Open Access
How to translate text using browser tools
14 April 2010 Differential Endometrial Gene Expression in Pregnant and Nonpregnant Sows
Esben Østrup, Stefan Bauersachs, Helmut Blum, Eckhard Wolf, Poul Hyttel
Author Affiliations +
Abstract

In an attempt to unveil molecular processes controlling the porcine placentation, we have investigated the pregnancy-induced gene expression in the endometrium using the Affymetrix GeneChip Porcine Genome Array. At Day 14 after insemination, at the time of initial placentation, samples were obtained from the endometrium of pregnant sows and sows inseminated with inactivated semen. Analysis of the microarray data revealed 263 genes to be significantly differentially expressed between the pregnant and nonpregnant sows. Most gene ontology terms significantly enriched at pregnancy had allocated more up-regulated genes than down-regulated genes. These terms included developmental process, transporter activity, calcium ion binding, apoptosis, cell motility, enzyme-linked receptor protein signaling pathway, positive regulation of cell proliferation, ion homeostasis, and hormone activity. Only the three terms oxidoreductase activity, lipid metabolic process, and organic acid metabolic process had an overrepresentation of down-regulated genes. A gene interaction network based on the genes identified in the gene ontology term developmental processes identified genes likely to be involved in the process of placentation. Pregnancy-specific localization of IL11RA to the surface epithelium of the endometrium suggests a role of interleukin 11 signaling in formation of the porcine epitheliochorial placenta. Furthermore, up-regulation of FGF9 mRNA in pregnant endometrium and localization of FGF9 to the apical cell domain of the glandular epithelium suggest the concept of endometrial FGF9 acting as an embryonic growth factor in the pig.

INTRODUCTION

Successful embryonic development is dependent on a complex molecular cross-talk between the embryo(s) and the maternal organism [1, 2]. In the pig, the embryo-maternal communication becomes evident at Day 12 of gestation, when the conceptuses release a surge of estrogen [3]. This embryonic signal for maternal recognition of pregnancy triggers changes in the prostaglandin metabolism of the endometrium to prevent regression of the corpora lutea by prostaglandin F2alpha (PGF2a) [4]. Recent research indicates that the estrogen signal from the conceptuses stimulates endometrial prostaglandin E2 (PGE2) synthesis. Combined with a positive PGE2 feedback loop in the endometrium, this synthesis leads to an increase in the PGE2:PGF2a ratio, which helps to overcome the luteolytic effect of PGF2a [5].

Following the embryonic signal for maternal recognition of pregnancy at Day 12, the porcine embryos remain free-floating in the uterine lumen until Days 13–14 of gestation, when they appose and subsequently attach to the uterine luminal epithelium [6]. Until this time of development, the embryos are supported from the endometrium by histiotrophic nutrition. From Days 15–20 of gestation, a gradual transition in embryonic nutrient takes place from being mainly histiotrophic to becoming primarily hemotrophic [6]. The functional changes are accompanied by extensive tissue remodeling of the endometrium [7, 8], where a pronounced vascularization is evident already from Day 13 of gestation [9, 10]. These processes, together with the attachment of the embryos to the surface epithelium of the uterus, initiate the placentation. The porcine placenta is, in contrast to that of many other species, epitheliochorial [11, 12]. That is, the placentation is noninvasive, and the placental barrier includes both the trophoblast and the epithelium of the endometrium.

During evolution, several subtypes of placentas have developed in eutherian mammals [11, 13]. However, the basic principles have not changed. Similar to all other subtypes of chorio-allantoic placentae, the epitheliochorial placenta is essential for the growth and development of the embryo and fetus. Despite the evolutionary changes in placental architecture, strong similarities also exist in the cellular functions between different placental subtypes [13]. Microarray investigations have been conducted successfully in several species in an attempt to disclose the mechanisms of placentation [1416]. A comparison of similar studies, however, reveals large differences in the gene expression profile between the species [16], indicating a highly species-specific molecular regulation of placentation. Differences in the embryo maternal communication are not necessarily related to differences in the placental structure. In sheep and cows, the signal for maternal recognition of pregnancy is interferon tau, produced by the conceptuses [17]. The porcine conceptuses also produce interferons; however, they do not appear to be antiluteolytic [18]. Instead, it has been suggested that interferon gamma, the main interferon produced by the porcine conceptuses, might be important in regulation of angiogenesis in the endometrium, similar to the human and murine placenta [19]. However, the pregnancy-specific role(s) of estrogen- and interferon-stimulated genes in the porcine endometrium remains largely conjectural. Furthermore, only a few genes, which are differentially expressed at gestation, have been investigated [18].

In the present study, we aim to investigate pregnancy-induced changes in the gene expression of the porcine endometrium in an attempt to identify specific mechanisms involved in the regulation of the initial porcine placentation.

MATERIALS AND METHODS

Animals

Three pairs of Danish Landrace and three pairs of Yorkshire sows, each pair from the same litter, were inseminated and slaughtered in pairs. One sow in each pair was inseminated with a standard dose of single Duroc semen, whereas the other littermate was inseminated with a dose of freeze-inactivated semen from the same boar. The animals were slaughtered at Day 14 postinsemination. The uteri were removed, and each uterine horn was flushed with PBS containing 1% fetal calf serum and subsequently opened longitudinally at the antimesometrial side. The sites of embryonic attachment were macroscopically visual in the endometrium on the mesometrial side in the form of hyperemic zones (Fig. 1). In pregnant sows, samples of the endometrium (lamina epithelialis, lamina propria, and tela submucosa, but not tunica muscularis) were taken from such hyperemic zones at three locations of each uterine horn: proximal (the end close to the ovaries), intermedial, and distal (next to the corpus uteri). Samples were taken from the endometrium of the nonpregnant animals at comparable locations. Tissue samples were transferred to RNAlater (Ambion) or 4% paraformaldehyde (PFA)/PBS within 25 min after slaughter. Samples in RNAlater were incubated overnight at 4°C and stored at −80°C until further processing. Samples in 4% PFA/PBS were incubated overnight at 4°C and stored in 1% PFA/PBS at 4°C until embedding in paraffin. All procedures involving animals were conducted in accordance with the national guidelines for agricultural animal care.

FIG. 1.

Hyperemic zone in the endometrium identifying the sites of implantation, as seen after uterine flushing.

i0006-3363-83-2-277-f01.tif

Microarray Analysis

Total RNA was isolated from endometrium using TRIzol (Invitrogen) according to the manufacturer's recommendations. An additional RNA selective precipitation was made using ammonium acetate. Purity and quantity of total RNA was measured by use of Nanodrop ND-1000 spectrophotometer (Thermo Fisher Scientific), and quality was assessed by electrophoresis. RNA samples were tested for genomic DNA contamination by performing a quantitative RT-PCR without reverse transcriptase in the RT step.

Equal amounts of total RNA from samples of proximal, intermedial, and distal endometrial sections were pooled for each animal. Probe synthesis and hybridization to the porcine GeneChip from Affymetrix were performed according to the recommendations of the manufacturer. Cel files were processed using the Robust Multi-Array normalization in the BioConductor package affy for R [20]. Microarray data have been deposited in the National Center for Biotechnology Information's Gene Expression Omnibus [21] and are accessible through GEO Series accession number GSE18641 ( http://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE18641). For quality control, normalized data were analyzed with a distance matrix and a heatmap based on pair-wise distances (BioConducter package geneplotter). Two samples were excluded from further analysis. Significance analysis was performed using the BioConductor package Linear Models for Microarray Data (LIMMA) [22]. Cut-off values were set to twofold difference in expression values and a false discovery rate of 1%. Identified probe sets were annotated by comparing two automated annotations [23, 24]. In case of disagreement between these annotations, the target sequences of the probe sets were manually annotated using BLAST [25]. If a gene was identified in the results by more than one probe set, the mean fold-change was calculated. Integrated analysis of different functional databases was done using the functional annotation clustering tool of the Database for Annotation, Visualization, and Integrated Discovery (DAVID) [26].

Furthermore, DAVID was used to calculate the fold-enrichment of identified Gene Ontology (GO) terms. The fold-enrichment for a particular GO term describes the ratio between the numbers of genes in the gene list belonging to a specific GO term and the total number of genes in the gene list, which have at least one GO annotation. This ratio is then compared to the ratio between the total number of genes in the GO term and the total number of genes in the human genome with at least one GO annotation. For example, if 8 (3.5%) of 223 genes in the gene list are involved in hormone activity and the ratio in the human genome is 127 (0.75%) of 16 968 genes associating with hormone activity, the fold-enrichment is roughly 3.5%/0.75% = 4.7. Fold-enrichment for the remaining GO terms was calculated in a similar manner.

An interaction network was built using Pathway Architect Software (version 2.0.1; Stratagene). Interactions provided by the Pathway Architect databases were cross-checked with literature, and additional interactions were assigned to the network. The human Entrez Gene IDs of the putative human orthologous genes for the identified porcine transcripts were used for these analyses.

Quantitative Real-Time RT-PCR

The same RNA as used for the microarrays was used to validate the results by real-time RT-PCR (see Table 4). One microgram of total RNA was reverse transcribed in a total volume of 25 μl, containing 1× buffer (Promega), 0.5 mM dNTPs (Fermentas), 16 ng/μL of random hexamer primers (Fermentas), 8 ng/μL (μM) of oligo-dT primers (Fermentas), 200 U of Moloney Murine Leukemia Virus Reverse Transcriptase (Promega), and 32 U of RNase H Minus, Point Mutant (Promega). Primers were designed to amplify specific fragments referring to selected regulated genes (Table 1). Amplified PCR fragments were sequenced with forward and reverse primers to verify the resulting PCR product. The specific melting point of the amplified product carried out within the LightCycler standard PCR protocol served as a verification of the product identity in the following PCR procedures. Real-time PCR reactions were performed in a 10-μl reaction mixture (2 μl of cDNA, 1 μM forward and reverse primer, and 1× Light Cycler DNA Master SYBR Green I [Roche]) using a real-time LightCycler 480 instrument (Roche). The annealing temperature was 60°C for all reactions. A no-template control was made in all runs, using water instead of cDNA.

TABLE 1.

Primer sequences used to amplify specific fragments of porcine transcripts.

i0006-3363-83-2-277-t01.tif

The reference gene ubiquitin B (UBB), which previously has been used as a reference gene in the endometrium [16], showed no statistical difference between pregnant and nonpregnant animals according to the microarray data verifying its suitability as reference gene. Expression levels of the selected target genes were calculated using relative quantification (LightCycler 480 software version 1.5.39). The data were normalized to the expression of UBB, and a calibrator was included to normalize between runs. Five biological and three technical replicates were made for all quantitative PCR reactions. Calculations of significant difference in the real-time RT-PCR expression data between samples from pregnant and nonpregnant animals were carried out in R using Wilcoxon signed-rank tests.

Immunohistochemistry

Endometrial samples stored in 1% PFA/PBS were dehydrated and embedded in paraffin. Paraffin sections (thickness, 5 μm) mounted on Superfrost Plus slides (Gerhard Menzel, Braunschweig, Germany) were dewaxed followed by blocking of endogenous peroxidase with 0.5% (v/v) H2O2 in 99% ETOH. After rehydration, sections were submitted to heat-induced epitope retrieval by microwave treatment in either 0.01 M sodium citrate buffer (pH 6.0) or in ethylenediaminetetra-acetic acid at either pH 8 or pH 9 (Table 2). Nonspecific binding of antibodies was blocked with PBS containing 1.25% normal horse serum (Vector Laboratories) and 1% bovine serum albumin (Sigma-Aldrich). Incubation with the primary antibodies was performed at room temperature for 2 h or overnight at 4°C (see Table 2 for details on specific antibodies). The antibodies against ERBB3 and IL6R were preadsorbed with Poly-l-lysine (Sigma-Aldrich) overnight at 4°C to reduce unspecific binding [27, 28]. Controls were made using preadsorption controls for all polyclonal antibodies using the specific blocking peptides IL6R (Santa Cruz), IL11RA (Santa Cruz), ERBB3 (Santa Cruz), LIFR (R&D Systems), and isotype controls for monoclonal antibodies using mouse IgG2a and IgG3 (Abcam). Detection of the primary antibodies was performed using ImmPRESS (Vector Laboratories). The ImmPRESS secondary antibodies were preadsorbed with 0.1% normal swine serum (DAKO) at 4°C overnight to minimize background. The color reaction was developed with an AEC-kit (Zymed Laboratories). Sections were counterstained with Mayer hematoxylin before mounting in Glycergel (Dako).

TABLE 2.

Primary antibodies, HIER treatment, and incubation time.

i0006-3363-83-2-277-t02.tif

RESULTS

Microarray Analysis

A heatmap analysis of the normalized microarray data (data not shown) revealed one pregnant and one nonpregnant animal as having highly abnormal gene expression when compared to the expression-pattern of the remaining 10 animals. One of these two animals presented signs of weak endometritis at the time of sample collections. The data from these two animals were not included in the further analyses.

When comparing the gene expression in the endometrium of pregnant sows (n = 5) with that of nonpregnant sows (n = 5), 323 probe sets were identified to be more than twofold significantly differentially expressed (Supplemental Tables S1 and S2 available at  www.biolreprod.org). Annotation of the 323 probe sets revealed 146 genes expressed at higher levels in pregnant animals (referred to hereafter as up-regulated genes). Another 117 genes were detected with lower expression in the pregnant animals (referred to hereafter as down-regulated genes). From 18 probe sets, which could not be annotated, 13 were up-regulated, and five were down-regulated.

The 263 genes identified to be differentially expressed were analyzed using the functional annotation chart and the functional annotation clustering tool from DAVID [26]. The analyses were based on the major categories Biological Process and Molecular Function. A combination of the most informative significantly enriched GO terms from the resulting functional clusters and GO terms that were not included in the functional clustering but were found to be significantly enriched by the functional annotation chart have been summarized in Table 3. Of the GO terms that were significantly enriched, most terms had allocated more up-regulated genes than down-regulated genes at pregnancy. Among these were GO terms such as developmental process, apoptosis, cell motility, and positive regulation of cell proliferation, which are likely to be enriched because of remodeling of the endometrium in relation to the placentation. Another group of enriched GO terms contains genes involved in formation and regulation of histiotrophe (e.g., transporter activity, calcium ion binding, and ion homeostasis). Furthermore, GO terms describing different aspects of communication and signaling, response to external stimulus, enzyme-linked receptor protein signaling pathway, and hormone activity were present in the group of significantly enriched GO terms, with an over-representation of up-regulated genes. Only three GO terms had an overrepresentation of down-regulated genes. These GO terms were oxidoreductase activity, lipid metabolic process, and organic acid metabolic process. The enriched terms with most down-regulated genes included genes involved in steroid hormone and prostaglandin metabolism.

TABLE 3.

Significantly enriched gene ontology terms.

i0006-3363-83-2-277-t03.tif

TABLE 4.

Validation of microarray results using real-time RT-PCR.

i0006-3363-83-2-277-t04.tif

Using Pathway Architect software (version 2.0.1; Stratagene), an interaction network was built based on the genes present in the GO term developmental process (Fig. 2). This network includes possible interactions between genes involved in cytokine and growth factor-mediated cell signaling. Among the genes identified in our results contributing to the cytokine-mediated signaling are genes related to the interleukin families. These include the receptors IL6R, LIFR, and IL11RA. Both IL6R and LIFR were found to be up-regulated, whereas IL11RA was down-regulated. Among the growth factor-related genes, FGF9 was up-regulated. The FGF9 receptor FGFR3 was, however, down-regulated. Another growth factor receptor, ERBB3, was identified by the microarray to be up-regulated. A possible interaction was found by the network between ERBB3 and the mucin MUC4. The mucin MUC4 was approximately ninefold higher expressed in the endometrium of the pregnant animals compared to the expression in the nonpregnant animals.

FIG. 2.

Interaction network based on differentially expressed genes belonging to the GO term developmental process. Genes highlighted with red frames were up-regulated at pregnancy. Genes highlighted with green frames were down-regulated at pregnancy. The numbers under the gene symbols indicate the fold-change in mRNA expression. Genes framed with a thin black line were not identified to be differentially expressed in the endometrium when comparing pregnant and nonpregnant sows. Purple polygons illustrate gene families. Green circles illustrate hormones. Arrows with a green square indicate positive (+) or negative (−) regulation. Lines with a blue square indicate binding. Arrows with a purple circle indicate gene family member.

i0006-3363-83-2-277-f02.tif

Quantitative RT-PCR

Based on the interaction network, seven genes (IL6R, LIFR, IL11RA, MUC4, ERBB3, FGF9, and FGFR3) were selected for mRNA quantification by quantitative RT-PCR (Table 4). The results obtained by array hybridization were clearly confirmed, and more accurate gene expression differences were obtained.

Immunohistochemistry

The localization of IL6R, LIFR, IL11RA, MUC4, ERBB3, FGF9, and FGFR3 proteins was investigated in the endometrial tissue (Fig. 3).

FIG. 3.

Immunohistochemical localization of IL6R (a and b), LIFR (d and e), IL11RA (g and h), MUC4 (j and k), ERBB3 (m and n), FGF9 (p and q), FGFR3 (s and t), in endometrial tissue from nonpregnant (a, d, g, j, m, p, and s) and pregnant (b, e, h, k, n, q, and t) sows. Corresponding negative controls: IL6R preadsorption control (c), LIFR preadsorption control (f), IL11RA preadsorption control (i), murine IgG2a Isotype control (l), ERBB3 preadsorption control (o), murine IgG3 Isotype control (r), and murine IgG2a Isotype control (u). DG, deep glands; LE, luminal epithelium; S, stroma; SG, superficial glands. Bar = 200 μm (af and ju) and 500 μm (g–i).

i0006-3363-83-2-277-f03.tif

IL6R.

Strong staining for IL6R was seen in surface epithelium, superficial and deep glandular epithelium, as well as stroma cells, endothelial cells, and leukocytes. No differences were observed in the localization of IL6R between pregnant and nonpregnant animals (Fig. 3, a and b).

LIFR.

Staining for LIFR was present in the surface epithelium as well as in superficial and deep glandular epithelium, with a tendency for weaker staining in the deeper glands. Staining for LIFR was also seen in some stromal cells, endothelial cells, and leukocytes. No differences were observed in the protein localization of LIFR between pregnant and nonpregnant animals (Fig. 3, d and e).

IL11RA.

Staining for IL11RA was found in superficial glandular epithelium and in most of the deeper glandular epithelium in both pregnant and nonpregnant animals (Fig. 3, g and h). Furthermore, IL11RA was detected in the surface epithelium of all pregnant animals, whereas it was only detected in few cells at this location in nonpregnant animals.

MUC4.

Strong staining for MUC4 was observed in the surface epithelium as well as in superficial and deep glandular epithelium. In the surface epithelium, MUC4 staining was primarily cytoplasmic, whereas in the glandular epithelium, MUC4 was strongly localized to the extracellular matrix and the apical domain of the cells. No differences were seen in the localization of the protein between pregnant and nonpregnant animals (Fig. 3, j and k).

ERBB3.

Staining for ERBB3 was found in the surface epithelial cells as well as in superficial glandular epithelium. Weaker staining was observed in the deeper-lying glandular epithelium. No differences were observed in the protein localization of ERBB3 between pregnant and nonpregnant animals (Fig. 3, m and n).

FGF9.

Strong staining for FGF9 was present in the apical cell domain of superficial and deep glandular epithelium. Weaker staining was observed in the surface epithelium and stromal cells. No differences in the localization of the protein were observed between pregnant and nonpregnant animals (Fig. 3, p and q).

FGFR3.

Staining for FGFR3 was primarily localized to the glandular epithelium and, to some degree, the surface epithelium. No differences were seen in the localization of FGFR3 between pregnant and nonpregnant animals (Fig. 3, s and t); however, a tendency was found for higher staining intensity in the luminal epithelium of the pregnant animals compared to the nonpregnant.

DISCUSSION

In the present study, we successfully identified 263 genes that were significantly differentially expressed in the porcine endometrium between pregnant and nonpregnant sows. Based on an interaction network, we selected the genes IL6R, LIFR, IL11RA, MUC4, ERBB3, FGF9, and FGFR3, which are potential candidate genes for regulation of placentation.

The interleukin 6 family of cytokines is well known to be a key regulator of implantation and placentation. In mice, these cytokines as well as their receptors are required for normal implantation and embryonic survival [29]. In the present study, we identified the specific receptors IL6R, LIFR, and IL11RA to be significantly differentially expressed; however, neither the cytokines nor the common signal transducer, IL6ST, was among the differentially expressed genes. This indicates that endometrial regulation of signaling in this cytokine family is, to a great extent, controlled by the expression of the specific receptors.

The receptor IL6R was localized in both surface and glandular epithelium, as well as to cells of the stroma, in both pregnant and nonpregnant animals. This localization of the IL6R in the porcine endometrium is similar to that reported in human endometrium [30].

LIFR was similar to IL6R localized to both surface and glandular epithelium, as well as to some stromal cells of the porcine endometrium. The LIFR mRNA expression was similar to that in mouse [31], increased in pregnant animals; however, no apparent differences were noticed in the localization of the LIFR between pregnant and nonpregnant animals. This localization of LIFR is similar to that reported in the endometrium of the rhesus monkey during the luteal phase and peri-implantation period [32]. In human and mouse, LIFR mRNA was shown to be exclusively expressed in the surface epithelium at the time of implantation [31, 33], and only weak staining for LIFR has been reported in human glandular epithelium [30, 34].

In contrast to the two other receptors in the IL6 cytokine family, IL11RA was significantly lower expressed in the endometrium from the pregnant animals compared to the nonpregnant (Table 4). In human endometrium, it is without any cyclic variation [35].

Interestingly, a marked increase in the number of cells stained for IL11RA protein was identified in the surface epithelium of pregnant compared to nonpregnant sows (Fig. 3), despite the down-regulation of IL11RA mRNA in the endometrial tissue samples from pregnant sows. The role of IL11 has been investigated in mice [36] and humans, in which IL11 stimulates migration of extravillous trophoblast cells but prevents their invasion [37, 38]. The crucial role of IL11 in prevention of invasion is also supported by studies in Il11ra knock-out mice. Hence, mice lacking Il11ra were infertile because of failure in the decidualization and, possibly, overinvasiveness of the trophoblast [36, 39]. Porcine embryos are capable of invading tissues when placed on ectopic locations [40]. Hence, noninvasiveness of the porcine implantation has been suggested to be controlled by the endometrium rather than by the trophoblast. The pregnancy-dependent localization of IL11RA to the surface epithelium suggests that IL11 signaling may play a role in the porcine endometrium by inhibiting the trophoblast invasion through the surface epithelium, similar to what is described in the mouse for the decidual zone.

It has been suggested previously that MUC4 expressed by the porcine endometrium also plays a role in protecting the surface epithelium from invasion [41]. Accordingly, in the present study, we identified MUC4 to have significantly higher expression in the pregnant compared to the nonpregnant endometrium. Experiments in rodents, on the contrary, show a down-regulation of mucins just before implantation of the blastocyst [42, 43]. The different placentation types between rodents and pigs, combined with the different expression pattern of MUC4, supports the idea that MUC4 might act as a protector of the porcine surface epithelium against invasion of the conceptuses [41].

On the contrary, the expression of MUC4 in the human endometrium is highest during the follicular (proliferative) phase of the menstrual cycle rather than during the luteal (secretory) phase [44]. Moreover, indications exist that MUC4 is not related to invasiveness in humans [45]. On the other hand, increased expression levels of MUC4 in cancers and hyperplastic diseases of the uterus instead support a role for MUC4 in regulating cell proliferation [44, 46].

The role of MUC4 as a regulator of differentiation and a modulator of proliferation can be explained by its C-terminal structure. It contains two epidermal growth factor (EGF)-like domains, which can interact with ERBB2 [47]. The receptor ERBB2 exhibits a strong kinase activity and is the preferred partner in forming dimers with other ERBB members [48]. The MUC4-ERBB2 complex interacts readily with ERBB3 in the presence of neuregulin, resulting in robust activation of the PI3K pathway and cell proliferation [49]. In the present study, we identified ERBB3 mRNA to have significantly higher expression in endometrium of pregnant compared to nonpregnant sows.

The mRNA coding for the growth factor FGF9 was significantly higher expressed in pregnant animals, and FGF9 has been identified previously as a growth factor in human endometrium [50]. The localization is, however, different. In pigs, the strongest staining for FGF9 was observed at the apical domain of the glandular epithelial cells; in humans, glandular epithelium only expresses the gene at low levels [50].

FGF9 binds with high affinity to FGFR2 and FGFR3 [51, 52]. In the human endometrium, FGF9 has no effect on proliferation of the epithelium despite the localization of FGFR2 in epithelial cells [50], indicating that FGF9 preferably acts through FGFR3 in the human endometrium. This may also be the case in the porcine endometrium, because another fibroblastic growth factor, FGF7, has been identified as a likely candidate for FGFR2-mediated signaling [53]. Interestingly, the expression of FGFR3 mRNA was significantly lower in the pregnant sows, and FGFR3 staining was strongest in the glandular epithelium. This indicates the involvement of factors other than FGF9 in the regulation of proliferation of the surface epithelium. Moreover, FGF9 might act in an endocrine manner [5456]. Hence, it could be speculated that an increase in secreted FGF9 from the endometrial glands during pregnancy may not be related to actions within the endometrium but, rather, may be implicated in the embryo-maternal communication.

In conclusion, we identified 263 genes to be differentially regulated in the endometrium of pregnant versus nonpregnant sows at the time of initial placentation. Furthermore, we showed a pregnancy-specific localization of IL11RA to the surface epithelium of the endometrium, suggesting that IL11 signaling may play an important role in formation of the porcine epitheliochorial placenta. We also showed that FGF9 is up-regulated in pregnant endometrium, whereas its receptor, FGFR3, is down-regulated. Combined with the localization of FGF9 to the apical cell domain of the glandular epithelium, this finding makes it intriguing to speculate that FGF9 of endometrial origin functions as an embryonic growth factor in the pig.

Acknowledgements

The authors thank Danish Pig Production for arranging housing and insemination of the animals used in the present study.

REFERENCES

1.

P Vigano, S Mangioni, F Pompei, and I Chiodo . Maternal-conceptus cross talk—a review. Placenta 2003. 24 (suppl B)::S56–S61. Google Scholar

2.

E Wolf, GJ Arnold, S Bauersachs, HM Beier, H Blum, R Einspanier, T Frohlich, A Herrler, S Hiendleder, S Kolle, K Prelle, HD Reichenbach, et al . Embryo-maternal communication in bovine—strategies for deciphering a complex cross-talk. Reprod Domest Anim 2003. 38:276–289. Google Scholar

3.

RD Geisert, MT Zavy, RJ Moffatt, RM Blair, and T Yellin . Embryonic steroids and the establishment of pregnancy in pigs. J Reprod Fertil Suppl 1990. 40:293–305. Google Scholar

4.

AJ Ziecik . Old, new and the newest concepts of inhibition of luteolysis during early pregnancy in pig. Domest Anim Endocrinol 2002. 23:265–275. Google Scholar

5.

A Waclawik, HN Jabbour, A Blitek, and AJ Ziecik . Estradiol-17beta, prostaglandin E2 (PGE2), and the PGE2 receptor are involved in PGE2 positive feedback loop in the porcine endometrium. Endocrinology 2009. 150:3823–3832. Google Scholar

6.

V Dantzer . Electron microscopy of the initial stages of placentation in the pig. Anat Embryol (Berl ) 1985. 172:281–293. Google Scholar

7.

LP Reynolds, PP Borowicz, KA Vonnahme, ML Johnson, AT Grazul-Bilska, JM Wallace, JS Caton, and DA Redmer . Animal models of placental angiogenesis. Placenta 2005. 26:689–708. Google Scholar

8.

A Cencic, M Guillomot, S Koren, and BC La . Trophoblastic interferons: do they modulate uterine cellular markers at the time of conceptus attachment in the pig? Placenta 2003. 24:862–869. Google Scholar

9.

JL Keys, GJ King, and TG Kennedy . Increased uterine vascular permeability at the time of embryonic attachment in the pig. Biol Reprod 1986. 34:405–411. Google Scholar

10.

JL Keys and GJ King . Morphological evidence for increased uterine vascular permeability at the time of embryonic attachment in the pig. Biol Reprod 1988. 39:473–487. Google Scholar

11.

DE Wildman, C Chen, O Erez, LI Grossman, M Goodman, and R Romero . Evolution of the mammalian placenta revealed by phylogenetic analysis. Proc Natl Acad Sci U S A 2006. 103:3203–3208. Google Scholar

12.

AM Carter and AC Enders . Comparative aspects of trophoblast development and placentation. Reprod Biol Endocrinol 2004. 2:46..  Google Scholar

13.

BA Croy, J Wessels, N Linton, and C Tayade . Comparison of immune cell recruitment and function in endometrium during development of epitheliochorial (pig) and hemochorial (mouse and human) placentas. Placenta 2009. 30 (suppl A)::S26–S31. Google Scholar

14.

D Haouzi, K Mahmoud, M Fourar, K Bendhaou, H Dechaud, VJ De, T Reme, D Dewailly, and S Hamamah . Identification of new biomarkers of human endometrial receptivity in the natural cycle. Hum Reprod 2009. 24:198–205. Google Scholar

15.

J Reese, SK Das, BC Paria, H Lim, H Song, H Matsumoto, KL Knudtson, RN DuBois, and SK Dey . Global gene expression analysis to identify molecular markers of uterine receptivity and embryo implantation. J Biol Chem 2001. 276:44137–44145. Google Scholar

16.

S Bauersachs, SE Ulbrich, K Gross, SE Schmidt, HH Meyer, H Wenigerkind, M Vermehren, F Sinowatz, H Blum, and E Wolf . Embryo-induced transcriptome changes in bovine endometrium reveal species-specific and common molecular markers of uterine receptivity. Reproduction 2006. 132:319–331. Google Scholar

17.

TE Spencer, GA Johnson, FW Bazer, RC Burghardt, and M Palmarini . Pregnancy recognition and conceptus implantation in domestic ruminants: roles of progesterone, interferons and endogenous retroviruses. Reprod Fertil Dev 2007. 19:65–78. Google Scholar

18.

GA Johnson, FW Bazer, RC Burghardt, TE Spencer, G Wu, and KJ Bayless . Conceptus-uterus interactions in pigs: endometrial gene expression in response to estrogens and interferons from conceptuses. Soc Reprod Fertil Suppl 2009. 66:321–332. Google Scholar

19.

SP Murphy, C Tayade, AA Ashkar, K Hatta, J Zhang, and BA Croy . Interferon gamma in successful pregnancies. Biol Reprod 2009. 80:848–859. Google Scholar

20.

R Development Core Team. R: A language and environment for statistical computing. Vienna, Austria: R Foundation for Statistical Computing. 2005.  Google Scholar

21.

R Edgar, M Domrachev, and AE Lash . Gene Expression Omnibus: NCBI gene expression and hybridization array data repository. Nucleic Acids Res 2002. 30:207–210. Google Scholar

22.

GK Smyth . Linear models and empirical Bayes methods for assessing differential expression in microarray experiments. Stat Appl Genet Mol Biol 2004. 3:Article3..  Google Scholar

23.

S Tsai, JP Cassady, BA Freking, DJ Nonneman, GA Rohrer, and JA Piedrahita . Annotation of the Affymetrix porcine genome microarray. Anim Genet 2006. 37:423–424. Google Scholar

24.

V Hintermair . Comparative analysis of Bos taurus and Homo sapiens DNA microarrays. Munich, Germany: LMU München. 2007. Thesis.  Google Scholar

25.

SF Altschul, W Gish, W Miller, EW Myers, and DJ Lipman . Basic local alignment search tool. J Mol Biol 1990. 215:403–410. Google Scholar

26.

G Dennis Jr, BT Sherman, DA Hosack, J Yang, W Gao, HC Lane, and RA Lempicki . DAVID: Database for Annotation, Visualization, and Integrated Discovery. Genome Biol 2003. 4:3..  Google Scholar

27.

LI Larsson, L Scopsi, D Modena, G Racchetti, and YM Galante . Immunocytochemical characterization of mouse monoclonal ACTH antibodies with a note on staining conditions and control procedures. Histochemistry 1989. 91:115–122. Google Scholar

28.

K Schauser and LI Larsson . Programmed cell death and cell extrusion in rat duodenum: a study of expression and activation of caspase-3 in relation to C-jun phosphorylation, DNA fragmentation and apoptotic morphology. Histochem Cell Biol 2005. 124:237–243. Google Scholar

29.

LA Salamonsen, E Dimitriadis, and L Robb . Cytokines in implantation. Semin Reprod Med 2000. 18:299–310. Google Scholar

30.

JR Sherwin, SK Smith, A Wilson, and AM Sharkey . Soluble gp130 is up-regulated in the implantation window and shows altered secretion in patients with primary unexplained infertility. J Clin Endocrinol Metab 2002. 87:3953–3960. Google Scholar

31.

H Ni, NZ Ding, MJ Harper, and ZM Yang . Expression of leukemia inhibitory factor receptor and gp130 in mouse uterus during early pregnancy. Mol Reprod Dev 2002. 63:143–150. Google Scholar

32.

ZP Yue, ZM Yang, P Wei, SJ Li, HB Wang, JH Tan, and MJ Harper . Leukemia inhibitory factor, leukemia inhibitory factor receptor, and glycoprotein 130 in rhesus monkey uterus during menstrual cycle and early pregnancy. Biol Reprod 2000. 63:508–512. Google Scholar

33.

EB Cullinan, SJ Abbondanzo, PS Anderson, JW Pollard, BA Lessey, and CL Stewart . Leukemia inhibitory factor (LIF) and LIF receptor expression in human endometrium suggests a potential autocrine/paracrine function in regulating embryo implantation. Proc Natl Acad Sci U S A 1996. 93:3115–3120. Google Scholar

34.

L Aghajanova, A Stavreus-Evers, Y Nikas, O Hovatta, and BM Landgren . Coexpression of pinopodes and leukemia inhibitory factor, as well as its receptor, in human endometrium. Fertil Steril 2003. 79 (suppl 1)::808–814. Google Scholar

35.

E Dimitriadis, CA White, RL Jones, and LA Salamonsen . Cytokines, chemokines and growth factors in endometrium related to implantation. Hum Reprod Update 2005. 11:613–630. Google Scholar

36.

L Robb, R Li, L Hartley, HH Nandurkar, F Koentgen, and CG Begley . Infertility in female mice lacking the receptor for interleukin 11 is due to a defective uterine response to implantation. Nat Med 1998. 4:303–308. Google Scholar

37.

P Paiva, LA Salamonsen, U Manuelpillai, C Walker, A Tapia, EM Wallace, and E Dimitriadis . Interleukin-11 promotes migration, but not proliferation, of human trophoblast cells, implying a role in placentation. Endocrinology 2007. 148:5566–5572. Google Scholar

38.

P Paiva, LA Salamonsen, U Manuelpillai, and E Dimitriadis . Interleukin 11 inhibits human trophoblast invasion indicating a likely role in the decidual restraint of trophoblast invasion during placentation. Biol Reprod 2009. 80:302–310. Google Scholar

39.

L Bao, S Devi, J Bowen-Shauver, S Ferguson-Gottschall, L Robb, and G Gibori . The role of interleukin-11 in pregnancy involves up-regulation of alpha2-macroglobulin gene through janus kinase 2-signal transducer and activator of transcription 3 pathway in the decidua. Mol Endocrinol 2006. 20:3240–3250. Google Scholar

40.

CA Samuel and JS Perry . The ultrastructure of pig trophoblast transplanted to an ectopic site in the uterine wall. J Anat 1972. 113:139–149. Google Scholar

41.

AD Ferrell, JR Malayer, KL Carraway, and RD Geisert . Sialomucin complex (Muc4) expression in porcine endometrium during the estrous cycle and early pregnancy. Reprod Domest Anim 2003. 38:63–65. Google Scholar

42.

GA Surveyor, SJ Gendler, L Pemberton, SK Das, I Chakraborty, J Julian, RA Pimental, CC Wegner, SK Dey, and DD Carson . Expression and steroid hormonal control of Muc-1 in the mouse uterus. Endocrinology 1995. 136:3639–3647. Google Scholar

43.

RR McNeer, CA Carraway, NL Fregien, and KL Carraway . Characterization of the expression and steroid hormone control of sialomucin complex in the rat uterus: implications for uterine receptivity. J Cell Physiol 1998. 176:110–119. Google Scholar

44.

F Alameda, R Mejias-Luque, M Garrido, and C de Bolo´s . Mucin genes (MUC2, MUC4, MUC5AC, and MUC6) detection in normal and pathological endometrial tissues. Int J Gynecol Pathol 2007. 26:61–65. Google Scholar

45.

NA Bersinger, DM Wunder, MH Birkhauser, and MD Mueller . Gene expression in cultured endometrium from women with different outcomes following IVF. Mol Hum Reprod 2008. 14:475–484. Google Scholar

46.

A Lopez-Ferrer, F Alameda, C Barranco, M Garrido, and C de Bolo´s . MUC4 expression is increased in dysplastic cervical disorders. Hum Pathol 2001. 32:1197–1202. Google Scholar

47.

KL Carraway III, EA Rossi, M Komatsu, SA Price-Schiavi, D Huang, PM Guy, ME Carvajal, N Fregien, CA Carraway, and KL Carraway . An intramembrane modulator of the ErbB2 receptor tyrosine kinase that potentiates neuregulin signaling. J Biol Chem 1999. 274:5263–5266. Google Scholar

48.

D Graus-Porta, RR Beerli, JM Daly, and NE Hynes . ErbB-2, the preferred heterodimerization partner of all ErbB receptors, is a mediator of lateral signaling. EMBO J 1997. 16:1647–1655. Google Scholar

49.

M Funes, JK Miller, C Lai, KL Carraway III, and C Sweeney . The mucin Muc4 potentiates neuregulin signaling by increasing the cell-surface populations of ErbB2 and ErbB3. J Biol Chem 2006. 281:19310–19319. Google Scholar

50.

SJ Tsai, MH Wu, HM Chen, PC Chuang, and LY Wing . Fibroblast growth factor-9 is an endometrial stromal growth factor. Endocrinology 2002. 143:2715–2721. Google Scholar

51.

D Hecht, N Zimmerman, M Bedford, A Avivi, and A Yayon . Identification of fibroblast growth factor 9 (FGF9) as a high affinity, heparin dependent ligand for FGF receptors 3 and 2 but not for FGF receptors 1 and 4. Growth Factors 1995. 12:223–233. Google Scholar

52.

DM Ornitz, J Xu, JS Colvin, DG McEwen, CA MacArthur, F Coulier, G Gao, and M Goldfarb . Receptor specificity of the fibroblast growth factor family. J Biol Chem 1996. 271:15292–15297. Google Scholar

53.

K Wollenhaupt, H Welter, KP Brussow, and R Einspanier . Regulation of endometrial fibroblast growth factor 7 (FGF-7) and its receptor FGFR2IIIb in gilts after sex steroid replacements, and during the estrous cycle and early gestation. J Reprod Dev 2005. 51:509–519. Google Scholar

54.

K Miyakawa, K Hatsuzawa, T Kurokawa, M Asada, T Kuroiwa, and T Imamura . A hydrophobic region locating at the center of fibroblast growth factor-9 is crucial for its secretion. J Biol Chem 1999. 274:29352–29357. Google Scholar

55.

JM Revest, L DeMoerlooze, and C Dickson . Fibroblast growth factor 9 secretion is mediated by a noncleaved amino-terminal signal sequence. J Biol Chem 2000. 275:8083–8090. Google Scholar

56.

M Miyamoto, K Naruo, C Seko, S Matsumoto, T Kondo, and T Kurokawa . Molecular cloning of a novel cytokine cDNA encoding the ninth member of the fibroblast growth factor family, which has a unique secretion property. Mol Cell Biol 1993. 13:4251–4259. Google Scholar

Notes

[1] Financial disclosure Supported by the Danish Pig Production, the Danish Agency for Science, Technology and Innovation grant 09-060623/FTP, and the German Federal Ministry for Education and Research (BMBF, FUGATOplus Compendium). Microarray data have been submitted to the NCBI Gene Expression Omnibus with accession no. GSE18641.

Esben Østrup, Stefan Bauersachs, Helmut Blum, Eckhard Wolf, and Poul Hyttel "Differential Endometrial Gene Expression in Pregnant and Nonpregnant Sows," Biology of Reproduction 83(2), 277-285, (14 April 2010). https://doi.org/10.1095/biolreprod.109.082321
Received: 13 November 2009; Accepted: 1 March 2010; Published: 14 April 2010
KEYWORDS
Endometrium
ERBB3
FGF9
FGFR3
IL11RA
IL6R
implantation
Back to Top