Estonia for their technical support.

Contributor Information

Marina Loid, Department of Obstetrics and Gynecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia; Competence Centre on Health Technologies, Tartu, Estonia.

Darina Obukhova, Department of Obstetrics and Gynecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia; Department of Clinical Genetics, Maastricht University Medical Centre+, Maastricht, The Netherlands; Department of Genetics and Cell Biology, GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands.

Keiu Kask, Department of Obstetrics and Gynecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia; Competence Centre on Health Technologies, Tartu, Estonia.

Apostol Apostolov, Department of Obstetrics and Gynecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia; Competence Centre on Health Technologies, Tartu, Estonia.

Alvin Meltsov, Competence Centre on Health Technologies, Tartu, Estonia; Department of Clinical Genetics, Maastricht University Medical Centre+, Maastricht, The Netherlands; Department of Genetics and Cell Biology, GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands.

Demis Tserpelis, Department of Clinical Genetics, Maastricht University Medical Centre+, Maastricht, The Netherlands.

Arthur van den Wijngaard, Department of Clinical Genetics, Maastricht University Medical Centre+, Maastricht, The Netherlands.

Signe Altmäe, Department of Biochemistry and Molecular Biology, Faculty of Sciences, University of Granada, Granada, Spain; Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain; Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden.

Galina Yahubyan, Department of Molecular Biology, Faculty of Biology, University of Plovdiv, Plovdiv, Bulgaria.

Vesselin Baev, Department of Molecular Biology, Faculty of Biology, University of Plovdiv, Plovdiv, Bulgaria.

Merli Saare, Department of Obstetrics and Gynecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia; Competence Centre on Health Technologies, Tartu, Estonia.

Maire Peters, Department of Obstetrics and Gynecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia; Competence Centre on Health Technologies, Tartu, Estonia.

Ave Minajeva, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia.

Priit Adler, Faculty of Science and Technology, Institute of Computer Science, University of Tartu, Tartu, Estonia.

Ganesh Acharya, Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden.

Kaarel Krjutškov, Department of Obstetrics and Gynecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia; Competence Centre on Health Technologies, Tartu, Estonia.

Maria Nikolova, Department of Molecular Biology, Faculty of Biology, University of Plovdiv, Plovdiv, Bulgaria; Center for Women’s Health, Plovdiv, Bulgaria.

Felipe Vilella, Research & Medical Department, Carlos Simon Foundation, INCLIVA Health Research Institute, Valencia, Spain.

Carlos Simon, Research & Medical Department, Carlos Simon Foundation, INCLIVA Health Research Institute, Valencia, Spain; Beth Israel Deaconess Medical Center, Harvard University, Boston, MA, USA; Department of Pediatrics, Obstetrics & Gynecology, University of Valencia, Valencia, Spain; Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, USA.

Masoud Zamani Esteki, Department of Clinical Genetics, Maastricht University Medical Centre+, Maastricht, The Netherlands; Department of Genetics and Cell Biology, GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands; Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden.

Andres Salumets, Department of Obstetrics and Gynecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia; Competence Centre on Health Technologies, Tartu, Estonia; Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden.

Supplementary data

Supplementary data are available at Human Reproduction Open online.

Data availability

FASTQ files for bulk RNA-seq and raw count matrices were deposited at the Gene Expression Omnibus under accession code GSE236128. All codes used for the analysis are available on GitHub (https://github.com/darinaobukhova/endo_ageing).

Authors’ roles

M.L. designed the study, performed the experiments, participated in the analysis of the data and graphical visualization of the results, and wrote the manuscript; D.O. performed the bioinformatic analysis and graphical visualization of the results, and participated in the manuscript writing; K.Kask performed the IHC analysis and participated in manuscript writing; A.M. performed the receptivity analysis; D.T. and A.v.d.W. participated in the sequencing experiments and initial data analysis; S.A. participated in the functional analysis; A.A., G.Y., and V.B. performed the miRNA analysis; A.M. performed the endometrial tissue histology analysis; P.A. performed power analysis; M.S., M.P., G.A., K.Krjutškov, and M.N. coordinated and performed the sample collections; F.V. and C.S. participated in the sample collections and reviewed the manuscript; M.Z.E participated in the study design, coordinated the bioinformatic analysis, and reviewed the manuscript; A.S. designed and coordinated the study. All authors have read and approved the manuscript.

Funding

Estonian Research Council (grant no. PRG1076), Horizon 2020 innovation grant (ERIN, grant no. EU952516), Enterprise Estonia (grant no. EU48695), MSCA-RISE-2020 project TRENDO (grant no. 101008193), EU 874867 project HUTER, the Horizon Europe NESTOR grant (grant no. 101120075) of the European Commission, the EVA (Erfelijkheid Voortplanting & Aanleg) specialty program (grant no. KP111513) of Maastricht University Medical Center (MUMC+), MICIU/AEI/10.13039/501100011033 and FEDER, EU projects Endo-Map (grant no. PID2021-12728OB-100), SY (grant no. CNS2022-135999), and the National Science Fund of Bulgaria (grant no. KII-06 H31/2).

Conflict of interest

The authors declare no competing interests.

References

  1. Abdalla HI, Wren ME, Thomas A, Korea L.. Age of the uterus does not affect pregnancy or implantation rates; a study of egg donation in women of different ages sharing oocytes from the same donor. Hum Reprod 1997;12:827–829. [DOI] [PubMed] [Google Scholar]
  2. Aging Atlas Consortium; Liu G-H, Bao Y, Qu J, Zhang W, Zhang T, Kang W. et al. Aging Atlas: a multi-omics database for aging biology. Nucleic Acids Res 2021;49:D825–D830. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Altmäe S, Koel M, Võsa U, Adler P, Suhorutšenko M, Laisk-Podar T, Kukushkina V, Saare M, Velthut-Meikas A, Krjutškov K. et al. Meta-signature of human endometrial receptivity: a meta-analysis and validation study of transcriptomic biomarkers. Sci Rep 2017;7:10077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Altmäe S, Tamm-Rosenstein K, Esteban FJ, Simm J, Kolberg L, Peterson H, Metsis M, Haldre K, Horcajadas JA, Salumets A. et al. Endometrial transcriptome analysis indicates superiority of natural over artificial cycles in recurrent implantation failure patients undergoing frozen embryo transfer. Reprod Biomed Online 2016;32:597–613. [DOI] [PubMed] [Google Scholar]
  5. Arribas J, Cajuso T, Rodio A, Marcos R, Leonardi A, Velázquez A.. NF-κB mediates the expression of TBX15 in cancer cells. PLoS One 2016;11:e0157761. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Bonser LR, Koh KD, Johansson K, Choksi SP, Cheng D, Liu L, Sun DI, Zlock LT, Eckalbar WL, Finkbeiner WE. et al. Flow-cytometric analysis and purification of airway epithelial-cell subsets. Am J Respir Cell Mol Biol 2021;64:308–317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Burger HG, Dudley EC, Hopper JL, Groome N, Guthrie JR, Green A, Dennerstein L.. Prospectively measured levels of serum follicle-stimulating hormone, estradiol, and the dimeric inhibins during the menopausal transition in a population-based cohort of women. J Clin Endocrinol Metab 1999;84:4025–4030. [DOI] [PubMed] [Google Scholar]
  8. Cano F, Simón C, Remohí J, Pellicer A.. Effect of aging on the female reproductive system: evidence for a role of uterine senescence in the decline in female fecundity. Fertil Steril 1995;64:584–589. [DOI] [PubMed] [Google Scholar]
  9. Chen Y, Wang X.. miRDB: an online database for prediction of functional microRNA targets. Nucleic Acids Res 2020;48:D127–D131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Chevalier B, Adamiok A, Mercey O, Revinski DR, Zaragosi L-E, Pasini A, Kodjabachian L, Barbry P, Marcet B.. miR-34/449 control apical actin network formation during multiciliogenesis through small GTPase pathways. Nat Commun 2015;6:8386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. de Magalhães J, Toussaint O.. GenAge: a genomic and proteomic network map of human ageing. FEBS Lett 2004;571:243–247. [DOI] [PubMed] [Google Scholar]
  12. Devesa-Peiro A, Sebastian-Leon P, Parraga-Leo A, Pellicer A, Diaz-Gimeno P.. Breaking the ageing paradigm in endometrium: endometrial gene expression related to cilia and ageing hallmarks in women over 35 years. Hum Reprod 2022;37:762–776. [DOI] [PubMed] [Google Scholar]
  13. Díaz-Gimeno P, Horcajadas JA, Martínez-Conejero JA, Esteban FJ, Alamá P, Pellicer A, Simón C.. A genomic diagnostic tool for human endometrial receptivity based on the transcriptomic signature. Fertil Steril 2011;95:50–60.e15. [DOI] [PubMed] [Google Scholar]
  14. Djerassi A, Coutifaris C, West VA, Asa SL, Kapoor SC, Pavlou SN, Snyder PJ.. Gonadotroph adenoma in a premenopausal woman secreting follicle-stimulating hormone and causing ovarian hyperstimulation. J Clin Endocrinol Metab 1995;80:591–594. [DOI] [PubMed] [Google Scholar]
  15. Enciso M, Carrascosa JP, Sarasa J, Martínez-Ortiz PA, Munné S, Horcajadas JA, Aizpurua J.. Development of a new comprehensive and reliable endometrial receptivity map (ER Map/ER Grade) based on RT-qPCR gene expression analysis. Hum Reprod 2018;33:220–228. [DOI] [PubMed] [Google Scholar]
  16. European Statistical System. 2022. https://ec.europa.eu/eurostat/databrowser/view/tps00017/default/table?lang=en (3 April 2023, date last accessed).
  17. Fischer AH, Jacobson KA, Rose J, Zeller R.. Hematoxylin and eosin staining of tissue and cell sections. CSH Protoc 2008;2008:pdb.prot4986. [DOI] [PubMed] [Google Scholar]
  18. Flamigni C, Borini A, Violini F, Bianchi L, Serrao L.. Oocyte donation: comparison between recipients from different age groups. Hum Reprod 1993;8:2088–2092. [DOI] [PubMed] [Google Scholar]
  19. Garcia-Alonso LUZ, Handfield L-F, Roberts K, Nikolakopoulou K, Fernando RC, Gardner L, Woodhams B, Arutyunyan A, Polanski K, Hoo R. et al. Mapping the temporal and spatial dynamics of the human endometrium in vivo and in vitro. Nat Genet 2021;53:1698–1711. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Glogovitis I, Yahubyan G, Würdinger T, Koppers-Lalic D, Baev V.. miRGalaxy: galaxy-based framework for interactive analysis of microRNA and isomiR sequencing data. Cancers (Basel) 2021;13:5663. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Haider S, Gamperl M, Burkard TR, Kunihs V, Kaindl U, Junttila S, Fiala C, Schmidt K, Mendjan S, Knöfler M. et al. Estrogen signaling drives ciliogenesis in human endometrial organoids. Endocrinology 2019;160:2282–2297. [DOI] [PubMed] [Google Scholar]
  22. Haouzi D, Mahmoud K, Fourar M, Bendhaou K, Dechaud H, De Vos J, Reme T, Dewailly D, Hamamah S.. Identification of new biomarkers of human endometrial receptivity in the natural cycle. Hum Reprod 2008;24:198–205. [DOI] [PubMed] [Google Scholar]
  23. Horree N, Heintz APM, Sie-Go DMDS, Diest P.. p16 is consistently expressed in endometrial tubal metaplasia. Cell Oncol 2007;29:37–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Hu S, Yao G, Wang Y, Xu H, Ji X, He Y, Zhu Q, Chen Z, Sun Y.. Transcriptomic changes during the pre-receptive to receptive transition in human endometrium detected by RNA-Seq. J Clin Endocrinol Metab 2014;99:E2744–E2753. [DOI] [PubMed] [Google Scholar]
  25. Idda ML, McClusky WG, Lodde V, Munk R, Abdelmohsen K, Rossi M, Gorospe M.. Survey of senescent cell markers with age in human tissues. Aging (Albany NY) 2020;12:4052–4066. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Klaes R, Friedrich T, Spitkovsky D, Ridder R, Rudy W, Petry U, Dallenbach-Hellweg G, Schmidt D, Knebel Doeberitz MV.. Overexpression of p16INK4A as a specific marker for dysplastic and neoplastic epithelial cells of the cervix uteri. Int J Cancer 2001;92:276–284. [DOI] [PubMed] [Google Scholar]
  27. Lambalk C. Editor’s choice: Delayed childbearing and medically assisted reproduction. Hum Reprod 2022;37:e1. [DOI] [PubMed] [Google Scholar]
  28. Li M-Q, Yao M-N, Yan J-Q, Li Z-L, Gu X-W, Lin S, Hu W, Yang Z-M.. The decline of pregnancy rate and abnormal uterine responsiveness of steroid hormones in aging mice. Reprod Biol 2017;17:305–311. [DOI] [PubMed] [Google Scholar]
  29. Lyons RA, Saridogan E, Djahanbakhch O.. The reproductive significance of human fallopian tube cilia. Hum Reprod Update 2006;12:363–372. [DOI] [PubMed] [Google Scholar]
  30. Marcet B, Chevalier B, Luxardi G, Coraux C, Zaragosi L-E, Cibois M, Robbe-Sermesant K, Jolly T, Cardinaud B, Moreilhon C. et al. Control of vertebrate multiciliogenesis by miR-449 through direct repression of the Delta/Notch pathway. Nat Cell Biol 2011;13:693–699. [DOI] [PubMed] [Google Scholar]
  31. Martin N, Beach D, Gil J.. Ageing as developmental decay: insights from p16INK4a. Trends Mol Med 2014;20:667–674. [DOI] [PubMed] [Google Scholar]
  32. Masterton R, Armstrong EM, More IAR.. The cyclical variation in the percentage of ciliated cells in the normal human endometrium. J Reprod Fertil 1975;42:537–540. [DOI] [PubMed] [Google Scholar]
  33. Meltsov A, Saare M, Teder H, Paluoja P, Arffman RK, Piltonen T, Laudanski P, Wielgoś M, Gianaroli L, Koel M. et al. Targeted gene expression profiling for accurate endometrial receptivity testing. Sci Rep 2023;13:13959. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Mendaza S, Fernández-Irigoyen J, Santamaría E, Zudaire T, Guarch R, Guerrero-Setas D, Vidal A, Santos-Salas J, Matias-Guiu X, Ausín K. et al. Absence of nuclear p16 is a diagnostic and independent prognostic biomarker in squamous cell carcinoma of the cervix. IJMS 2020;21:2125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Moore L, Leongamornlert D, Coorens THH, Sanders MA, Ellis P, Dentro SC, Dawson KJ, Butler T, Rahbari R, Mitchell TJ. et al. The mutational landscape of normal human endometrial epithelium. Nature 2020;580:640–646. [DOI] [PubMed] [Google Scholar]
  36. More IAR, Masterton RG.. The role of oestrogen in the control of ciliated cells of the human endometrium. J Reprod Fertil 1976;47:19–24. [DOI] [PubMed] [Google Scholar]
  37. Navot D, Drews MR, Bergh PA, Guzman I, Karstaedt A, Scott RT, Garrisi GJ, Hofmann GE.. Age-related decline in female fertility is not due to diminished capacity of the uterus to sustain embryo implantation. Fertil Steril 1994;61:97–101. [DOI] [PubMed] [Google Scholar]
  38. Niu G, Hao J, Sheng S, Wen F.. Role of T‐box genes in cancer, epithelial‐mesenchymal transition, and cancer stem cells. J Cell Biochem 2022;123:215–230. [DOI] [PubMed] [Google Scholar]
  39. Noyes RW, Hertig AT, Rock J.. Dating the endometrial biopsy. Fertil Steril 1950;1:3–25. [DOI] [PubMed] [Google Scholar]
  40. Onogi S, Ezoe K, Nishihara S, Fukuda J, Kobayashi T, Kato K.. Endometrial thickness on the day of the LH surge: an effective predictor of pregnancy outcomes after modified natural cycle-frozen blastocyst transfer. Hum Reprod Open 2020;2020:hoaa060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Owusu-Akyaw A, Krishnamoorthy K, Goldsmith LT, Morelli SS.. The role of mesenchymal–epithelial transition in endometrial function. Hum Reprod Update 2019;25:114–133. [DOI] [PubMed] [Google Scholar]
  42. Parvanov D, Ganeva R, Vidolova N, Stamenov G.. Decreased number of p16-positive senescent cells in human endometrium as a marker of miscarriage. J Assist Reprod Genet 2021;38:2087–2095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Perkins KM, Boulet SL, Jamieson DJ, Kissin DM, National Assisted Reproductive Technology Surveillance System (NASS) Group. Trends and outcomes of gestational surrogacy in the United States. Fertil Steril 2016;106:435–442.e2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Reimand J, Kull M, Peterson H, Hansen J, Vilo J.. g:Profiler—a web-based toolset for functional profiling of gene lists from large-scale experiments. Nucleic Acids Res 2007;35:W193–W200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Ren X, Kuan PF.. RNAAgeCalc: a multi-tissue transcriptional age calculator. PLoS One 2020;15:e0237006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Sacchi L, Albani E, Cesana A, Smeraldi A, Parini V, Fabiani M, Poli M, Capalbo A, Levi-Setti PE.. Preimplantation genetic testing for aneuploidy improves clinical, gestational, and neonatal outcomes in advanced maternal age patients without compromising cumulative live-birth rate. J Assist Reprod Genet 2019;36:2493–2504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Sanders KD, Silvestri G, Gordon T, Griffin DK.. Analysis of IVF live birth outcomes with and without preimplantation genetic testing for aneuploidy (PGT-A): UK Human Fertilisation and Embryology Authority data collection 2016–2018. J Assist Reprod Genet 2021;38:3277–3285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Schindelin J, Arganda-Carreras I, Frise E, Kaynig V, Longair M, Pietzsch T, Preibisch S, Rueden C, Saalfeld S, Schmid B. et al. Fiji: an open-source platform for biological-image analysis. Nat Methods 2012;9:676–682. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Sigurgeirsson B, Åmark H, Jemt A, Ujvari D, Westgren M, Lundeberg J, Gidlöf S.. Comprehensive RNA sequencing of healthy human endometrium at two time points of the menstrual cycle. Biol Reprod 2017;96:24–33. [DOI] [PubMed] [Google Scholar]
  50. Soares SR, Troncoso C, Bosch E, Serra V, Simón C, Remohí J, Pellicer A.. Age and uterine receptiveness: predicting the outcome of oocyte donation cycles. J Clin Endocrinol Metab 2005;90:4399–4404. [DOI] [PubMed] [Google Scholar]
  51. Song R, Walentek P, Sponer N, Klimke A, Lee JS, Dixon G, Harland R, Wan Y, Lishko P, Lize M. et al. miR-34/449 miRNAs are required for motile ciliogenesis by repressing cp110. Nature 2014;510:115–120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Suhorutshenko M, Kukushkina V, Velthut-Meikas A, Altmäe S, Peters M, Mägi R, Krjutškov K, Koel M, Codoñer FM, Martinez-Blanch JF. et al. Endometrial receptivity revisited: endometrial transcriptome adjusted for tissue cellular heterogeneity. Hum Reprod 2018;33:2074–2086. [DOI] [PubMed] [Google Scholar]
  53. Tasca A, Helmstädter M, Brislinger MM, Haas M, Mitchell B, Walentek P.. Notch signaling induces either apoptosis or cell fate change in multiciliated cells during mucociliary tissue remodeling. Dev Cell 2021;56:525–539.e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Toner JP, Grainger DA, Frazier LM.. Clinical outcomes among recipients of donated eggs: an analysis of the U.S. national experience, 1996–1998. Fertil Steril 2002;78:1038–1045. [DOI] [PubMed] [Google Scholar]
  55. Vassilieva IO, Reshetnikova GF, Shatrova AN, Tsupkina NV, Kharchenko MV, Alekseenko LL, Nikolsky NN, Burova EB.. Senescence-messaging secretome factors trigger premature senescence in human endometrium-derived stem cells. Biochem Biophys Res Commun 2018;496:1162–1168. [DOI] [PubMed] [Google Scholar]
  56. 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. [DOI] [PubMed] [Google Scholar]
  57. Wu H, Wang C, Wu Z.. PROPER: comprehensive power evaluation for differential expression using RNA-seq. Bioinformatics 2015;31:233–241. [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

hoae048_Supplementary_Data

Data Availability Statement

FASTQ files for bulk RNA-seq and raw count matrices were deposited at the Gene Expression Omnibus under accession code GSE236128. All codes used for the analysis are available on GitHub (https://github.com/darinaobukhova/endo_ageing).


Articles from Human Reproduction Open are provided here courtesy of Oxford University Press

RESOURCES