Skip to main content
Erschienen in: Reproductive Biology and Endocrinology 1/2014

Open Access 01.12.2014 | Research

Endometrial gene expression reveals compromised progesterone signaling in women refractory to embryo implantation

verfasst von: Alejandro Tapia-Pizarro, Paula Figueroa, Julio Brito, Juan Carlos Marín, David J Munroe, Horacio B Croxatto

Erschienen in: Reproductive Biology and Endocrinology | Ausgabe 1/2014

Abstract

Background

Endometrial function is essential for embryo implantation. The aim of this study was to analyze gene expression profiles from individual endometrial samples obtained from women with repeated implantation failure after IVF in oocyte donation programs.

Methods

Seventeen volunteers were recruited: women who had previously participated as recipients in oocyte donation cycles and repeatedly exhibited implantation failure (Group A, study group, n = 5) or had at least one successful cycle (Group B, control group, n = 6) and spontaneously fertile women (Group C, normal fertility group, n = 6). An endometrial cycle was induced with exogenous estradiol (E2) and progesterone (P) and an endometrial sample was collected on the seventh day of P treatment.

Results

Transcriptome analysis showed 82 genes with consistent differential gene expression when comparing A vs. B and A vs. C. One hundred transcripts differentially expressed in group A vs. B have been shown to be regulated by P, suggesting compromised P signaling in the endometrium. The P receptor (PR) mutation PROGINS was not detected in women from group A. Semi-quantitation of immunoreactive PRA/B, PRB and Sp1 (a transcription factor related to P signaling) in paraffin-embedded endometrial sections, did not show statistically significant differences amongst groups. However immunostaining glycodelin was significantly decreased in endometrial samples from group A

Conclusions

We conclude that some cases of repeated implantation failure could be associated with an aberrant gene expression profile. Compromised P signaling might be the underlying mechanism for such endometrial gene expression deregulation in women with repeated implantation failure.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1477-7827-12-92) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

AT conceived the study, participated in its design, carried out the microarrays and bioinformatic analyses and helped to draft the manuscript. PF and JB participated in PROGINS detection. J.C.M participated in PROGINS detection, data analysis and interpretation. D.J.M. participated in microarrays analysis. HBC conceived the study, participated in its design, data interpretation and helped to draft the manuscript. All authors read and approved the final manuscript.
Abkürzungen
DAVID
Database for annotation visualization and integrated discovery
E2
Estradiol
ELS
Expression level score
GATHER
Gene annotation tool to help explain relationships
IHC
Immunohistochemistry
mAdb
National cancer institute’s microarrays data base webtool
P
Progesterone
PAEP
Progestagen-associated endometrial protein
PCA
Principal component analysis
Pgr
Progesterone receptor gene
PR
Progesterone receptor
PR-A/B
Progesterone receptor isoforms A and B
PR-B
Progesterone receptor isoform B
PREs
Progesterone-response elements
PROGINS
Alu insertion in intron G of the progesterone receptor gene
RFLP
Restriction fragment length polymorphism
RT-PCR
Reverse transcription coupled to polymerase chain reaction
Sp1
Specificity Protein 1..

Background

Cellular and molecular events in the uterine milieu that lead to successful blastocyst implantation are required in the endometrium to become receptive and ready for implantation. Acquisition of receptivity is driven by estradiol (E2) and progesterone (P), which acting through their receptors, change the transcription rate of target genes [1]. Particularly, the postovulatory rise in P triggers a sequence of highly coordinated responses beginning with the detention of the estrogen-induced epithelial cell proliferation and followed by the transformation to a secretory phenotype of the gland, recruitment of leukocytes and angiogenesis [2]. The P action is mediated primarily through binding to and activation of its cognate receptors; the full length B- and N-terminally truncated A isoforms of the P receptor (PR), classically defined as ligand-activated transcription factors [3]. Upon exposure to P, the ligand-activated receptor can directly interact with specific P-response elements (PREs) in the promoter regions of target genes. It is accepted that P acts on an estrogen primed endometrium to initiate a pattern of gene expression important for achievement of receptivity and an altered PR signaling has been associated with human endometrial dysfunction [4]. The antiprogestin mifepristone binds to the PR with high affinity blocking the biological effects of P. In women, the administration of a single dose of oral mifepristone (200 mg) during the secretory phase of the cycle rapidly renders the endometrium unreceptive and modifies gene expression in the uterus within 6 h of administration [57].
The development of microarray technology has led to many large-scale gene expression profiling studies of human endometrium [810]. Although there seems to be very few consensus genes that have been identified across similar studies [11], collectively they demonstrate that a multitude of genes are associated with the endometrial transcriptome, whose regulation for the acquisition of the receptive phenotype is ultimately driven by P. The approach our group has used to identify the endometrial receptivity transcript profile in a previous report from our laboratory [12] was to analyze endometrial tissue obtained from women during a mock hormonal treatment cycle for oocyte donation as a recipient. The endometrial samples are collected during the time interval corresponding to the window of implantation [13] comparing gene expression profiles from women who were refractory to implantation and those who achieved pregnancy in previous oocyte donation cycles [12]. Although this previous study provided interesting insights to endometrial gene expression associated with implantation failure, the microarrays analysis was performed with only 3(from a total of 5) samples from women with repeated embryo implantation failure that were pooled and using a microarrays platform that examined only one-third of the human genome; providing only a partial view of the whole picture.
The aim of the present report was to examine the individual gene expression profiles in the endometrium from women with implantation failure and compare them with those obtained from fertile women in order to identify compromised transcripts and pathways in the infertile group. We used a microarrays platform for complete coverage of the human genome and bioinformatics tools for data interpretation. Here we report that several transcripts, whose expression level is aberrant in the infertile group, have been described as regulated by P and are related to immune function.

Methods

Subjects

This study was approved by the Ethics Review Committee for Investigations in Human Beings of Faculty of Medicine, University of Chile: protocol No. 093–2008, approved 12-29-2008, initiated 01-05-2009 finished 03-31-2014. Each volunteer participating read and signed the informed consent approved by the respective Ethics Review Committee.
Three groups of women were recruited as has been described elsewhere [12]. Group A (n = 5) consisted of women that had never been pregnant and had previously participated in two or more cycles as recipients in an oocyte donation program with no evidence of embryo implantation. Male partners had normal seminal parameters and transferred embryos had good morphology, at least equivalent to embryos transferred to the oocyte donor who became pregnant. Since good quality embryos with the ability to implant and develop normally derive from good quality oocytes, it was required that the oocyte donor had become pregnant from the same oocyte pool. Women from group A were recruited within 3 years following the last failed cycle. Group B (n = 6) comprised of women who became pregnant as recipients in previous oocyte donation cycles and delivered live infants. Group C (n = 6) included normal fertile women who conceived in natural cycles and had three or more live births and had elective tubal ligation at least 1 year prior to their participation for reasons unrelated to this study. Women from groups B and C were recruited within 5 years following the last successful pregnancy. The general exclusion criteria for all volunteers included: metabolic or endocrine diseases other than those leading to ovarian failure, chronic use of medication other than HRT, polycystic ovary syndrome, drug abuse, obesity, endometriosis, pelvic inflammatory disease and current genital tract infection. Age and body mass index from recruited women as well as the plasma P and endometrial thickness measured on the day of the endometrial collection are shown in Table 1.
Table 1
Characteristics of women participating in the study and parameters evaluated during the hormonal replacement cycle
 
Group A (n = 5)
Group B (n = 6)
Group C (n = 6)
P value
Age (years)
35.4 (26–43)
41.9 (34–46)
41.3 (36–47)
0.1117
Body mass index
25.2 (22.6-29.4)
25.6 (22.5-27.4)
25.4 (23.3-28.1)
0.573
Plasma progesterone* (nmol/L)
75.7 (38–122)
88.1 (36–192)
63.7 (43.3-75)
0.7601
Endometrial thickness* (mm)
10.8 (9–12)
9.1 (8–10)
11.2 (8.5-12.5)
0.6162
*On the day of endometrial collection.

Induction of endometrial cycle

All subjects underwent the induction of an artificial endometrial cycle with exogenous ethinyl E2 for 20 days and for the last 7 days, this treatment was administered concomitantly with micronized P as described previously [12]. An endometrial sample was obtained on 20th day of the endometrial cycle. One part of the specimen was snap frozen in liquid nitrogen and kept at -80 ° C until use and the remaining portion was fixed in paraformaldehyde for histological dating, according to the criteria of Noyes et al., [14] and for immunohistochemistry (IHC) studies. All biopsies were classified as normal secretory endometrium with no signs of inflammatory processes.

Gene expression profiling

Total RNA was extracted from frozen endometrial tissue samples using Trizol (Invitrogen, Gaithersburg, MD, USA) as directed by the manufacturer and then checked for yield and quality as described before [12]. The Human Genome U133 plus 2.0 GeneChip oligonucleotide microarrays (Affymetrix, Sunnyvale, CA, USA); corresponding to 47,000 transcripts and variants, including 38,500 well-characterized human genes, was used for gene expression analysis according to the manufacturer’s instruction.
Microarrays Data Analysis: Replicate hybridizations were performed for each RNA sample and raw data obtained from the GeneChip Microarray Suite v 1.4 was subsequently analyzed using the National Cancer Institute’s Microarrays Data Base webtool (mAdb) (http://​nciarray.​nci.​nih.​gov).

Statistical analyses of microarrays data

Significant genes were defined as ≥2, p-value < 0.001 and a false discovery rate (FDR) < 0.1 [15]. T-test was performed to determine statistical differences and from the significant genes identified, Venn diagrams were constructed to identify coincident transcripts.

Hierarchical clustering

Was performed based on uncentered correlations with average linkage clustering using mAdb. The resulting dendogram allows data structure visualization of endometrial samples according to total gene expression, revealing samples with similar patterns of gene expression and relationships between the specimens.

Principal component analysis (PCA)

Was performed for simplifying the large amount of data derived from microarray analysis [16]. We applied the unbiased PCA algorithm to all samples using all transcripts analyzed with the microarray chip to look for expression patterns and underlying cluster structures of endometrial samples.

Functional clustering

To increase the effectiveness of DNA microarray analysis, data sets of differentially expressed genes from the comparison between A vs. B and A vs. C were intersected to define those transcripts consistently up- or down-regulated and combined with external data sources, such as gene annotation, in order to associate the expression patterns of this particular set of genes with the biological processes that they may represent. In our analysis, we submitted our gene lists to the web-based tools DAVID (Database for Annotation, Visualization and Integrated Discovery) [17] and GATHER (Gene Annotation Tool to Help Explain Relationships) [18] for functional annotation analysis in order to gain an in-depth understanding of their biological themes, which otherwise would require laborious and somewhat subjective manual literature searches.

DAVID

Up- and down-regulated genes were submitted to DAVID database for systematically extracting biological meaning for them by retrieving pathway maps from the Kyoto Encyclopedia of Genes and Genomes (KEGG) [19] and Biocarta pathways database (http://​www.​biocarta.​com/​genes/​index.​asp) along with Gene Ontology (GO) functional annotations from Entrez Gene [20]. The parameters of the “Functional Annotation Clustering” (a part of the “Functional Annotation Tool”) were set to the highest level of stringency in order to obtain the smallest number of maps. The DAVID database associates each annotation to a gene group using a contingency table representation and calculates its significance.

GATHER

Regulated genes were submitted as well to GATHER database (http://​gather.​genome.​duke.​edu/​) that contains the GO annotations and KEGG pathways. The GATHER database associates each group of transcripts with the same functional annotation and calculates a Bayes factor [18] which is a measure of the strength of the evidence supporting an association of an annotation with the submitted gene list. We have selected a low Bayes factor (≥3) for presenting the preponderant evidences for associations.

Immunohistochemistry (IHC)

A portion of each endometrial sample was fixed in paraformaldehyde, included in paraffin blocks and 5 μm sections were prepared. PR-A/B, PR-B, glycodelin and Specificity protein 1 (Sp1) were evaluated by IHC in the endometrial samples using the antibodies and dilutions shown in Table 2 and the broad spectrum Histostain-SP kit (Life Technologies, Carlsbad, CA, USA) as described previously [21]. Immunoreactive PRA/B, PRB, Sp1 and glycodelin in endometrial sections was semi-quantified using the expression level score (ELS), calculated by means of Image Pro Plus software (Media Cybernetics Rockville, MD, USA) as described previously [21]. Briefly, ELS = Mean Optical Density of immunostaining x Percent Area Positively Stained x 100.
Table 2
Antibodies and dilutions used for immunohistochemistry
Antibody
Source
Dilution
Progesterone receptor (PR)-A/B
Santa Cruz Biotech. (sc-810)
1:50
PR-B
Novocastra (NCL-PGR-B)
1:100
Glycodelin
H. Koistinen [22]
1:1000
Specificity protein-1 (Sp1)
Santa Cruz Biotech. (sc-14027)
1:100

DNA isolation and PROGINSdetection

Genomic DNA was isolated from leukocytes derived from peripheral blood obtained by venipuncture using the PAXgene Blood DNA Validation kit (Qiagen, Valencia, CA, USA) following the manufacturer’s protocol. The detection of Alu insertion in intron G and restriction fragment length polymorphism (RFLP) analysis in exon 5 to confirm the presence of PROGINS mutation was performed as described by Pisarska et al. [23].

Results

Gene expression profile analysis

Women with implantation failure (group A, n = 5), women with 2 or more livebirths conceived either by oocyte donation (group B, n = 6) or naturally (group C, n = 6) were subjected to an oocyte donation mock cycle as recipients and on the seventh day of P administration an endometrial sample was obtained. Total RNA was extracted from each tissue sample and used to individually probe the HG_U133 plus 2.0 human gene microarray comprising of 54,675 genes and expressed sequence tags.
We performed PCA for all the endometrial samples analyzed using their respective gene expression profiles for their representation on a three-dimensional graphic (Figure 1A). Each point in a PCA graph represents the gene expression profile of an endometrial sample and the distance between two plotted points is proportional to the degree of similarity between the gene expression profiles. The PCA plot comprising of a projection on the first three principal components, which together explain 48.8% (21%, 14%, and 13%) of the total variance, showed that endometrial samples from infertile subjects clustered apart from samples belonging to the control groups. In addition, gene expression profiles from endometrial samples obtained from microarray analysis were subjected to unsupervised hierarchical clustering analysis in order to generate a dendogram, which is a tree-structured graph that illustrates the similarities in gene expression profiles between endometrial samples from all groups. The dendogram obtained displayed a striking segregation of samples into two major clustering branches, corresponding to the implantation failure group (Group A) and the successful implantation groups (Groups B and C, Figure 1B).
The microarrays data analysis revealed that 747 transcripts were down-regulated in group A compared to group B; whereas 218 transcripts were up-regulated (Figure 1C and 1D). When group A was compared to group C, 156 and 884 transcripts were decreased and increased respectively in group A (Figure 1C and 1D). Only 31 and 51 transcripts down- and up-regulated respectively were common when comparing group A with the control groups B and C (Figure 1C and 1D). The lists of common differentially expressed genes in the comparison of gene expression profiles from group A vs. Group B and Group A vs. group C is in Table 3 for down- and Table 4 for up-regulated transcripts. An independent validation by Real Time RT-PCR for some of the differentially expressed genes found in the samples used in this study has been reported elsewhere [12], supporting our microarrays findings.
Table 3
Genes whose transcript level was down-regulated in Group A (n = 5) when compared with Group B (n = 6) and Group C (n = 6) in the microarray analyses
UniGene ID
Gene symbol
Gene title
A vs. B
p value
A vs. C
p value
Average
Hs.699841
IGHA1
Immunoglobulin heavy constant alpha 1
0.1
0.00086
0.08
0.00043
0.09
Hs.436657
CLU
Clusterin
0.11
0.00043
0.17
0.00013
0.14
Hs.356624
NID1
Nidogen 1
0.19
0.00013
0.14
0.00022
0.16
Hs.82071
CITED2
Cbp/p300-interacting transactivator. with Glu/Asp-rich carboxy-terminal domain. 2
0.15
0.00086
0.19
0.00043
0.17
Hs.532325
PAEP
Progestagen-associated endometrial protein (PAEP)
0.09
0.00086
0.29
0.00086
0.19
Hs.38972
TSPAN1
Tetraspanin 1
0.14
0.00022
0.24
0.00086
0.19
Hs.445705
RRM1
Ribonucleotide reductase M1
0.18
0.00043
0.23
0.00022
0.2
Hs.1012
C4BPA
Complement component 4 binding protein. alpha
0.21
0.00043
0.22
0.00043
0.22
Hs.513261
HN1L
Hematological and neurological expressed 1-like
0.18
0.00022
0.35
0.00043
0.26
Hs.80658
UCP2
Uncoupling protein 2 (mitochondrial. proton carrier)
0.1
0.00043
0.49
0.00013
0.29
Hs.502989
UNC93B1
Unc-93 homolog B1 (C. elegans)
0.23
0.00013
0.37
0.00043
0.3
Hs.414099
CNPY3
Canopy 3 homolog (zebrafish)
0.29
0.00013
0.32
0.00022
0.31
Hs.110571
GADD45B
Growth arrest and DNA-damage-inducible. beta
0.2
0.00022
0.44
0.00086
0.32
Hs.320151
AGPAT2
1-acylglycerol-3-phosphate O-acyltransferase 2 (lysophosphatidic acid acyltransferase. beta)
0.23
0.00086
0.43
0.00013
0.33
Hs.77422
PLP2
Proteolipid protein 2 (colonic epithelium-enriched)
0.26
0.00043
0.42
0.00043
0.34
Hs.1497
RARG
Retinoic acid receptor. gamma
0.33
0.00043
0.35
0.00043
0.34
Hs.389700
MGST1
Glutathione S-transferase. microsomal
0.4
0.00022
0.29
0.00043
0.34
Hs.292078
LARP1
La ribonucleoprotein domain family. member 1
0.34
0.00043
0.36
0.00086
0.35
Hs.334587
RBPMS
RNA binding protein with multiple splicing
0.29
0.00086
0.43
0.00022
0.36
Hs.5298
ADIPOR1
Adiponectin receptor 1
0.3
0.00022
0.42
0.00013
0.36
Hs.439894
CASZ1
Castor zinc finger 1
0.24
0.00022
0.49
0.00086
0.37
Hs.371727
SCNN1G
Sodium channel. nonvoltage-gated 1. gamma
0.29
0.00013
0.45
0.00043
0.37
Hs.474596
LIMK2
LIM domain kinase 2
0.26
0.00043
0.5
0.00013
0.38
Hs.459940
LITAF
Lipopolysaccharide-induced TNF factor
0.37
0.00022
0.39
0.00086
0.38
Hs.442449
CHST14
Carbohydrate (N-acetylgalactosamine 4–0) sulfotransferase 14
0.42
0.00086
0.35
0.00086
0.38
Hs.518525
GLUL
Glutamate-ammonia ligase
0.42
0.00043
0.48
0.00086
0.45
Hs.119177
ARF3
ADP-ribosylation factor 3
0.47
0.00013
0.45
0.00043
0.46
Hs.497417
KIAA0317
KIAA0317
0.49
0.00086
0.44
0.00013
0.46
Hs.501728
RHOG
Ras homolog gene family. member G (rho G)
0.46
0.00043
0.47
0.00043
0.47
Hs.414614
SCNN1B
Sodium channel. nonvoltage-gated 1. beta
0.48
0.00013
0.47
0.00022
0.47
Hs.436896
POLR3A
Polymerase (RNA) III (DNA directed) polypeptide A. 155 kDa
0.48
0.00043
0.47
0.00043
0.48
Data includes genes with decreased transcript levels displaying a ≥2-fold difference in average A vs. B and A vs. C.
Table 4
Genes whose transcript level was up-regulated in Group A (n = 5) when compared with Group B (n = 6) and Group C (n = 6) in the microarray analyses
UniGene ID
Gene symbol
Gene title
A vs. B
p value
A vs. C
p value
Average
Hs.35086
USP1
Ubiquitin specific protease 1 (USP1), mRNA.
42.52
0.00022
44.32
0.00043
43.42
Hs.436977
SYTL3
Synaptotagmin-like 3
20.68
0.00043
26.72
0.00013
23.70
Hs.133421
LIFR
Leukemia inhibitory factor receptor
43.71
0.00086
3.32
0.00022
23.52
Hs.160211
THRAP3
Thyroid hormone receptor associated protein 3 (THRAP3), mRNA.
29.04
0.00013
3.66
0.00043
16.35
Hs.532399
ZC3H11A
KIAA0663 gene product (KIAA0663), mRNA.
4.38
0.00043
28.05
0.00086
16.21
Hs.652169
PLGLB2
Plasminogen-like B2
21.71
0.00086
9.45
0.00086
15.58
Hs.524809
CLIP1
Restin (Reed-Steinberg cell-expressed intermediate filament-associated protein) (RSN), transcript variant 2, mRNA.
2.19
0.00043
25.81
0.00043
14.00
Hs.16355
MYH10
Myosin, heavy polypeptide 10, non-muscle (MYH10), mRNA.
2.6
0.00013
24.08
0.00022
13.34
Hs.502829
SF1
Splicing factor 1 (SF1), transcript variant 4, mRNA.
21.86
0.00043
4.08
0.00086
12.97
Hs.517949
MAP4
Microtubule-associated protein 4 (MAP4), transcript variant 1, mRNA.
10.41
0.00043
11.96
0.00013
11.18
Hs.8118
SMCHD1
KIAA0650 protein
18.77
0.00013
2.17
0.00043
10.47
Hs.514806
GALNT1
UDP-N-acetyl-alpha-D-galactosamine:polypeptide N-acetylgalactosaminyltransferase 1 (GalNAc-T1) (GALNT1), mRNA.
11
0.00043
9.38
0.00043
10.19
Hs.130293
LUC7L3
Cisplatin resistance-associated overexpressed protein (CROP), transcript variant 2, mRNA.
6.32
0.00043
11.88
0.00022
9.10
Hs.143728
WASL
Wiskott-Aldrich syndrome-like (WASL), mRNA.
2.07
0.00022
14.32
0.00086
8.20
Hs.532082
IL6ST
Interleukin 6 signal transducer (gp130, oncostatin M receptor) (IL6ST), transcript variant 2, mRNA.
2.95
0.00013
13
0.00043
7.97
Hs.2913
EPHB3
EphB3 = HEK2 = tyrosine kinase receptor = large erk kinase
11.39
0.00086
4.41
0.00086
7.90
Hs.431081
USP53
Ubiquitin specific protease 53
2.23
0.00013
13.55
0.00043
7.89
Hs.194726
BAG4
BCL2-associated athanogene 4 (BAG4), mRNA.
11.71
0.00013
3.16
0.00086
7.44
Hs.464971
PIK3C3
Phosphoinositide-3-kinase, class 3
3.92
0.00043
10.41
0.00086
7.16
Hs.9997
SECISBP2L
KIAA0256 gene product (KIAA0256), mRNA.
5.43
0.00086
7.36
0.00013
6.39
Hs.497788
EPRS
Glutamyl-prolyl-tRNA synthetase (EPRS), mRNA.
4.41
0.00043
8.11
0.00043
6.26
Hs.101014
CEP57
Translokin (KIAA0092), mRNA.
5.03
0.00086
7.31
0.00086
6.17
Hs.143600
GOLIM4
Golgi phosphoprotein 4 (GOLPH4), mRNA.
2.46
0.00043
9.85
0.00022
6.16
Hs.24485
SMC3
Chondroitin sulfate proteoglycan 6 (bamacan) (CSPG6), mRNA.
2.53
0.00022
9.65
0.00043
6.09
Hs.193832
GPATCH4
G patch domain containing 4 (GPATC4), transcript variant 3, mRNA.
3.32
0.00013
7.94
0.00086
5.63
Hs.406695
PRDM7
PR domain containing 7 (PRDM7), mRNA.
2.04
0.00086
8.46
0.00086
5.25
Hs.42194
SPCS3
Signal peptidase complex subunit 3 homolog (S. cerevisiae) (SPCS3), mRNA.
2.1
0.00043
8.34
0.00022
5.22
Hs.458418
KIAA1731
PREDICTED: KIAA1731 protein (KIAA1731), mRNA.
2.04
0.00013
8.4
0.00086
5.22
Hs.49853
CCAR1
Cell division cycle and apoptosis regulator 1
2.75
0.00086
7.67
0.00013
5.21
Hs.496414
ATP7A
ATPase, Cu++ transporting, alpha polypeptide (Menkes syndrome)
2.89
0.00086
7.16
0.00043
5.02
Hs.481181
NEK1
NIMA (never in mitosis gene a)-related kinase 1 (NEK1), mRNA.
3.07
0.00086
5.35
0.00043
4.21
Hs.440833
PKN2
protein kinase N2 (PKN2), mRNA.
6.02
0.00022
2.14
0.00043
4.08
Hs.524009
AASDHPPT
Aminoadipate-semialdehyde dehydrogenase-phosphopantetheinyl transferase
3.89
0.00013
4.2
0.00086
4.04
Hs.26904
SEC63
SEC63 homolog (S. cerevisiae)
2.95
0.00086
3.78
0.00043
3.37
Hs.93485
SCN2A
MRNA; cDNA DKFZp761D191 (from clone DKFZp761D191)
2.08
0.00043
4.5
0.00013
3.29
Hs.31082
TMEM33
Transmembrane protein 33
2.19
0.00086
4.35
0.00043
3.27
Hs.371372
CWC27
Serologically defined colon cancer antigen 10 (SDCCAG10), mRNA.
2.95
0.00043
3.51
0.00086
3.23
Hs.523299
EIF3A
Eukaryotic translation initiation factor 3, subunit 10 theta, 150/170 kDa (EIF3S10), mRNA.
2.68
0.00022
3.58
0.00022
3.13
Hs.440320
CUL5
Cullin 5 (CUL5), mRNA.
2.6
0.00043
3.25
0.00043
2.93
Hs.203965
PHTF2
Putative homeodomain transcription factor 2
3.56
0.00013
2.3
0.00086
2.93
Hs.335068
TGS1
Nuclear receptor coactivator 6 interacting protein (NCOA6IP), mRNA.
3.63
0.00086
2.16
0.00013
2.89
Hs.189075
TWF1
Twinfilin, actin-binding protein, homolog 1 (Drosophila)
2.36
0.00086
2.97
0.00043
2.67
Hs.127310
UHMK1
U2AF homology motif (UHM) kinase 1 (UHMK1), mRNA.
2.85
0.00086
2.46
0.00043
2.66
Hs.430849
OSBPL8
Oxysterol binding protein-like 8 (OSBPL8), transcript variant 1, mRNA.
2.03
0.00022
3.05
0.00022
2.54
Hs.150557
KLF9
Basic transcription element binding protein 1 (BTEB1), mRNA.
2.6
0.00086
2.39
0.00043
2.50
Hs.210850
HECTD1
HECT domain containing 1 (HECTD1), mRNA.
2.35
0.00013
2.6
0.00086
2.47
Hs.142442
HP1BP3
Heterochromatin protein 1, binding protein 3
2.08
0.00086
2.41
0.00013
2.25
Hs.369284
ESF1
Chromosome 20 open reading frame 6 (C20orf6), mRNA.
2.13
0.00013
2.3
0.00086
2.21
Hs.119023
SMC2
SMC2 structural maintenance of chromosomes 2-like 1 (yeast) (SMC2L1), mRNA.
2.25
0.00022
2
0.00022
2.13
Hs.481927
NIPBL
Nipped-B homolog (Drosophila)
2.11
0.00043
2.04
0.00086
2.08
Hs.374201
KIF21A
kinesin family member 21A (KIF21A), mRNA.
2.13
0.00013
2
0.00013
2.06
Data includes genes with increased transcript levels displaying a ≥2-fold difference in average A vs. B and A vs. C.

Functional associations of transcripts dysregulated in group A vs. control groups

In order to gain further understanding of the potential functional roles of dysregulated endometrial transcripts from group A, we obtained the functional annotations from each gene and determined the enriched processes associated to them from two different web-based tools. Within the down-regulated transcripts, the functional classifications immune response and complement activation, classical pathway were found to be statistically over-represented using the web based applications DAVID and GATHER respectively (p < 0.01). The Bayes factor obtained with the analysis using the GATHER database was 3, which indicates that the association of this particular function with the total of the transcripts in our gene list is weak. The up-regulated transcript list was not enriched with transcripts related to a particular function.

P-regulated genes in women with implantation failure (group A) vs. control (group B)

We reasoned that the endometrium of women from group A might have a dysregulation in P-regulated transcripts as it has been described for endometriosis and also these genes might be coincident with those whose expression in the endometrium is altered upon treatment with the PR antagonist mifepristone. Since women from groups A and B only differ on the embryo implantation outcome, the list of dysregulated transcripts in group A vs. group B during the receptive phase of the endometrium was selected. Within this repertoire, we searched for those genes known to be regulated in normal cycling endometrium by P as it has been described before [4]. For that we accounted for those transcripts that, directed by P, get regulated for the acquisition of endometrial receptivity [2432] and/or dysregulated in conditions that render the endometrium with an unreceptive phenotype (i.e., endometriosis and mifepristone treatment) and that intersected with our list of up and down regulated genes (i.e., A vs. B). We considered only those that had the opposite regulation compared with receptive endometrium, and same regulation in endometrium from women with compromised P signaling in the endometrium such as treated with mifepristone [7] and/or from women with endometriosis [4]. We found 14 and 86 up- and down-regulated genes respectively in the endometrium during the receptive period of women with implantation failure vs. control group B (Tables 5 and 6).
Table 5
Genes previously described to be progesterone regulated that are down-regulated in endometrium of subjects with repeated embryo implantation failure
UniGene ID
Gene symbol
Gene title
Up regulated in window of implantation
Down regulated in endometriosis or mifepristone
Fold change
p value
Hs.386793
GPX3
Glutathione peroxidase 3 (plasma) (GPX3), mRNA.
[25, 27, 29, 32]
 
0.01
0.00013
Hs.458355
C1S
Complement component 1, s subcomponent, transcript variant 1, mRNA.
[28, 29]
 
0.02
0.00043
Hs.647023
CLDN3
Claudin 3
[24]
 
0.07
0.00086
Hs.89603
MUC1
Mucin 1, transmembrane, mRNA.
[25]
[4]
0.11
0.00022
Hs.436657
CLU
Clusterin (complement lysis inhibitor, SP-40,40, sulfated glycoprotein 2, testosterone-repressed prostate message 2, apolipoprotein J), transcript variant 1, mRNA.
[27, 29, 31, 32]
 
0.12
0.00086
Hs.276770
CD52
CD52 molecule
 
[7]
0.14
0.00043
Hs.498173
SMPD1
Sphingomyelin phosphodiesterase 1, acid lysosomal (acid sphingomyelinase), transcript variant 1, mRNA.
[24, 26]
 
0.14
0.00013
Hs.523414
LOC492304
Putative insulin-like growth factor II associated protein, mRNA.
[27]
 
0.15
0.00022
Hs.532325
PAEP
Progestagen-associated endometrial protein
[24, 25, 27, 31, 32]
[39]
0.15
0.00086
Hs.590970
AXL
AXL receptor tyrosine kinase
[24]
 
0.15
0.00022
Hs.163893
PICALM
Phosphatidylinositol binding clathrin assembly protein
 
[4]
0.16
0.00086
Hs.525607
TNFAIP2
Tumor necrosis factor, alpha-induced protein 2, mRNA.
[2729, 32]
 
0.18
0.00043
Hs.654439
APOE
Apolipoprotein E
[24, 29]
 
0.18
0.00086
Hs.201978
PTGS1
Prostaglandin-endoperoxide synthase 1 (prostaglandin G/H synthase and cyclooxygenase), transcript variant 2, mRNA.
[29]
[7]
0.19
0.00013
Hs.82071
CITED2
Cbp/p300-interacting transactivator, with Glu/Asp-rich carboxy-terminal domain, 2, mRNA.
[25]
 
0.19
0.00086
Hs.524518
STAT6
Signal transducer and activator of transcription 6, interleukin-4 induced, mRNA.
[29]
 
0.19
0.00022
Hs.478588
BCL6
B-cell CLL/lymphoma 6 (zinc finger protein 51), transcript variant 1, mRNA.
[25, 27, 29]
 
0.20
0.00086
Hs.1012
C4BPA
Complement component 4 binding protein, alpha
[24, 25, 27, 29, 31, 32]
[39]
0.22
0.00043
Hs.21765
FADS3
Fatty acid desaturase 3, mRNA.
[26]
 
0.23
0.00043
Hs.4055
KLF6
Kruppel-like factor 6
[25]
 
0.23
0.00013
Hs.332708
FBLN5
Fibulin 5, mRNA.
[27, 29]
 
0.23
0.00022
Hs.25292
JUNB
Jun B proto-oncogene, mRNA.
[25, 26]
 
0.25
0.00043
Hs.431048
ABL1
V-abl Abelson murine leukemia viral oncogene homolog 1, transcript variant b, mRNA.
[24]
 
0.27
0.00086
Hs.190783
HAL
Histidine ammonia-lyase
[26, 32]
 
0.27
0.00086
Hs.513984
FLII
Flightless I homolog (Drosophila), mRNA.
[24, 32]
 
0.27
0.00043
Hs.643357
ADAMTS1
ADAM metallopeptidase with thrombospondin type 1 motif, 1
[29]
 
0.29
0.00022
Hs.44227
HPSE
Heparanase
[29]
 
0.29
0.00086
Hs.515536
RRAS
Related RAS viral (r-ras) oncogene homolog, mRNA.
[27]
 
0.29
0.00013
Hs.409578
STK38
Serine/threonine kinase 38
[26]
 
0.29
0.00043
Hs.549171
C1orf56
Chromosome 1 open reading frame 56
 
[7]
0.29
0.00022
Hs.494457
NINJ1
Ninjurin 1, mRNA.
[26]
 
0.29
0.00013
Hs.270291
ACTN4
Actinin, alpha 4 (ACTN4), mRNA.
[29]
 
0.29
0.00086
Hs.381099
LCP1
Lymphocyte cytosolic protein 1 (L-plastin), mRNA.
[28, 29]
 
0.31
0.00043
Hs.185172
GNB2
Guanine nucleotide binding protein (G protein), beta polypeptide 2, mRNA.
[26]
 
0.31
0.00013
Hs.1497
RARG
Retinoic acid receptor, gamma
 
[7]
0.33
0.00043
Hs.474751
MYH9
Myosin, heavy polypeptide 9, non-muscle, mRNA.
[29]
 
0.33
0.00043
Hs.255093
PFKL
Phosphofructokinase, liver, transcript variant 2, mRNA.
[24]
 
0.33
0.00086
Hs.503911
NNMT
Nicotinamide N-methyltransferase
[25, 27]
 
0.33
0.00043
Hs.504877
ARHGDIB
Rho GDP dissociation inhibitor (GDI) beta , mRNA.
[24, 26, 27, 29]
 
0.33
0.00022
Hs.210995
CA12
Carbonic anhydrase XII, transcript variant 2, mRNA.
[27, 32]
 
0.35
0.00043
Hs.520640
ACTB
Actin, beta, mRNA.
 
[7]
0.35
0.00086
Hs.514819
AP2B1
Adaptor-related protein complex 2, beta 1 subunit, mRNA.
[24]
 
0.35
0.00013
Hs.511605
ANXA2
Annexin A2, transcript variant 2, mRNA.
[29]
 
0.35
0.00086
Hs.87752
MSN
Moesin, mRNA.
[29]
 
0.35
0.00022
Hs.654958
ABCF2
ATP-binding cassette, sub-family F (GCN20), member 2
 
[7]
0.35
0.00013
Hs.443577
TNFRSF21
Tumor necrosis factor receptor superfamily, member 21
[29]
[4]
0.35
0.00086
Hs.591868
ZBTB10
Zinc finger and BTB domain containing 10
 
[4]
0.35
0.00043
Hs.25348
VAMP2
Vesicle-associated membrane protein 2 (synaptobrevin 2)
 
[4]
0.38
0.00086
Hs.159161
ARHGDIA
Rho GDP dissociation inhibitor (GDI) alpha, mRNA.
[24]
[7]
0.38
0.00022
Hs.131269
RARRES1
Retinoic acid receptor responder (tazarotene induced) 1
[27]
 
0.38
0.00086
Hs.513915
CLDN7
Claudin 7, mRNA.
[25]
 
0.38
0.00013
Hs.10326
COPE
Coatomer protein complex, subunit epsilon, transcript variant 2, mRNA.
[24]
 
0.38
0.00013
Hs.416024
NRSN2
Neurensin 2
 
[7]
0.38
0.00043
Hs.434248
PLEC
Plectin
[26, 29]
[39]
0.38
0.00086
Hs.584854
AVIL
Advillin
[26, 29]
 
0.41
0.00022
Hs.183109
MAOA
Monoamine oxidase A
[24, 25, 27, 28, 31, 32]
 
0.41
0.00013
Hs.365405
SELO
Selenoprotein O
 
[4]
0.41
0.00013
Hs.645228
KIR3DL1
Killer cell immunoglobulin-like receptor, three domains, long cytoplasmic tail, 1
[29]
 
0.41
0.00086
Hs.528299
HTATIP
HIV-1 Tat interacting protein, 60 kDa, transcript variant 3, mRNA.
[26]
 
0.41
0.00043
Hs.164226
THBS1
Thrombospondin 1, mRNA.
[29]
 
0.41
0.00086
Hs.647078
CDK5
Cyclin-dependent kinase 5
 
[7]
0.41
0.00043
Hs.278573
CD59
CD59 antigen p18-20 (antigen identified by monoclonal antibodies 16.3A5, EJ16, EJ30, EL32 and G344), transcript variant 2, mRNA.
[29]
 
0.41
0.00022
Hs.515162
CALR
Calreticulin
 
[7]
0.41
0.00043
Hs.465744
INSR
Insulin receptor
[26]
 
0.41
0.00013
Hs.274256
ELOVL7
ELOVL family member 7, elongation of long chain fatty acids (yeast)
 
[4]
0.44
0.00086
Hs.450230
IGFBP3
Insulin-like growth factor binding protein 3
[27, 29, 32]
 
0.44
0.00086
Hs.504687
MYL9
Myosin, light polypeptide 9, regulatory
[27]
 
0.44
0.00022
Hs.446641
ARAF
V-raf murine sarcoma 3611 viral oncogene homolog, mRNA.
[25]
 
0.44
0.00086
Hs.2030
THBD
Thrombomodulin
[25, 27, 29]
 
0.44
0.00013
Hs.104672
FILIP1L
Filamin A interacting protein 1-like
[27]
 
0.44
0.00086
Hs.75862
SMAD4
SMAD family member 4
 
[4]
0.44
0.00022
Hs.520757
TBXAS1
Thromboxane A synthase 1 (platelet, cytochrome P450, family 5, subfamily A), transcript variant TXS-II, mRNA.
[29]
 
0.47
0.00013
Hs.283741
EXOSC5
Exosome component 5
 
[7]
0.47
0.00086
Hs.174312
TLR4
Toll-like receptor 4, transcript variant 2, mRNA.
[29]
 
0.47
0.00043
Hs.24601
FBLN1
Fibulin 1
[31]
[7]
0.47
0.00086
Hs.501728
RHOG
Ras homolog gene family, member G (rho G)
 
[7]
0.47
0.00043
Hs.220864
CHD2
Chromodomain helicase DNA binding protein 2
 
[4]
0.47
0.00086
Hs.524809
CLIP1
CAP-GLY domain containing linker protein 1
[29]
 
0.47
0.00043
Hs.92236
MLL4
Myeloid/lymphoid or mixed-lineage leukemia 4
 
[7]
0.47
0.00086
Hs.654688
MKL1
Megakaryoblastic leukemia (translocation) 1
[26]
 
0.47
0.00043
Hs.279837
GSTM2
Glutathione S-transferase mu 2 (muscle)
[26]
 
0.47
0.00086
Hs.645227
TGFB1
Transforming growth factor, beta 1
 
[7]
0.50
0.00086
Hs.149261
RUNX1
Runt-related transcription factor 1
[27]
 
0.50
0.00013
Hs.522818
L1CAM
L1 cell adhesion molecule (hydrocephalus, stenosis of aqueduct of Sylvius 1, MASA (mental retardation, aphasia, shuffling gait and adducted thumbs) syndrome, spastic paraplegia 1)
[26]
 
0.50
0.00043
Hs.840
IDO1
Indoleamine 2,3-dioxygenase 1
[24, 25, 29]
 
0.50
0.00086
Hs.2256
MMP7
Matrix metalloproteinase 7 (matrilysin, uterine)
[27]
 
0.50
0.00043
Data is expressed as fold change for endometrial genes down-regulated ≥2-fold in group A vs. group B that have been shown either up-regulated during the window of implantation or down-regulated in women with endometriosis or treated with mifepristone. Bolded transcripts are decreased also in group A vs. Group C.
Table 6
Genes previously described to be progesterone regulated that are up-regulated in endometrium of subjects with repeated embryo implantation failure
UniGene ID
Gene symbol
Gene title
Down regulated in window of implantation
Up regulated in endometriosis or RU486
Fold change
p value
Hs.208854
CD69
CD69 antigen (p60, early T-cell activation antigen)
 
[7]
2,3
0.00022
Hs.406515
NQO1
NAD(P)H dehydrogenase, quinone 1
[29]
 
2,2
0.00043
Hs.335614
SEC14L2
SEC14-like 2 (S. cerevisiae), mRNA.
 
[4]
1,9
0.00013
Hs.481181
NEK1
NIMA (never in mitosis gene a)-related kinase 1 (NEK1), mRNA.
[29]
 
1,6
0.00043
Hs.86368
CLGN
Calmegin, mRNA.
 
[7]
1,5
0.00086
Hs.189075
TWF1
Twinfilin, actin-binding protein, homolog 1 (Drosophila)
 
[4]
1,5
0.00043
Hs.127680
LOC389332
PREDICTED: hypothetical LOC389332 (LOC389332), mRNA.
 
[4]
1,4
0.00013
Hs.369430
PAM
Peptidylglycine alpha-amidating monooxygenase, transcript variant 3, mRNA.
[24, 28]
 
1,1
0.00022
Hs.514806
GALNT1
UDP-N-acetyl-alpha-D-galactosamine:polypeptide N-acetylgalactosaminyltransferase 1 (GalNAc-T1), mRNA.
 
[7]
1,1
0.00043
Hs.509447
GRLF1
Glucocorticoid receptor DNA binding factor 1
 
[7]
1,1
0.00086
Hs.481927
NIPBL
Nipped-B homolog (Drosophila)
 
[7]
1,1
0.00086
Hs.444558
KHDRBS3
KH domain containing, RNA binding, signal transduction associated 3, mRNA.
[24, 29, 32]
[4]
1,1
0.00043
Hs.495710
GPM6B
Glycoprotein M6B (GPM6B), transcript variant 4, mRNA.
[29]
 
1,0
0.00022
Hs.496414
ATP7A
ATPase, Cu++ transporting, alpha polypeptide (Menkes syndrome)
[29]
 
1,0
0.00043
Data is expressed as fold change for endometrial genes up-regulated ≥2-fold in group A vs. group B that have been shown either down-regulated during the window of implantation or up-regulated in women with endometriosis or treated with mifepristone. Bolded transcripts are increased also in group A vs. Group C.

PROGINS detection

Since the comparative gene expression analysis of P-regulated genes in endometrial samples from group A, suggested an altered P response, we determined the presence of the Alu insertion in intron G of the PR gene (PROGINS) in women from groups A, B and C. Restriction fragment length polymorphism (RFLP) analysis was also carried out on exon 5 of PR gene for confirmation. We found 4 heterozygous subjects for pgr (Figures 2A and B). Two were from group B and two from group C, whereas no PROGINS alleles were detected in women from group A.

IHC analysis

Since the levels of both isoforms of PR in human endometrium have been found to be abnormal in patients with endometriosis [33, 34], we evaluated the immunoreactive presence of PR-A/B (Figures 3A and 3C), PR-B (Figures 3D and 3F) along with Sp1 (Figure 3G and 3I) and the P-regulated glycoprotein glycodelin (Figures 3 J and 3L) in paraformaldehyde-fixed paraffin embedded endometrial tissue from groups A, B and C by IHC. Immunostaining was semi-quantified by calculating the respective ELS scores for each detected molecule in all groups of women (Figure 4). ELS for glycodelin in groups B and C was 10.6 and 12.1 fold from group A respectively (p = 0.00509, Figure 4A). The presence of PR-A/B and PR-B in endometrial tissue was evaluated (Figures 3A-C and 3D-F, respectively), since a possible post-translational dysregulation of PR expression (not detected by transcript analysis) might explain the differential gene expression of P-regulated genes in the endometrium from women of group A such as glycodelin. The ELS scores obtained for PR-A/B and PRB did not show significant differences amongst groups (Figures 4B and 4C respectively). In addition, semi-quantitation of immunoreactive Sp1, a known co-activator and trans-activator of the PR that mediates P-induced glycodelin expression, did not show significant differences amongst groups A, B and C (Figure 4D).

Discussion

Uterine receptivity is defined as a restricted time-related period when the uterus is receptive to blastocyst attachment and implantation. The establishment of this endometrial transition, which supports embryo implantation, is primarily coordinated by ovarian hormones, E2 and P that modulate uterine events in a spatiotemporal manner.
Endometrial factors, at the molecular level, have been suggested to explain some cases of infertility, recurrent miscarriages and implantation failure after IVF. In the present study we assessed the endometrial gene expression profile during the receptive period in mock oocyte donation cycles of women with repeated embryo implantation failure (Group A). Their profiles were compared with those obtained from women who achieved embryo implantation and pregnancy in oocyte donation cycles (Group B), or which got pregnant in natural spontaneous cycles (Group C). The data suggest a strong association between an aberrant endometrial gene expression and implantation failure. The stimulation protocol with steroid hormones performed before the endometrial sample collection was the same for all participating women in this study. Hence, the differential transcript profile in Group A suggests a long-term dysregulation of endometrial gene regulation rendering it not suitable for embryo implantation. The functional annotation analysis of dysregulated transcripts showed an enrichment of decreased genes involved in immune response and complement activation in women with repeated implantation failure.
Integration and cross-validation of endometrial transcripts regulated by P could increase the confidence in expression results for many more genes than is tractable with classical one-by-one validation of differentially expressed genes and should provide the up- and down-regulated genes that together orchestrate the acquisition of the receptive phenotype of the endometrium for embryo implantation. Such exploration and integration could help to get a comprehensive view of existing data needed to better prioritize experimental efforts. We identified a subset of P-regulated transcripts with differential expression in the endometrium of women from group A compared to the control group B revealing compromised P-signalling in the endometrium.
Pisarska et al. [23] reported that 42% of women with unexplained infertility carry the allele for the PROGINS mutation compared with 14% of control fertile women (with at least 1 term pregnancy). We analyzed the presence of the PROGINS allele in women from groups A, B and C and found no correlation between the PROGINS carrier women and altered transcript levels of P-regulated genes in the endometrium. This result is in line with a study from Coulam et al. [35] that did not find an association between PR polymorphisms with recurrent implantation failure in women after in vitro fertilization and embryo transfer.
Glycodelin, encoded by the gene PAEP [36, 37] is the main P-regulated glycoprotein secreted by the endometrial epithelium during the secretory phase and early pregnancy [38]. The transcript levels for PAEP have also been consistently identified to be one of the most abundant in the endometrium by several gene expression profiling studies [24, 25, 27, 31], and it has been shown to be decreased in women with endometriosis [39]. In the present study we found that the transcript levels for PAEP were decreased in the endometrium of women from group A compared to both control groups (Table 5). In addition, immunoreactive glycodelin evaluation in endometrial sections showed the protein to be significantly decreased in group A which is in line with the microarrays data. These results are consistent with the reduced concentrations of glycodelin in uterine flushing reported for patients with unexplained infertility [40]. In normal ovulatory cycles, P secretion is followed by endometrial glycodelin synthesis in epithelial glands from 4 to 5 postovulatory days onwards [37, 41]. Endometrial epithelial cells stimulated in vitro with progestins showed an increase in glycodelin transcription, synthesis, and secretion [42] however a PR-antagonist failed to prevent the induction of glycodelin [43]. In silico analysis of PAEP gene promoter sequence identified a potential P response element [44], however functional studies found that the transcription factor Sp1 mediates the effect of P and PR on human glycodelin expression in endometrial cells [45]. We semiquantified the immunoreactive levels of PR-A/B, PR-B and Sp1 in endometrial sections of women from groups A, B and C and found no significant differences between the groups.
The endometrial response to the increased circulating levels of P during the luteal phase has shown to be remarkably different in women with endometriosis compared to healthy controls. Such response has been evidenced by dysregulated specific gene networks of P-dependent genes in patients with endometriosis compared to non-diseased patients in eutopic secretory endometrium [4, 39, 46, 47]. This transcriptional behavior has led to the concept of ‘P resistance’ which may explain the association between pelvic endometriosis and infertility. We have found that the endometrial transcript profile from women with repeated implantation failure (group A) presents altered gene expression profile including several transcripts reported to be P-regulated, suggesting a women from group A have a compromised P signalling in the endometrium.
The cause of this endometrial defect is unknown, although the apparent intrinsic dysregulation in P signalling that renders the endometrium unreceptive in women with repeated embryo implantation failure seems to be beyond perturbations in PR expression such as chaperone proteins involved in receptor recycling and ligand binding [48], coregulators [4951], as well as associated transcription factors and a variety of upstream signal transduction pathways capable of modifying PR and its coregulators [5256]. In addition, the action of the P is not limited to the cell type in which is PR expressed since steroid hormone regulation can be mediated also through epithelial-stromal cross talk in the endometrium [57]. Also P can elicit a variety of rapid signalling events, independently of a direct transcriptional regulation or even in the absence of its cognate nuclear receptors [58] which may modulate gene expression. The molecular mechanism behind the defect in P-regulated gene networks in the endometrium of women with repeated embryo implantation failure is yet to be determined.

Conclusions

We conclude that some cases of repeated implantation failure could be associated with an aberrant gene expression profile, particularly of transcripts related to the immune function and complement activation. Compromised P signaling might be the underlying mechanism for such endometrial gene expression deregulation in women with repeated implantation failure. Future research should focus on determining the causes of incomplete P signalling in the endometrium from these women.

Acknowledgements

We acknowledge to all volunteer women participating in the present study for their generous contribution. Also we acknowledge Dr. Fernando Gabler for dating the endometrial samples, Dr. H. Koistinen for providing the glycodelin antibody and Felipe Argandona for excellent technical assistance. This work was supported by FONDECYT 11100443 / 1140614 and FONDAP 15010006.
This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://​creativecommons.​org/​licenses/​by/​4.​0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly credited. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

AT conceived the study, participated in its design, carried out the microarrays and bioinformatic analyses and helped to draft the manuscript. PF and JB participated in PROGINS detection. J.C.M participated in PROGINS detection, data analysis and interpretation. D.J.M. participated in microarrays analysis. HBC conceived the study, participated in its design, data interpretation and helped to draft the manuscript. All authors read and approved the final manuscript.
Anhänge

Authors’ original submitted files for images

Literatur
1.
Zurück zum Zitat O’Malley BW, Tsai MJ: Molecular pathways of steroid receptor action. Biol Reprod. 1992, 46: 163-167. 10.1095/biolreprod46.2.163.CrossRefPubMed O’Malley BW, Tsai MJ: Molecular pathways of steroid receptor action. Biol Reprod. 1992, 46: 163-167. 10.1095/biolreprod46.2.163.CrossRefPubMed
2.
Zurück zum Zitat Gellersen B, Brosens IA, Brosens JJ: Decidualization of the human endometrium: mechanisms, functions, and clinical perspectives. Semin Reprod Med. 2007, 25: 445-453. 10.1055/s-2007-991042.CrossRefPubMed Gellersen B, Brosens IA, Brosens JJ: Decidualization of the human endometrium: mechanisms, functions, and clinical perspectives. Semin Reprod Med. 2007, 25: 445-453. 10.1055/s-2007-991042.CrossRefPubMed
3.
Zurück zum Zitat Beato M: Gene regulation by steroid hormones. Cell. 1989, 56: 335-344. 10.1016/0092-8674(89)90237-7.CrossRefPubMed Beato M: Gene regulation by steroid hormones. Cell. 1989, 56: 335-344. 10.1016/0092-8674(89)90237-7.CrossRefPubMed
4.
Zurück zum Zitat Burney RO, Talbi S, Hamilton AE, Vo KC, Nyegaard M, Nezhat CR, Lessey BA, Giudice LC: Gene expression analysis of endometrium reveals progesterone resistance and candidate susceptibility genes in women with endometriosis. Endocrinology. 2007, 148: 3814-3826. 10.1210/en.2006-1692.CrossRefPubMed Burney RO, Talbi S, Hamilton AE, Vo KC, Nyegaard M, Nezhat CR, Lessey BA, Giudice LC: Gene expression analysis of endometrium reveals progesterone resistance and candidate susceptibility genes in women with endometriosis. Endocrinology. 2007, 148: 3814-3826. 10.1210/en.2006-1692.CrossRefPubMed
5.
Zurück zum Zitat Hapangama DK, Critchley HO, Henderson TA, Baird DT: Mifepristone-induced vaginal bleeding is associated with increased immunostaining for cyclooxygenase-2 and decrease in prostaglandin dehydrogenase in luteal phase endometrium. J Clin Endocrinol Metab. 2002, 87: 5229-5234. 10.1210/jc.2002-020429.CrossRefPubMed Hapangama DK, Critchley HO, Henderson TA, Baird DT: Mifepristone-induced vaginal bleeding is associated with increased immunostaining for cyclooxygenase-2 and decrease in prostaglandin dehydrogenase in luteal phase endometrium. J Clin Endocrinol Metab. 2002, 87: 5229-5234. 10.1210/jc.2002-020429.CrossRefPubMed
6.
Zurück zum Zitat Danielsson KG, Marions L, Bygdeman M: Effects of mifepristone on endometrial receptivity. Steroids. 2003, 68: 1069-1075. 10.1016/S0039-128X(03)00131-4.CrossRefPubMed Danielsson KG, Marions L, Bygdeman M: Effects of mifepristone on endometrial receptivity. Steroids. 2003, 68: 1069-1075. 10.1016/S0039-128X(03)00131-4.CrossRefPubMed
7.
Zurück zum Zitat Catalano RD, Critchley HO, Heikinheimo O, Baird DT, Hapangama D, Sherwin JR, Charnock-Jones DS, Smith SK, Sharkey AM: Mifepristone induced progesterone withdrawal reveals novel regulatory pathways in human endometrium. Mol Hum Reprod. 2007, 13: 641-654. 10.1093/molehr/gam021.CrossRefPubMed Catalano RD, Critchley HO, Heikinheimo O, Baird DT, Hapangama D, Sherwin JR, Charnock-Jones DS, Smith SK, Sharkey AM: Mifepristone induced progesterone withdrawal reveals novel regulatory pathways in human endometrium. Mol Hum Reprod. 2007, 13: 641-654. 10.1093/molehr/gam021.CrossRefPubMed
8.
Zurück zum Zitat Horcajadas JA, Pellicer A, Simon C: Wide genomic analysis of human endometrial receptivity: new times, new opportunities. Hum Reprod Update. 2007, 13: 77-86.CrossRefPubMed Horcajadas JA, Pellicer A, Simon C: Wide genomic analysis of human endometrial receptivity: new times, new opportunities. Hum Reprod Update. 2007, 13: 77-86.CrossRefPubMed
9.
Zurück zum Zitat Ruiz-Alonso M, Blesa D, Simon C: The genomics of the human endometrium. Biochim Biophys Acta. 1822, 2012: 1931-1942. Ruiz-Alonso M, Blesa D, Simon C: The genomics of the human endometrium. Biochim Biophys Acta. 1822, 2012: 1931-1942.
10.
Zurück zum Zitat Haouzi D, Dechaud H, Assou S, De Vos J, Hamamah S: Insights into human endometrial receptivity from transcriptomic and proteomic data. Reprod Biomed Online. 2012, 24: 23-34. 10.1016/j.rbmo.2011.09.009.CrossRefPubMed Haouzi D, Dechaud H, Assou S, De Vos J, Hamamah S: Insights into human endometrial receptivity from transcriptomic and proteomic data. Reprod Biomed Online. 2012, 24: 23-34. 10.1016/j.rbmo.2011.09.009.CrossRefPubMed
11.
Zurück zum Zitat Tapia A, Vilos C, Marin JC, Croxatto HB, Devoto L: Bioinformatic detection of E47, E2F1 and SREBP1 transcription factors as potential regulators of genes associated to acquisition of endometrial receptivity. Reprod Biol Endocrinol. 2011, 9: 14-10.1186/1477-7827-9-14.PubMedCentralCrossRefPubMed Tapia A, Vilos C, Marin JC, Croxatto HB, Devoto L: Bioinformatic detection of E47, E2F1 and SREBP1 transcription factors as potential regulators of genes associated to acquisition of endometrial receptivity. Reprod Biol Endocrinol. 2011, 9: 14-10.1186/1477-7827-9-14.PubMedCentralCrossRefPubMed
12.
Zurück zum Zitat Tapia A, Gangi LM, Zegers-Hochschild F, Balmaceda J, Pommer R, Trejo L, Pacheco IM, Salvatierra AM, Henriquez S, Quezada M, Vargas M, Ríos M, Munroe DJ, Croxatto HB, Velasquez L: Differences in the endometrial transcript profile during the receptive period between women who were refractory to implantation and those who achieved pregnancy. Hum Reprod. 2008, 23: 340-351.CrossRefPubMed Tapia A, Gangi LM, Zegers-Hochschild F, Balmaceda J, Pommer R, Trejo L, Pacheco IM, Salvatierra AM, Henriquez S, Quezada M, Vargas M, Ríos M, Munroe DJ, Croxatto HB, Velasquez L: Differences in the endometrial transcript profile during the receptive period between women who were refractory to implantation and those who achieved pregnancy. Hum Reprod. 2008, 23: 340-351.CrossRefPubMed
13.
Zurück zum Zitat Damario MA, Lesnick TG, Lessey BA, Kowalik A, Mandelin E, Seppala M, Rosenwaks Z: Endometrial markers of uterine receptivity utilizing the donor oocyte model. Hum Reprod. 2001, 16: 1893-1899. 10.1093/humrep/16.9.1893.CrossRefPubMed Damario MA, Lesnick TG, Lessey BA, Kowalik A, Mandelin E, Seppala M, Rosenwaks Z: Endometrial markers of uterine receptivity utilizing the donor oocyte model. Hum Reprod. 2001, 16: 1893-1899. 10.1093/humrep/16.9.1893.CrossRefPubMed
14.
Zurück zum Zitat Noyes RW, Hertig AT, Rock J: Dating the endometrial biopsy. Am J Obstet Gynecol. 1975, 122: 262-263.PubMed Noyes RW, Hertig AT, Rock J: Dating the endometrial biopsy. Am J Obstet Gynecol. 1975, 122: 262-263.PubMed
15.
Zurück zum Zitat Benjamini Y, Hochberg Y: Controlling the False Discovery Rate: A Practical and Powerful Approach to Multiple Testing. J Roy Statist Soc Ser B (Method). 1995, 57: 289-300. Benjamini Y, Hochberg Y: Controlling the False Discovery Rate: A Practical and Powerful Approach to Multiple Testing. J Roy Statist Soc Ser B (Method). 1995, 57: 289-300.
16.
Zurück zum Zitat Joliffe IT, Morgan BJ: Principal component analysis and exploratory factor analysis. Stat Methods Med Res. 1992, 1: 69-95. 10.1177/096228029200100105.CrossRefPubMed Joliffe IT, Morgan BJ: Principal component analysis and exploratory factor analysis. Stat Methods Med Res. 1992, 1: 69-95. 10.1177/096228029200100105.CrossRefPubMed
17.
Zurück zum Zitat Dennis G, Sherman BT, Hosack DA, Yang J, Gao W, Lane HC, Lempicki RA: DAVID: Database for Annotation, Visualization, and Integrated Discovery. Genome Biol. 2003, 4: P3-10.1186/gb-2003-4-5-p3.CrossRefPubMed Dennis G, Sherman BT, Hosack DA, Yang J, Gao W, Lane HC, Lempicki RA: DAVID: Database for Annotation, Visualization, and Integrated Discovery. Genome Biol. 2003, 4: P3-10.1186/gb-2003-4-5-p3.CrossRefPubMed
18.
Zurück zum Zitat Chang JT, Nevins JR: GATHER: a systems approach to interpreting genomic signatures. Bioinformatics. 2006, 22: 2926-2933. 10.1093/bioinformatics/btl483.CrossRefPubMed Chang JT, Nevins JR: GATHER: a systems approach to interpreting genomic signatures. Bioinformatics. 2006, 22: 2926-2933. 10.1093/bioinformatics/btl483.CrossRefPubMed
19.
Zurück zum Zitat Kanehisa M, Goto S, Hattori M, Aoki-Kinoshita KF, Itoh M, Kawashima S, Katayama T, Araki M, Hirakawa M: From genomics to chemical genomics: new developments in KEGG. Nucleic Acids Res. 2006, 34: D354-357. 10.1093/nar/gkj102.PubMedCentralCrossRefPubMed Kanehisa M, Goto S, Hattori M, Aoki-Kinoshita KF, Itoh M, Kawashima S, Katayama T, Araki M, Hirakawa M: From genomics to chemical genomics: new developments in KEGG. Nucleic Acids Res. 2006, 34: D354-357. 10.1093/nar/gkj102.PubMedCentralCrossRefPubMed
20.
Zurück zum Zitat Ashburner M, Ball CA, Blake JA, Botstein D, Butler H, Cherry JM, Davis AP, Dolinski K, Dwight SS, Eppig JT, Harris MA, Hill DP, Issel-Tarver L, Kasarskis A, Lewis S, Matese JC, Richardson JE, Ringwald M, Rubin GM, Sherlock G: Gene ontology: tool for the unification of biology. Gene Ontology Consortium Nat Genet. 2000, 25: 25-29.PubMed Ashburner M, Ball CA, Blake JA, Botstein D, Butler H, Cherry JM, Davis AP, Dolinski K, Dwight SS, Eppig JT, Harris MA, Hill DP, Issel-Tarver L, Kasarskis A, Lewis S, Matese JC, Richardson JE, Ringwald M, Rubin GM, Sherlock G: Gene ontology: tool for the unification of biology. Gene Ontology Consortium Nat Genet. 2000, 25: 25-29.PubMed
21.
Zurück zum Zitat Gonzalez-Ramos R, Rocco J, Rojas C, Sovino H, Poch A, Kohen P, Alvarado-Diaz C, Devoto L: Physiologic activation of nuclear factor kappa-B in the endometrium during the menstrual cycle is altered in endometriosis patients. Fertil Steril. 2012, 97: 645-651. 10.1016/j.fertnstert.2011.12.006.CrossRefPubMed Gonzalez-Ramos R, Rocco J, Rojas C, Sovino H, Poch A, Kohen P, Alvarado-Diaz C, Devoto L: Physiologic activation of nuclear factor kappa-B in the endometrium during the menstrual cycle is altered in endometriosis patients. Fertil Steril. 2012, 97: 645-651. 10.1016/j.fertnstert.2011.12.006.CrossRefPubMed
22.
Zurück zum Zitat Koistinen H, Koistinen R, Dell A, Morris HR, Easton RL, Patankar MS, Oehninger S, Clark GF, Seppala M: Glycodelin from seminal plasma is a differentially glycosylated form of contraceptive glycodelin-A. Mol Hum Reprod. 1996, 2: 759-765. 10.1093/molehr/2.10.759.CrossRefPubMed Koistinen H, Koistinen R, Dell A, Morris HR, Easton RL, Patankar MS, Oehninger S, Clark GF, Seppala M: Glycodelin from seminal plasma is a differentially glycosylated form of contraceptive glycodelin-A. Mol Hum Reprod. 1996, 2: 759-765. 10.1093/molehr/2.10.759.CrossRefPubMed
23.
Zurück zum Zitat Pisarska MD, Carson SA, Casson PR, Tong X, Buster JE, Kieback DG: A mutated progesterone receptor allele is more prevalent in unexplained infertility. Fertil Steril. 2003, 80: 651-653. 10.1016/S0015-0282(03)00755-6.CrossRefPubMed Pisarska MD, Carson SA, Casson PR, Tong X, Buster JE, Kieback DG: A mutated progesterone receptor allele is more prevalent in unexplained infertility. Fertil Steril. 2003, 80: 651-653. 10.1016/S0015-0282(03)00755-6.CrossRefPubMed
24.
Zurück zum Zitat 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. 10.1210/endo.143.6.8885.CrossRefPubMed 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. 10.1210/endo.143.6.8885.CrossRefPubMed
25.
Zurück zum Zitat Borthwick JM, Charnock-Jones DS, Tom BD, Hull ML, Teirney R, Phillips SC, Smith SK: Determination of the transcript profile of human endometrium. Mol Hum Reprod. 2003, 9: 19-33. 10.1093/molehr/gag004.CrossRefPubMed Borthwick JM, Charnock-Jones DS, Tom BD, Hull ML, Teirney R, Phillips SC, Smith SK: Determination of the transcript profile of human endometrium. Mol Hum Reprod. 2003, 9: 19-33. 10.1093/molehr/gag004.CrossRefPubMed
26.
Zurück zum Zitat Carson DD, Lagow E, Thathiah A, Al-Shami R, Farach-Carson MC, Vernon M, Yuan L, Fritz MA, Lessey B: Changes in gene expression during the early to mid-luteal (receptive phase) transition in human endometrium detected by high-density microarray screening. Mol Hum Reprod. 2002, 8: 871-879. 10.1093/molehr/8.9.871.CrossRefPubMed Carson DD, Lagow E, Thathiah A, Al-Shami R, Farach-Carson MC, Vernon M, Yuan L, Fritz MA, Lessey B: Changes in gene expression during the early to mid-luteal (receptive phase) transition in human endometrium detected by high-density microarray screening. Mol Hum Reprod. 2002, 8: 871-879. 10.1093/molehr/8.9.871.CrossRefPubMed
27.
Zurück zum Zitat Riesewijk A, Martin J, van Os R, Horcajadas JA, Polman J, Pellicer A, Mosselman S, Simon C: Gene expression profiling of human endometrial receptivity on days LH + 2 versus LH + 7 by microarray technology. Mol Hum Reprod. 2003, 9: 253-264. 10.1093/molehr/gag037.CrossRefPubMed Riesewijk A, Martin J, van Os R, Horcajadas JA, Polman J, Pellicer A, Mosselman S, Simon C: Gene expression profiling of human endometrial receptivity on days LH + 2 versus LH + 7 by microarray technology. Mol Hum Reprod. 2003, 9: 253-264. 10.1093/molehr/gag037.CrossRefPubMed
28.
Zurück zum Zitat Mirkin S, Arslan M, Churikov D, Corica A, Diaz JI, Williams S, Bocca S, Oehninger S: In search of candidate genes critically expressed in the human endometrium during the window of implantation. Hum Reprod. 2005, 20: 2104-2117. 10.1093/humrep/dei051.CrossRefPubMed Mirkin S, Arslan M, Churikov D, Corica A, Diaz JI, Williams S, Bocca S, Oehninger S: In search of candidate genes critically expressed in the human endometrium during the window of implantation. Hum Reprod. 2005, 20: 2104-2117. 10.1093/humrep/dei051.CrossRefPubMed
29.
Zurück zum Zitat Talbi S, Hamilton AE, Vo KC, Tulac S, Overgaard MT, Dosiou C, Le Shay N, Nezhat CN, Kempson R, Lessey BA, Nayak NR, Giudice LC: Molecular phenotyping of human endometrium distinguishes menstrual cycle phases and underlying biological processes in normo-ovulatory women. Endocrinology. 2006, 147: 1097-1121. 10.1210/en.2005-1076.CrossRefPubMed Talbi S, Hamilton AE, Vo KC, Tulac S, Overgaard MT, Dosiou C, Le Shay N, Nezhat CN, Kempson R, Lessey BA, Nayak NR, Giudice LC: Molecular phenotyping of human endometrium distinguishes menstrual cycle phases and underlying biological processes in normo-ovulatory women. Endocrinology. 2006, 147: 1097-1121. 10.1210/en.2005-1076.CrossRefPubMed
30.
Zurück zum Zitat Ace CI, Okulicz WC: Microarray profiling of progesterone-regulated endometrial genes during the rhesus monkey secretory phase. Reprod Biol Endocrinol. 2004, 2: 54-10.1186/1477-7827-2-54.PubMedCentralCrossRefPubMed Ace CI, Okulicz WC: Microarray profiling of progesterone-regulated endometrial genes during the rhesus monkey secretory phase. Reprod Biol Endocrinol. 2004, 2: 54-10.1186/1477-7827-2-54.PubMedCentralCrossRefPubMed
31.
Zurück zum Zitat 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. 2009, 24: 198-205.CrossRefPubMed 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. 2009, 24: 198-205.CrossRefPubMed
32.
Zurück zum Zitat Diaz-Gimeno P, Horcajadas JA, Martinez-Conejero JA, Esteban FJ, Alama P, Pellicer A, Simon C: A genomic diagnostic tool for human endometrial receptivity based on the transcriptomic signature. Fertil Steril. 2011, 95: 50-60. 10.1016/j.fertnstert.2010.04.063. 60 e51-15CrossRefPubMed Diaz-Gimeno P, Horcajadas JA, Martinez-Conejero JA, Esteban FJ, Alama P, Pellicer A, Simon C: A genomic diagnostic tool for human endometrial receptivity based on the transcriptomic signature. Fertil Steril. 2011, 95: 50-60. 10.1016/j.fertnstert.2010.04.063. 60 e51-15CrossRefPubMed
33.
Zurück zum Zitat Bulun SE, Cheng YH, Yin P, Imir G, Utsunomiya H, Attar E, Innes J, Julie Kim J: Progesterone resistance in endometriosis: link to failure to metabolize estradiol. Mol Cell Endocrinol. 2006, 248: 94-103. 10.1016/j.mce.2005.11.041.CrossRefPubMed Bulun SE, Cheng YH, Yin P, Imir G, Utsunomiya H, Attar E, Innes J, Julie Kim J: Progesterone resistance in endometriosis: link to failure to metabolize estradiol. Mol Cell Endocrinol. 2006, 248: 94-103. 10.1016/j.mce.2005.11.041.CrossRefPubMed
34.
Zurück zum Zitat Attia GR, Zeitoun K, Edwards D, Johns A, Carr BR, Bulun SE: Progesterone receptor isoform A but not B is expressed in endometriosis. J Clin Endocrinol Metab. 2000, 85: 2897-2902.PubMed Attia GR, Zeitoun K, Edwards D, Johns A, Carr BR, Bulun SE: Progesterone receptor isoform A but not B is expressed in endometriosis. J Clin Endocrinol Metab. 2000, 85: 2897-2902.PubMed
35.
Zurück zum Zitat Coulam CB, Jeyendran RS, Roussev R: Association of progesterone receptor polymorphisms with recurrent implantation failure after in vitro fertilization and embryo transfer. J Assist Reprod Genet. 2008, 25: 119-122. 10.1007/s10815-008-9210-9.PubMedCentralCrossRefPubMed Coulam CB, Jeyendran RS, Roussev R: Association of progesterone receptor polymorphisms with recurrent implantation failure after in vitro fertilization and embryo transfer. J Assist Reprod Genet. 2008, 25: 119-122. 10.1007/s10815-008-9210-9.PubMedCentralCrossRefPubMed
36.
Zurück zum Zitat Seppala M, Bohn H, Tatarinov Y: Glycodelins. Tumour Biol. 1998, 19: 213-220. 10.1159/000030009.CrossRefPubMed Seppala M, Bohn H, Tatarinov Y: Glycodelins. Tumour Biol. 1998, 19: 213-220. 10.1159/000030009.CrossRefPubMed
37.
Zurück zum Zitat Seppala M, Taylor RN, Koistinen H, Koistinen R, Milgrom E: Glycodelin: a major lipocalin protein of the reproductive axis with diverse actions in cell recognition and differentiation. Endocr Rev. 2002, 23: 401-430. 10.1210/er.2001-0026.CrossRefPubMed Seppala M, Taylor RN, Koistinen H, Koistinen R, Milgrom E: Glycodelin: a major lipocalin protein of the reproductive axis with diverse actions in cell recognition and differentiation. Endocr Rev. 2002, 23: 401-430. 10.1210/er.2001-0026.CrossRefPubMed
38.
Zurück zum Zitat Bell SC, Hales MW, Patel S, Kirwan PH, Drife JO: Protein synthesis and secretion by the human endometrium and decidua during early pregnancy. Br J Obstet Gynaecol. 1985, 92: 793-803. 10.1111/j.1471-0528.1985.tb03048.x.CrossRefPubMed Bell SC, Hales MW, Patel S, Kirwan PH, Drife JO: Protein synthesis and secretion by the human endometrium and decidua during early pregnancy. Br J Obstet Gynaecol. 1985, 92: 793-803. 10.1111/j.1471-0528.1985.tb03048.x.CrossRefPubMed
39.
Zurück zum Zitat Kao LC, Germeyer A, Tulac S, Lobo S, Yang JP, Taylor RN, Osteen K, Lessey BA, Giudice LC: Expression profiling of endometrium from women with endometriosis reveals candidate genes for disease-based implantation failure and infertility. Endocrinology. 2003, 144: 2870-2881. 10.1210/en.2003-0043.CrossRefPubMed Kao LC, Germeyer A, Tulac S, Lobo S, Yang JP, Taylor RN, Osteen K, Lessey BA, Giudice LC: Expression profiling of endometrium from women with endometriosis reveals candidate genes for disease-based implantation failure and infertility. Endocrinology. 2003, 144: 2870-2881. 10.1210/en.2003-0043.CrossRefPubMed
40.
Zurück zum Zitat Mackenna A, Li TC, Dalton C, Bolton A, Cooke I: Placental protein 14 levels in uterine flushing and plasma of women with unexplained infertility. Fertil Steril. 1993, 59: 577-582.PubMed Mackenna A, Li TC, Dalton C, Bolton A, Cooke I: Placental protein 14 levels in uterine flushing and plasma of women with unexplained infertility. Fertil Steril. 1993, 59: 577-582.PubMed
41.
Zurück zum Zitat Seppala M, Koistinen H, Koistinen R, Chiu PC, Yeung WS: Glycosylation related actions of glycodelin: gamete, cumulus cell, immune cell and clinical associations. Hum Reprod Update. 2007, 13: 275-287. 10.1093/humupd/dmm004.CrossRefPubMed Seppala M, Koistinen H, Koistinen R, Chiu PC, Yeung WS: Glycosylation related actions of glycodelin: gamete, cumulus cell, immune cell and clinical associations. Hum Reprod Update. 2007, 13: 275-287. 10.1093/humupd/dmm004.CrossRefPubMed
42.
Zurück zum Zitat Taylor RN, Vigne JL, Zhang P, Hoang P, Lebovic DI, Mueller MD: Effects of progestins and relaxin on glycodelin gene expression in human endometrial cells. Am J Obstet Gynecol. 2000, 182: 841-847. 10.1016/S0002-9378(00)70333-4. discussion 847–849CrossRefPubMed Taylor RN, Vigne JL, Zhang P, Hoang P, Lebovic DI, Mueller MD: Effects of progestins and relaxin on glycodelin gene expression in human endometrial cells. Am J Obstet Gynecol. 2000, 182: 841-847. 10.1016/S0002-9378(00)70333-4. discussion 847–849CrossRefPubMed
43.
Zurück zum Zitat Taylor RN, Savouret JF, Vaisse C, Vigne JL, Ryan I, Hornung D, Seppala M, Milgrom E: Promegestone (R5020) and mifepristone (RU486) both function as progestational agonists of human glycodelin gene expression in isolated human epithelial cells. J Clin Endocrinol Metab. 1998, 83: 4006-4012.PubMed Taylor RN, Savouret JF, Vaisse C, Vigne JL, Ryan I, Hornung D, Seppala M, Milgrom E: Promegestone (R5020) and mifepristone (RU486) both function as progestational agonists of human glycodelin gene expression in isolated human epithelial cells. J Clin Endocrinol Metab. 1998, 83: 4006-4012.PubMed
44.
Zurück zum Zitat Vaisse C, Atger M, Potier B, Milgrom E: Human placental protein 14 gene: sequence and characterization of a short duplication. DNA Cell Biol. 1990, 9: 401-413. 10.1089/dna.1990.9.401.CrossRefPubMed Vaisse C, Atger M, Potier B, Milgrom E: Human placental protein 14 gene: sequence and characterization of a short duplication. DNA Cell Biol. 1990, 9: 401-413. 10.1089/dna.1990.9.401.CrossRefPubMed
45.
Zurück zum Zitat Gao J, Mazella J, Seppala M, Tseng L: Ligand activated hPR modulates the glycodelin promoter activity through the Sp1 sites in human endometrial adenocarcinoma cells. Mol Cell Endocrinol. 2001, 176: 97-102. 10.1016/S0303-7207(01)00450-6.CrossRefPubMed Gao J, Mazella J, Seppala M, Tseng L: Ligand activated hPR modulates the glycodelin promoter activity through the Sp1 sites in human endometrial adenocarcinoma cells. Mol Cell Endocrinol. 2001, 176: 97-102. 10.1016/S0303-7207(01)00450-6.CrossRefPubMed
46.
Zurück zum Zitat Aghajanova L, Velarde MC, Giudice LC: Altered gene expression profiling in endometrium: evidence for progesterone resistance. Semin Reprod Med. 2010, 28: 51-58. 10.1055/s-0029-1242994.CrossRefPubMed Aghajanova L, Velarde MC, Giudice LC: Altered gene expression profiling in endometrium: evidence for progesterone resistance. Semin Reprod Med. 2010, 28: 51-58. 10.1055/s-0029-1242994.CrossRefPubMed
47.
Zurück zum Zitat Taylor HS, Bagot C, Kardana A, Olive D, Arici A: HOX gene expression is altered in the endometrium of women with endometriosis. Hum Reprod. 1999, 14: 1328-1331. 10.1093/humrep/14.5.1328.CrossRefPubMed Taylor HS, Bagot C, Kardana A, Olive D, Arici A: HOX gene expression is altered in the endometrium of women with endometriosis. Hum Reprod. 1999, 14: 1328-1331. 10.1093/humrep/14.5.1328.CrossRefPubMed
48.
Zurück zum Zitat Hirota Y, Tranguch S, Daikoku T, Hasegawa A, Osuga Y, Taketani Y, Dey SK: Deficiency of immunophilin FKBP52 promotes endometriosis. Am J Pathol. 2008, 173: 1747-1757. 10.2353/ajpath.2008.080527.PubMedCentralCrossRefPubMed Hirota Y, Tranguch S, Daikoku T, Hasegawa A, Osuga Y, Taketani Y, Dey SK: Deficiency of immunophilin FKBP52 promotes endometriosis. Am J Pathol. 2008, 173: 1747-1757. 10.2353/ajpath.2008.080527.PubMedCentralCrossRefPubMed
49.
Zurück zum Zitat Aghajanova L, Velarde MC, Giudice LC: The progesterone receptor coactivator Hic-5 is involved in the pathophysiology of endometriosis. Endocrinology. 2009, 150: 3863-3870. 10.1210/en.2009-0008.PubMedCentralCrossRefPubMed Aghajanova L, Velarde MC, Giudice LC: The progesterone receptor coactivator Hic-5 is involved in the pathophysiology of endometriosis. Endocrinology. 2009, 150: 3863-3870. 10.1210/en.2009-0008.PubMedCentralCrossRefPubMed
50.
Zurück zum Zitat Caballero V, Ruiz R, Sainz JA, Cruz M, Lopez-Nevot MA, Galan JJ, Real LM, de Castro F, Lopez-Villaverde V, Ruiz A: Preliminary molecular genetic analysis of the Receptor Interacting Protein 140 (RIP140) in women affected by endometriosis. J Exp Clin Assist Reprod. 2005, 2: 11-10.1186/1743-1050-2-11.PubMedCentralCrossRefPubMed Caballero V, Ruiz R, Sainz JA, Cruz M, Lopez-Nevot MA, Galan JJ, Real LM, de Castro F, Lopez-Villaverde V, Ruiz A: Preliminary molecular genetic analysis of the Receptor Interacting Protein 140 (RIP140) in women affected by endometriosis. J Exp Clin Assist Reprod. 2005, 2: 11-10.1186/1743-1050-2-11.PubMedCentralCrossRefPubMed
51.
Zurück zum Zitat Suzuki A, Horiuchi A, Oka K, Miyamoto T, Kashima H, Shiozawa T: Immunohistochemical detection of steroid receptor cofactors in ovarian endometriosis: involvement of down-regulated SRC-1 expression in the limited growth activity of the endometriotic epithelium. Virchows Arch. 2010, 456: 433-441. 10.1007/s00428-010-0884-x.CrossRefPubMed Suzuki A, Horiuchi A, Oka K, Miyamoto T, Kashima H, Shiozawa T: Immunohistochemical detection of steroid receptor cofactors in ovarian endometriosis: involvement of down-regulated SRC-1 expression in the limited growth activity of the endometriotic epithelium. Virchows Arch. 2010, 456: 433-441. 10.1007/s00428-010-0884-x.CrossRefPubMed
52.
Zurück zum Zitat Kim JJ, Taylor HS, Lu Z, Ladhani O, Hastings JM, Jackson KS, Wu Y, Guo SW, Fazleabas AT: Altered expression of HOXA10 in endometriosis: potential role in decidualization. Mol Hum Reprod. 2007, 13: 323-332. 10.1093/molehr/gam005.CrossRefPubMed Kim JJ, Taylor HS, Lu Z, Ladhani O, Hastings JM, Jackson KS, Wu Y, Guo SW, Fazleabas AT: Altered expression of HOXA10 in endometriosis: potential role in decidualization. Mol Hum Reprod. 2007, 13: 323-332. 10.1093/molehr/gam005.CrossRefPubMed
53.
Zurück zum Zitat Lee B, Du H, Taylor HS: Experimental murine endometriosis induces DNA methylation and altered gene expression in eutopic endometrium. Biol Reprod. 2009, 80: 79-85. 10.1095/biolreprod.108.070391.PubMedCentralCrossRefPubMed Lee B, Du H, Taylor HS: Experimental murine endometriosis induces DNA methylation and altered gene expression in eutopic endometrium. Biol Reprod. 2009, 80: 79-85. 10.1095/biolreprod.108.070391.PubMedCentralCrossRefPubMed
54.
Zurück zum Zitat Shazand K, Baban S, Prive C, Malette B, Croteau P, Lagace M, Racine JB, Hugo P: FOXO1 and c-jun transcription factors mRNA are modulated in endometriosis. Mol Hum Reprod. 2004, 10: 871-877. 10.1093/molehr/gah119.CrossRefPubMed Shazand K, Baban S, Prive C, Malette B, Croteau P, Lagace M, Racine JB, Hugo P: FOXO1 and c-jun transcription factors mRNA are modulated in endometriosis. Mol Hum Reprod. 2004, 10: 871-877. 10.1093/molehr/gah119.CrossRefPubMed
55.
Zurück zum Zitat Wu Y, Halverson G, Basir Z, Strawn E, Yan P, Guo SW: Aberrant methylation at HOXA10 may be responsible for its aberrant expression in the endometrium of patients with endometriosis. Am J Obstet Gynecol. 2005, 193: 371-380. 10.1016/j.ajog.2005.01.034.CrossRefPubMed Wu Y, Halverson G, Basir Z, Strawn E, Yan P, Guo SW: Aberrant methylation at HOXA10 may be responsible for its aberrant expression in the endometrium of patients with endometriosis. Am J Obstet Gynecol. 2005, 193: 371-380. 10.1016/j.ajog.2005.01.034.CrossRefPubMed
56.
Zurück zum Zitat Yang S, Fang Z, Suzuki T, Sasano H, Zhou J, Gurates B, Tamura M, Ferrer K, Bulun S: Regulation of aromatase P450 expression in endometriotic and endometrial stromal cells by CCAAT/enhancer binding proteins (C/EBPs): decreased C/EBPbeta in endometriosis is associated with overexpression of aromatase. J Clin Endocrinol Metab. 2002, 87: 2336-2345.PubMed Yang S, Fang Z, Suzuki T, Sasano H, Zhou J, Gurates B, Tamura M, Ferrer K, Bulun S: Regulation of aromatase P450 expression in endometriotic and endometrial stromal cells by CCAAT/enhancer binding proteins (C/EBPs): decreased C/EBPbeta in endometriosis is associated with overexpression of aromatase. J Clin Endocrinol Metab. 2002, 87: 2336-2345.PubMed
57.
Zurück zum Zitat Cunha GR, Cooke PS, Kurita T: Role of stromal-epithelial interactions in hormonal responses. Arch Histol Cytol. 2004, 67: 417-434. 10.1679/aohc.67.417.CrossRefPubMed Cunha GR, Cooke PS, Kurita T: Role of stromal-epithelial interactions in hormonal responses. Arch Histol Cytol. 2004, 67: 417-434. 10.1679/aohc.67.417.CrossRefPubMed
58.
Zurück zum Zitat Gellersen B, Fernandes MS, Brosens JJ: Non-genomic progesterone actions in female reproduction. Hum Reprod Update. 2009, 15: 119-138.CrossRefPubMed Gellersen B, Fernandes MS, Brosens JJ: Non-genomic progesterone actions in female reproduction. Hum Reprod Update. 2009, 15: 119-138.CrossRefPubMed
Metadaten
Titel
Endometrial gene expression reveals compromised progesterone signaling in women refractory to embryo implantation
verfasst von
Alejandro Tapia-Pizarro
Paula Figueroa
Julio Brito
Juan Carlos Marín
David J Munroe
Horacio B Croxatto
Publikationsdatum
01.12.2014
Verlag
BioMed Central
Erschienen in
Reproductive Biology and Endocrinology / Ausgabe 1/2014
Elektronische ISSN: 1477-7827
DOI
https://doi.org/10.1186/1477-7827-12-92

Weitere Artikel der Ausgabe 1/2014

Reproductive Biology and Endocrinology 1/2014 Zur Ausgabe

Update Gynäkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert – ganz bequem per eMail.