ArticlePDF Available
Effect of conceptus on expression of prostaglandin F
2
a
receptor in the
porcine endometrium
Piotr Kaczynski, Agnieszka Waclawik
*
Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland
article info
Article history:
Received 8 August 2012
Received in revised form 10 December 2012
Accepted 15 December 2012
Keywords:
Endometrium
Conceptus
Pregnancy
Prostaglandin F
2
a
receptor
Pig
Prostaglandin F
2
a
abstract
Increased synthesis of prostaglandin F
2
a
(PGF
2
a
) in the endometrium and conceptus during
the implantation period results in elevated concentration of PGF
2
a
in the uterine lumen in
pregnant gilts. PGF
2
a
exerts its effects through PGF
2
a
receptor (PTGFR), a Gprotein-coupled
receptor. However, besides studies concerning the function of PTGFR in endometrial
abnormalities, the role of PTGFR in the endometrium during early pregnancy has not been
elucidated. Therefore, the aim of this study was: (1) to evaluate the prole of PTGFR gene
and protein expression in the porcine endometrium during early pregnancy and the estrous
cycle; (2) to determine if the effect of conceptus on PTGFR expression is dependent on type
of endometrial cellsdluminal epithelial (LE) or stromal (ST) cells; and (3) to elucidate if the
putative effect of conceptus on endometrial PTGFR expression is mediated by estrogen
receptor. We evaluated the expression pattern of PTGFR gene and protein in the endome-
trium during day 9, 11,12, 15, and 18 of the estrous cycle and pregnancy (N ¼46 per group).
The expression of PTGFR mRNA was greateron day 18 of pregnancy and the estrous cycle (vs.
days 915 of the estrous cycle and pregnancy, P <0.05). Expression of PTGFR protein was
approximately 10-fold upregulated in the endometrium on day 15 of pregnancy when
compared with day 15 of the estrous cycle (P <0.01). Endometrial expression of PTGFR
protein increased from day 12 to 18 of pregnancy (P <0.05). PTGFR mRNA was expressed in
LE and ST cells. In a subsequent experiment, we used a coculture model in which LE cells
were cultured on collagen-coated inserts placed in wells plated with ST cells. Day 11 or 15
conceptus-exposed medium (CEM) elevated expression of PTGFR mRNA (2- and 1.5-fold,
respectively, P <0.05) in LE cells cocultured with ST cells. CEM did not have an effect on
PTGFR mRNA expression in ST cells. The 11-day CEM-induced increase of PTGFR mRNA was
abolished by incubation of LE cells in the presence of the estrogen receptor antagonist (ICI-
182,780; P <0.01). Summarizing, the conceptus upregulated expression of PTGFR in the
endometrium during the implantation period. Moreover, this study indicates that expres-
sion of PTGFR gene was elevated in LE cells of endometrium by embryonic signal of estradiol.
Our results suggest para- and autocrine effects of PGF
2
a
through its receptor PTGFR in the
porcine endometrium, especially in luminal epithelium which is in direct contact with the
conceptus during the implantation period.
Ó2013 Elsevier Inc. All rights reserved.
1. Introduction
Prostaglandin F
2
a
(PGF
2
a
) as a tissue specic hormone is
involved in many physiologic processes. In most mammals,
PGF
2
a
is claimed to be the main luteolytic factor which
controls progesterone secretion by acting on luteal cells [14].
PGF
2
a
exerts its function through PGF
2
a
receptor
(PTGFR) which belongs to the Gprotein-coupled receptor
family. Activated PTGFR binds to Gq protein, that leads to
protein kinase C-mediated elevation of intracellular Ca
2þ
concentration [5]. PTGFR can also activate other signal
*Corresponding author. Tel.: þ48 89 5357422; fax: þ48 89 5357421.
E-mail address: waclawik@pan.olsztyn.pl (A. Waclawik).
Contents lists available at SciVerse ScienceDirect
Theriogenology
journal homepage: www.theriojournal.com
0093-691X/$ see front matter Ó2013 Elsevier Inc. All rights reserved.
http://dx.doi.org/10.1016/j.theriogenology.2012.12.003
Theriogenology 79 (2013) 784790
transduction pathways such as Rho, focal adhesion kinase
signaling, and mitogen-activated protein kinase [6,7].
PTGFR is approximately a 40 kDa protein with a seven
transmembrane domain that is highly conserved among
prostanoid receptors. PTGFR gene consists of three exons
and two introns [8].
In the porcine corpus luteum (CL) the expression of
PTGFR is lower on days 7 and 8 of the estrous cycle, which is
believed to protect luteal cells from the lytic effects of PGF
2
a
.
The upregulation of PTGFR expression in the porcine CL on
days 13 and 15 of the estrous cycle coincides with onset of
luteolysis [9]. However, during early pregnancy in the pig,
estradiol (E
2
) produced by the conceptus in a biphasic
manner on days 11 to 12 and 15 to 30 of pregnancy prolongs
luteal progesterone production [10].
Interestingly, average concentration, amplitude, and the
frequency of PGF
2
a
pulses are lower in the porcine utero-
ovarian vein on day 13 to 17 of pregnancy than in pigs on
day 13 to 17 of the estrous cycle [11]. To avoid luteolytic
inuence of PGF
2
a
on the CL in ovary, PGF
2
a
is sequestered
in the uterus probably by redirection of its secretion from
the uterine venous drainage (endocrine) into the uterine
lumen (exocrine) by conceptus signaldE
2
[12]. The anti-
luteolytic mechanism could also involve the retrograde
transfer of PGF
2
a
from the venous blood and uterine lymph
into the uterine lumen and ability of uterine veins and
arterial walls to accumulate PGF
2
a
[13]. Another potential
mechanism by which the conceptus can contribute to
prevention of luteolysis is by changing prostaglandin (PG)
synthesis in favor of PGE
2
during the maternal recognition
of pregnancy, on days 10 to 13 in the pig [1416]. PGE
2
plays
an important role as a luteoprotective factor and a local
mediator of embryo signal on uterus [15,1720]. Our
previous study indicated the existence of a putative PGE
2
autoamplication loop in the endometrium that can be
involved in an increase of the PGE
2
/PGF
2
a
ratio on days 10
to 13 of pregnancy [16]. In this period, synthesis of PGF
2
a
seems to be undesired because of its luteolytic effect on the
CL. Indeed, our results indicate low expression of PGF
2
a
synthase mRNA and protein in the porcine embryo and
endometrium on days 10 to 13 of pregnancy. However, in
later periods, during implantation and early placentation
(days between 14 and 25 of pregnancy) expression of PGF
2
a
synthase signicantly increases in the endometrium and
conceptus [14,15] resulting in elevated PGF
2
a
content in the
uterine lumen [21]. To our knowledge, until now there is no
information concerning the role of an increased amount of
PGF
2
a
secreted into the uterine lumen during early preg-
nancy in the pig, or in any other species. Therefore, a key
question arises as to whether PTGFR is present in the
porcine endometrium during early pregnancy, especially at
the implantation period, when embryo and endometrium
secrete PGF
2
a
[14,15]. Hereby, the aim of this study was: (1)
to evaluate the prole of PTGFR gene and protein expres-
sion in the porcine endometrium during early pregnancy
and the estrous cycle; (2) to determine if the possible effect
of conceptus on PTGFR expression is dependent on the type
of endometrial cellsthe luminal epithelial (LE) and stromal
(ST) cells; and (3) to elucidate if the putative effect of
conceptus on PTGFR expression in endometrial cells is
mediated by estrogen receptor in the endometrium.
2. Materials and methods
2.1. Tissue collection
Peripubertal crossbred gilts (Polish Landrace Duroc)
of similar age (55.5 months) and genetic background from
one commercial herd were observed daily for onset of
estrus. After two natural estrous cycles, gilts were divided
randomly in two groups: cyclic and pregnant. Gilts
assigned to the pregnant group were inseminated at 12 and
24 hours after detection of estrus. Animals were slaugh-
tered in a local abattoir on either day 9, 11, 12, 15, or 18 of
the estrous cycle (N ¼46 per group) or pregnancy (N ¼
46 animals with conrmed pregnancy per group). Preg-
nancy was conrmed by the presence of conceptuses. The
conceptuses were ushed from each uterine horn with
sterile phosphate-buffered saline (137 mmol/L NaCl, 27
mmol/L KCl, 10 mmol/L Na
2
HPO
4
, and 2 mmol/L KH
2
PO
4
,
pH 7.4).
Endometrial fragments were collected from uteri at the
middle part of the uterine horn and were dissected from
myometrium. In pregnant gilts on days 14 and 18, the
endometrium collected from implantation sites was also
separated from trophoblasts.
Endometrial samples weresnap-frozen in liquid nitrogen
and stored at 80
C for further analyses. All procedures
involving animals were approved by the Local Research
Ethics Committee and were conducted in accordance with
the national guidelines for agricultural animal care.
2.2. Incubation of conceptusesdobtaining conceptus-exposed
medium
Day 11 or 15 conceptuseses were recovered from uteri of
pregnant gilts (N ¼5 and N ¼4, respectively) by gentle
ushing of each uterine horn with phenol red-free Medium
199 (M3769; Sigma-Aldrich, St. Louis, MO, USA) containing
0.1% (wt/vol) bovine serum albumin (BSA; ICN Biomedicals,
Inc., Costa Mesa, CA, USA), penicillin (100 IU/mL), and
streptomycin (100
m
g/mL; Sigma-Aldrich), warmed to 37
C. Then, conceptuses were washed two times with fresh
Medium 199 with 0.1% BSA and antibiotics, weighed, and
placed separately in culture asks containing phenol red-
free Medium 199 (1 mL of medium per 1530 mg of
conceptus), supplemented with penicillin, streptomycin,
and 10% (vol/vol) charcoal-stripped newborn calf serum
(steroids-free NCS; Sigma-Aldrich). Conceptuses were
incubated with culture medium for 24 hours at 37
Cin
a humidied atmosphere containing 95% air and 5% CO
2
.
After incubation, media from all conceptuses obtained from
each gilt were pooled together and centrifuged at 200 g
for 5 minutes to separate fragments of conceptus tissue.
Medium was stored at 80
C, and used in in vitro experi-
ments as day 11 or 15 conceptus-exposed medium (11d
CEM or 15d CEM).
2.3. Isolation of luminal epithelial and stromal cells of
endometrium
Luminal epithelial and stromal cells of the porcine
endometrium were isolated as previously described with
P. Kaczynski, A. Waclawik / Theriogenology 79 (2013) 784790 785
some modications [22]. Briey, uterine horns collected
from gilts (N ¼5) after slaughter on day 11 of the estrous
cycle were washed four times with sterile PBS. Endometrial
tissue was separated from the myometrium by scissors.
Separated fragments of endometrium were then digested
with 0.48% (wt/vol) dispase (Life Technologies) in Hanks
balanced salt solution (Ca-, Mg-, and phenol-free, pH 7.4;
Sigma-Aldrich) at 37
C for 50 minutes with gentle shaking.
LE cells released after this digestion were pelleted by
centrifugation at 200 gfor 10 minues at 8
C. LE cells
were then washed with Medium 199 (Sigma-Aldrich)
supplemented with 1% (wt/vol) BSA, 100 IU/mL penicillin,
and 100
m
g/mL streptomycin. Red blood cells were lysed by
Red Blood Cell Lysing Buffer (Sigma-Aldrich). Isolated LE
cells were then washed three times with fresh Medium 199
with 1% (wt/vol) BSA. After washing, LE cells were resus-
pended in culture medium (Medium 199 supplemented
with 1% [wt/vol] BSA, 10% (vol/vol) steroids-free NCS, 100
IU/mL penicillin, and 100
m
g/mL streptomycin) and coun-
ted. The cell viability was assessed by 0.5% (wt/vol) Trypan
blue dye exclusion and was greater than 90%.
Endometrial tissue remaining after digestion in dispase
was minced with a lancet and digested in 0.06% (wt/vol)
collagenase (Sigma-Aldrich) in Medium 199 supplemented
with 1% (wt/vol) BSA, for 1.5 hours at 37
C. ST cells were
pelleted by centrifugation at 200 gfor 10 minutes at 8
C.
Red blood cells were removed with the same procedure
described for LE cells. ST cells were then washed three
times with fresh Medium 199 supplemented with 1% (wt/
vol) BSA. Isolated cells were resuspended with culture
medium and counted. Cell viability was assessed by the
method described for LE cells and was greater than 90%.
Isolated ST cells were washed 24 hours after plating to
remove contaminating epithelial cells.
2.4. Incubation of luminal epithelial and stromal cells with
CEM in a coculture model
To study the effect of conceptus products on PTGFR
expression, isolated endometrial cells, obtained from gilts
(N ¼5) on day 11 of the estrous cycle, were plated in
a coculture model, in which ST cells were cultured at the
bottom of six-well culture plates and LE cells were cultured
on collagen-coated inserts placed inside each well (Biocoat
Cell Culture Inserts Collagen Type I; BD Biosciences, Bed-
ford, MA USA; apical compartment). We used the coculture
model, established previously by our group [23] because it
corresponds more adequately to in vivo conditions in which
interactions of LE and ST cells are enabled than culturing
these cells separately. ST and LE cells were cultured for
72 hours in a phenol-red free Medium 199 containing 10%
(vol/vol) steroids-free NCS, 1% (wt/vol) BSA, antibiotics
(100 IU/mL penicillin and 100
m
g/mL streptomycin), addi-
tionally supplemented with E
2
(10 nmol/L) and proges-
terone (100 nmol/L). After reaching full conuence, LE cells
were pretreated for 1 hour with phenol red-free Medium
199 containing 10% (vol/vol) steroids-free NCS and antibi-
otics with or without 1
m
M estrogen receptor antagonist
(ICI-182,78; Sigma-Aldrich). After pretreatment, LE cells
were treated with: conditioned control medium without
any contact with conceptus (phenol red-free Medium 199
supplemented with 10% steroid-free NCS, and antibiotics)
or CEM mixed 3:1 with the fresh control medium in
presence or absence of ICI-182,78. ICI-182,780 was only
used in incubation with 11d CEM but not with 15d CEM. ST
cells plated in the basal compartment were treated with
the control medium only. Moreover, to study a direct effect
of conceptus products on ST cell expression of PTGFR gene,
ST cells were cultured in six-well plates (Nunc, Rochester,
NY, USA), and stimulated with 11d CEM or 15d CEM in
the same way as LE cells in apical compartments in the
coculture model. Both types of treatments were per-
formed for 24 hours, at 37
C in a humidied atmosphere
containing 95% air and 5% CO
2
. After incubation, cells were
lysed with Fenozol buffer (A&A Biotechnology, Gdansk,
Poland), harvested, and stored at 80
C until total RNA
isolation.
2.5. Total RNA isolation
Total RNA was isolated using the Total RNA Prep Plus kit
(A&A Biotechnology) and treated with DNase I (Life Tech-
nology Inc., Carlsbad, CA, USA) according to the manufac-
turers protocol to exclude possible DNA contamination.
After isolation, the amount of RNA and its quality were
analyzed with an Agilent 2100 Bioanalyzer (Agilent Tech-
nologies, Waldbronn, Germany) and Nanodrop 1000
(Thermo Fisher Scientic Inc.). Isolated RNA was stored at
80
C for further analyses.
2.6. Reverse transcription
Isolated RNA was used to generate cDNA for real-time
polymerase chain reaction (PCR). Total RNA sample (1
m
g)
was reverse transcribed with MultiScribe Reverse Tran-
scriptase kit (Life Technologies) according to the manu-
facturers protocol. cDNA samples were stored at 80
C for
further real-time PCR analyses.
2.7. Real-time PCR
Real-time PCR was performed with the Applied Bio-
systems 7900 Real-Time PCR system (Life Technlogies)
using Power SYBR Green master mix (Life Technlogies).
Reverse transcribed cDNA was amplicated in the 25
m
Lof
reaction mixture (12.5
m
L Power SYBR Green master mix,
2.5
m
L of each sense and antisense primer (1
m
mol/L), 3.5
m
L
of cDNA, and 4
m
L of water). Specic primers used for real-
time PCR are listed in Table 1. For quantication, standard
Table 1
Primers used for real-time polymerase chain reaction.
Gene Primer sequence (5
0
3
0
) Reference
PTGFR Sense: TCAGCAGCACAGACAAGG [24]
Antisense: TTCACAGGCATCCAGATAATC
b
-actin Sense: ACATCAAGGAGAAGCTCTGCTACG [14]
Antisense: GAGGGGCGATGATCTTGATCTTCA
GAPDH Sense: CAGCAATGCCTCCTGTACCA [25]
Antisense: GATGCCGAAGTTGTCATGGA
Cyclophylin Sense: TAACCCCACCGTCTTCTT [26]
Antisense: TGCCATCCAACCACTCAG
P. Kaczynski, A. Waclawik / Theriogenology 79 (2013) 784790786
curves based on serial dilutions of the cDNA were included.
Before amplication samples were initially denaturated at
95
C for 15 minutes. The PCR program for PTGFR gene was
performed as follows: 36 cycles of denaturation ( 95
C for
15 seconds), and annealing and elongation (60
C for 1
minute). For
b
-actin amplication the PCR program was:
36 cycles of denaturation (95
C for 15 seconds), annealing
(55
C for 30 seconds), and elongation (72
C for 1 minute).
After PCR, melting curves were acquired by gradual
increases in the temperature from 60
Cto95
C to ensure
that a single product was amplied in the PCR reaction.
Obtained products were then sent for sequencing.
Sequences were checked by nBLAST to conrm the
sequence homology with the sequence present in Gene-
Bank. Stability of the reference gene was assessed using the
statistical algorythms Normnder 2.0 [27]. Three reference
genes were analyzed: GAPDH,cyclophylin, and
b
-actin. The
most stably expressed gene was
b
-actin.
2.8. Western blot
Endometrial tissues were homogenized on ice in buffer
containing 50 mmol/L TRIS-HCl, pH 8.0; 150 mmol/L NaCl,
and 1 mmol/L EDTA supplemented with protease inhibitor
cocktail (Sigma-Aldrich). Homogenates were then centri-
fuged for 15 minutes at 800 gat 8
C and stored at 80
C
for further analysis. The protein concentration was deter-
mined by Bradford Assay [28].
Protein samples from endometrial tissue homogenates
(40
m
g) were dissolved in SDS gel-loading buffer (50 mmol/L
TRIS-HCl, pH 6.8; 4% SDS, 20% glycerol, and 2%
b
-mercap-
toethanol), heated at 95
C for 4 minutes, and separated on
10% SDS-PAGE. Separated proteins were electroblotted
onto 0.2 mm nitrocellulose membrane in transfer buffer
(20 mmol/L TRIS-HCl buffer, pH 8.2; 150 mmol/L glycine,
and 20% methanol). After blocking in 5% nonfat dry milk in
TRIS-buffered saline buffer (TBS-T, containing 0.1% Tween-
20) for 1.5 hours at 25.6
C, the membranes were incu-
bated overnight with polyclonal anti-PTGFR antibodies at
4
C (Santa Cruz Biotechnology, Inc., Santa Cruz, CA, USA;
sc-67029). After incubation membranes were washed three
times in TBS-T and incubated with anti-rabbit secondary
antibodies (A8025; Sigma-Aldrich) dissolved 1:20,000 in
TBS-T, in room temperature for 90 minutes. After incuba-
tion membranes were also washed three times in TBS-T.
Immune complexes were visualized using alkaline phos-
phatase visualization procedure. Western blots were
quantied using Kodak 1D software (Eastman Kodak).
Sample loading was standardized to expression of GAPDH
using specic antibodies (1:100; A4312; Sigma-Aldrich).
2.9. Statistical analysis
All statistical analyses were conducted using GraphPad
Prism v. 5.02 software (GraphPad Software, Inc., San Diego,
CA, USA). To study the endometrial expression of PTGFR
mRNA and protein on days 9,11,12, 15, and 18 of the estrous
cycle and pregnancy, two-way ANOVA followed by Bon-
ferroni post hoc test was performed. This analysis included
the effect of day, reproductive status (the estrous cycle or
pregnancy), and day by reproductive status interaction. To
test the effect of 11d CEM and 15d CEM on PTGFR gene
abundance in endometrial cells, one-way ANOVA followed
by Bonferroni post hoc test was conducted. To study the
effect of CEM in the presence or absence of ICI-182,780 on
PTGFR mRNA abundance in LE cells, statistical analysis was
performed using two-way ANOVA followed by Bonferroni
post hoc test. This analysis included the effect of ICI pres-
ence, CEM treatment, and ICI presence by CEM treatment
interaction.
3. Results
3.1. Expression of PTGFR mRNA in the porcine endometrium
during early pregnancy and the estrous cycle
Real-time reverse transcription PCR analysis did not
reveal signicant differences in PTGFR gene expression in
the porcine endometrium between pregnant and cyclic
gilts. However, effect of day on PTGFR gene expression was
observed (P <0.05). On day 18 of pregnancy and the
estrous cycle PTGFR mRNA levels were increased when
compared with previous (915) days (Fig. 1A). PTGFR mRNA
Fig. 1. Relative expression of PGF
2
a
receptor (PTGFR) mRNA (A) and protein
(B) in the porcine endometrium during early pregnancy and the estrous
cycle. The representative samples of Western blots are shown in the upper
panels. Values from Western blot analyses were normalized to GAPDH. Data
are represented as the mean SEM. Bars with different letters differ
signicantly (P <0.05) within the estrous cycle (a and b) and the pregnancy
(x and y). Asterisks indicate differences (P <0.05) between animals during
the estrous cycle and animals during pregnancy.
P. Kaczynski, A. Waclawik / Theriogenology 79 (2013) 784790 787
was expressed in luminal epithelial and stromal cells of the
endometrium (Fig. 2, upper panel).
3.2. Expression of PTGFR protein in the porcine endometrium
during early pregnancy and the estrous cycle
The effects of reproductive status and day on PTGFR
protein expression were detected in the endometrium
(P <0.05). During the estrous cycle expression of PTGFR
was low on day 9, intermediate on day 12, and elevated on
day 18 (vs. days 9, 11, and 15; P <0.05). During early
pregnancy, content of PTGFR protein was greater starting at
day 12, and until day 18 (P <0.05; Fig. 1B). The endometrial
expression of PTGFR protein was upregulated on day 15 of
pregnancy when compared with that of nonpregnant
animals on day 15 (Fig. 1B).
3.3. Effect of conceptus (CEM) on expression of PTGFR mRNA
in luminal epithelial and stromal cells of endometrium
cultured in vitro
We further studied if the embryo regulates PTGFR
expression in the different types of endometrial cellsdLE
and ST. Incubation with CEM from day 11 (P <0.01), and
from day 15 of pregnancy (P <0.05) increased expression
of PTGFR mRNA in LE cells cultured in inserts of the
coculture model with stromal cells (Fig. 2A). Moreover,
expression of PTGFR mRNA was higher in LE incubated with
11d CEM than in LE incubated with 15d CEM (P <0.01).
There was no effect of treatment with 11d CEM or 15d CEM
on the expression of PTGFR mRNA in ST cells in the cocul-
ture model with LE cells (data not shown), and in ST cells
cultured separately in six-well plates (Fig. 2B).
3.4. Effect of products secreted by conceptus on PTGFR mRNA
expression in luminal epithelial cells is mediated through
estrogen receptor
Since secretion of E
2
and expression of estrogen
receptor are elevated on day 11 of pregnancy, we studied
further the effect of 11d CEM with the presence of estrogen
receptor antagonist (ICI-182,780) on PTGFR expression. The
11d CEM-induced increase of PTGFR mRNA was abolished
by incubation LE cells in the presence of ICI-182,780
(P <0.01; Fig. 3). There were no differences in PTGFR
mRNA abundance in LE cells treated with control medium
with or without the estrogen receptor antagonist (Fig. 3).
4. Discussion
During establishment of pregnancy autocrine and
paracrine interactions between the conceptus and endo-
metrium are required for implantation and the establish-
ment of pregnancy [20,29]. Appropriate balance between
PG synthesis and secretion is important either during
the luteolysis initiation or the establishment of pregnancy
[29]. Despite the differences in mechanisms of placentation
in diverse species of animals, the inhibition of PG synthesis
before the implantation led to the failure of pregnancy
development [3032].
Fig. 2. Effect of medium exposed to conceptuses from day 11 and 15 of pregnancy (11d CEM and 15d CEM, respectively) on expression of PTGFR mRNA in luminal
epithelial cells (A) in the coculture model with stromal cells, and in stromal cells cultured separately (B). Data are represented as the mean SEM. Asterisks
indicate differences between cells treated with 11d CEM and 15d CEM in comparison with the cells treated with control medium (* P <0.05; ** P <0.01).
Expression of PTGFR mRNA was detected by reverse transcription PCR in luminal epithelial (LE) and stromal (ST) cells as indicated in the upper panel. C, control
medium.
P. Kaczynski, A. Waclawik / Theriogenology 79 (2013) 784790788
In this study, we demonstrated elevated PTGFR protein
expression in the porcine endometrium during initializa-
tion of implantation and during the implantation period
when compared with the preimplanation period (Fig. 1).
These results correspond with the ndings which revealed
elevation of PGF
2
a
synthase protein expression in porcine
endometrium and trophoblasts on days 14 to 25 of preg-
nancy [14,15]. Moreover, the upregulation of PTGFR on
day 15 and 18 of pregnancy correspond to the increased
concentrations of PGF
2
a
in uterine ushings from preg-
nant gilts on day 14 to 18 of pregnancy than those from
cyclic animals [21]. Moreover, increased expression of
PTGFR on day 15 of pregnancy coincides with greater
expression of vascular endothelial growth factor A at the
peri-implantation period and elevated expression of its
receptor fms-related tyrosine kinase 1 on day 15 of preg-
nancy in the porcine endometrium [33]. Interestingly,
activation of PTGFR by PGF
2
a
mediates expression of
angiogenic factors in other cell typesdhuman endometrial
adenocarcinomas [7]. Therefore, it can be speculated that
the increase of PTGFR in the porcine endometrium during
implantation could be involved in angiogenesis. However, it
requires further studies.
Presence of PTGFR in the porcine endometrium is in
agreement with detection of this receptor in uterus of other
species [30,3436]. Immunohistochemical analysis of
PTGFR in the ovine endometrium revealed strong signals in
the endometrium on days 14 and 20 of pregnancy [30].
Interestingly, abundance of PTGFR mRNA was greater in the
endometrium which had a direct contact with conceptus
(in placentome caruncles) when compared with intercar-
uncles in the bovine uterus through the whole pregnancy
period [35]. Elevated expression of PTGFR mRNA was
observed in pregnant when compared with nonpregnant
rat uterus [36]. In contrast to our results, in the mare
endometrium expression of PTGFR was decreased on days
14 to 15 of pregnancy when compared with days 14 to 15 of
the estrous cycle [34].
Based on the present results indicating upregulation of
PTGFR protein during implantation period in the pig, we
further studied if the embryo regulates PTGFR expression in
the different types of endometrial cellsdLE and ST. Our
results demonstrated that the medium obtained during
incubation with conceptuses from day 11 or 15 of preg-
nancy stimulated expression of PTGFR mRNA in LE but not
in ST cells of the porcine endometrium. This nding is
interesting considering that pigs have a true epi-
theliochorial placenta in which luminal epithelium remains
intact and the conceptus has a direct contact with LE but
not with ST cells in vivo.
We hypothesized that if PGF
2
a
and PTGFR indeed exerts
a pivotal role during early pregnancy, then the porcine
conceptus, especially acting through the primary porcine
embryonic signal E
2
, might regulate expression of PTGFR.
Since secretion of estradiol and estrogen receptor expres-
sion in LE are elevated on day 11 of pregnancy [10,37],we
further studied the effect of CEM obtained during incuba-
tion with day 11 conceptuses on endometrial PTGFR
expression in presence of estrogen receptor antagonist ICI-
182,780. Our study indicated that the porcine conceptus
mediates its effect by E
2
because blocking its receptor
abolished the effect of CEM on luminal epithelial cells.
Moreover, our results are in agreement with earlier studies
showing that E
2
administrated for ovariectomized rats
enhances the PTGFR mRNA expression in rat uterus [36].
4.1. Conclusions
Our ndings indicate an important role of PTGFR in the
porcine endometrium during the implantation period.
Upregulation of PTGFR gene expression in luminal epithe-
lial cells mediated through estrogen receptor suggests
regulation of PTGFR by embryonic signaldE
2
. Moreover,
our results suggest a paracrine and autocrine effect of PGF
2
a
through its receptor PTGFR on the porcine endometrium,
especially on luminal epithelium which is in a direct
contact with conceptus during the implantation period.
The presented results support the hypothesis of a local
inuence of PGF
2
a
on the endometrium to promote uterine
function during this period and verify the established view
on the role of PGF
2
a
during early pregnancy as an undesired
factor. Thus, this potential role of PGF
2
a
and its receptor in
pregnancy establishment requires further studies.
Acknowledgments
The authors thank J. Klos and M. Blitek for help in care
and handling of animals. This research was supported by
the Ministry for Science and Higher Education in Poland
(grant 717/N-COST/2010/0) and National Science Centre in
Poland (grant 2012/05/E/NZ9/03493). A. Waclawik was
awarded by Stipendium for Outstanding Young Researchers
from the Ministry of Science and Higher Education in
Poland.
References
[1] McCracken JA, Custer EE, Lamsa JC. Luteolysis: a neuroendocrine-
mediated event. Pysiol Rev 1999;79:263323.
[2] Sakamoto K, Ezashi T, Miwa K, Okuda-Ashitaka E, Houtani T,
Sugimoto T, et al. Molecular cloning and expression of a cDNA of
the bovine prostaglandin F2a receptor. J Biol Chem 1994;269:
38818.
Fig. 3. Effect of the day 11 conceptus-exposed medium (CEM) in presence or
absence of 1
m
mol/L estrogen receptor antagonist ICI-182,780 (ICI) on relative
expression of PTGFR mRNA in porcine luminal epithelial (LE) cells of endo-
metrium cultured in a coculture model of LE and stromal cells. Data are
represented as the mean SEM. Asterisks indicate differences between cells
treated with CEM containing ICI-182,780 in comparison with cells treated
with control medium without ICI-182,780 (** P <0.01). C, control medium.
P. Kaczynski, A. Waclawik / Theriogenology 79 (2013) 784790 789
[3] Sugimoto Y, Hasumoto K, Namba T, Irie A, Katsuyama M, Negishi M,
et al. Cloning and expression of a cDNA for mouse prostaglandin F2
a
receptor. J Biol Chem 1994;269:135660.
[4] Tsai SJ, Anderson LE, Juengel J, Niswender GD, Wiltbank MC.
Regulation of prostaglandin F2 alpha and E receptor mRNA by
prostaglandin F 2 alpha in ovine corpora lutea. J Reprod Fertil 1998;
114:6975.
[5] Bos CL, Richel DJ, Ritsema T, Peppelenbosch MP, Versteeg HH.
Prostanoids and prostanoid receptors in signal transduction. Int J
Biochem Cell Biol 2004;36:1187205.
[6] Pierce KL, Fujino H, Srinivasan D, Regan JW. Activation of FP pros-
tanoid receptor isoforms leads to Rho-mediated changes in cell
morphology and in the cell cytoskeleton. J Biol Chem 1999;274:
359449.
[7] Sales KJ, List T, Boddy SC, Williams AR, Anderson RA, Naor Z, et al. A
novel angiogenic role for prostaglandin F2alpha-FP receptor inter-
action in human endometrial adenocarcinomas. Cancer Res 2005;
65:770716.
[8] Narumiya S, Sugimoto Y, Ushikubi F. Prostanoid receptors: struc-
tures, properties, and functions. Physiol Rev 1999;79:1193226.
[9] Boonyaprakob U, Gadsby JE, Hedgpeth V, Routh P, Almond GW.
Cloning of pig prostaglandin F2alphaFP receptor cDNA and expres-
sion of its mRNA in the corpora lutea. Reproduction 2003;125:5364.
[10] Geisert RD, Zavy MT, Moffatt RJ, Blair RM, Yellin T. Embryonic
steroids and the establishment of pregnancy in pigs. J Reprod Fertil
Suppl 1990;40:293305.
[11] Moeljono MP, Thatcher WW, Bazer FW, Frank M, Owens LJ,
Wilcox CJ. A study of prostaglandin F2a as the luteolysin in swine: II.
Characterization and comparison of prostaglandin F, estrogens and
progestin concentrations in utero-ovarian vein plasma of
nonpregnant and pregnant gilts. Prostaglandins 1977;14:54355.
[12] Bazer FW, Thatcher WW. Theory of maternal recognition of preg-
nancy in swine based on estrogen controlled endocrine versus
exocrine secretion of prostaglandin F2a by the uterine endome-
trium. Prostaglandins 1977;14:397400.
[13] Krzymowski T, Stefanczyk-Krzymowska S. The oestrous cycle and
early pregnancya new concept of local endocrine regulation. Vet J
2004;168:28596.
[14] Waclawik A, Rivero-Muller A, Blitek A, Kaczmarek MM, Brokken LJ,
WatanabeK, et al. Molecular cloning and spatiotemporal expressionof
prostaglandin F synthase and microsomal prostaglandinE synthase-1
in porcine endometrium. Endocrinology 2006;147:21021.
[15] Waclawik A, Ziecik AJ. Differential expression of prostaglandin (PG)
synthesis enzymes in conceptus during peri-implantation period
and endometrial expression of carbonyl reductase/PG 9-
ketoreductase in the pig. J Endocrinol 2007;194:499510.
[16] Waclawik A, Jabbour HN, Blitek A, Ziecik AJ. Estradiol-17beta,
prostaglandin E2 (PGE2), and the PGE2 receptor are involved in
PGE2 positive feedback loop in the porcine endometrium. Endo-
crinology 2009;150:382332.
[17] Akinlosotu BA, Diehl JR, Gimenez T. Sparing effects of intrauterine
treatment with prostaglandin E2 on luteal function in cycling gilts.
Prostaglandins 1986;32:2919.
[18] Christenson LK, Farley DB, Anderson LH, Ford SP. Luteal mainte-
nance during early pregnancy in the pig: role for prostaglandin E2.
Prostaglandins 1994;47:6175.
[19] Waclawik A, Blitek A, Ziecik AJ. Oxytocin and tumor necrosis factor
alpha stimulateexpression of prostaglandin E2 synthase and secretion
of prostaglandin E2 by luminal epithelial cells of the porcine endo-
metrium during early pregnancy. Reproduction 2010;140:61322.
[20] Waclawik A. Novel insights into the mechanisms of pregnancy
establishment: regulation of prostaglandin synthesis and signaling.
Reproduction 2011;142:38999.
[21] Zavy MT, Bazer FW, Thatcher WW, Wilcox CJ. A study of prosta-
glandin F2a as the luteolysin in swine: V. Comparison of prosta-
glandin F, progestins, estrone and estradiol in uterine ushings from
pregnant and nonpregnant gilts. Prostaglandins 1980;20:83751.
[22] Blitek A, Ziecik AJ. Prostaglandins F2
a
and E2 secretion by porcine
epithelial and stromal endometrial cells on different days of the
oestrous cycle. Reprod Domest Anim 2004;39:3406.
[23] Blitek A, Morawska E, Kiewisz J, Ziecik AJ. Effect of conceptus
secretions on HOXA10 and PTGS2 gene expression, and PGE2
release in co-cultured luminal epithelial and stromal cells of the
porcine endometrium at the time of early implantation. Ther-
iogenology 2011;76:95466.
[24] Zannoni A, Bernardini C, Rada T, Ribeiro LA, Forni M, Bacci ML.
Prostaglandin F2-alpha receptor (FPr) expression on porcine corpus
luteum microvascular endothelial cells (pCL-MVECs). Reprod Biol
Endocrinol 2007;5:31.
[25] Vallée M, Beaudry D, Roberge C, Matte JJ, Blouin R, Palin MF.
Isolation of differentially expressed genes in conceptuses and
endometrial tissue of sows in early gestation. Biol Reprod 2003;69:
1697706.
[26] Dozois CM, Oswald E, Gautier N, Serthelon JP, Fairbrother JM,
Oswald IP. A reverse transcription-polymerase chain reaction
method to analyze porcine cytokine gene expression. Vet Immunol
Immunopathol 1997;58:287300.
[27] Andersen CL, Jensen JL, Ørntoft TF. Normalization of real-time
quantitative reverse transcription-PCR data: a model-based vari-
ance estimation approach to identify genes suited for normalization,
applied to bladder and colon cancer data sets. Cancer Res 2004;64:
524550.
[28] Bradford MM. A rapid and sensitive method for the quantitation of
microgram quantities of protein utilizing the principle of protein-
dye binding. Anal Biochem 1976;72:24854.
[29] Waclawik A, Blitek A, Kaczmarek MM, Kiewisz J, Ziecik AJ. Anti-
luteolytic mechanisms and the establishment of pregnancy in the
pig. Soc Reprod Fertil Suppl 2009;66:30720.
[30] Dorniak P, Bazer FW, Spencer TE. Prostaglandins regulate conceptus
elongation and mediate effects of interferon tau on the ovine
uterine endometrium. Biol Reprod 2011;84:111927.
[31] Erdem H, Guzeloglu A. Effect of meloxicam treatment during early
pregnancy in Holstein heifers. Reprod Domest Anim 2010;45:
6258.
[32] Kraeling RR, Rampacek GB, Fiorello NA. Inhibition of pregnancy with
indomethacin in mature gilts and prepuberal gilts induced to
ovulate. Biol Reprod 1985;32:10510.
[33] Kaczmarek MM, Waclawik A, Blitek A, Kowalczyk AE, Schams D,
Ziecik AJ. Expression of the vascular endothelial growth factor
receptor system in the porcine endometrium throughout the oes-
trous cycle and early pregnancy. Mol Reprod Dev 2008;75:36272.
[34] Atli MO, Kurar E, Kayis SA, Aslan S, Semacan A, Celik S, et al. Eval-
uation of genes involved in prostaglandin action in equine endo-
metrium during estrous cycle and early pregnancy. Anim Reprod Sci
2010;122:12432.
[35] Fortier MA, Arosh JA, Banu SA, Chapdelaine P. Temporal and tissue-
specic expression of prostaglandin receptors EP2, EP3, EP4, FP, and
cyclooxygenases 1 and 2 in uterus and fetal membranes during
bovine pregnancy. Endocrinology 2004;145:40717.
[36] Yallampalli C, Dong YL. Pregnancy and exogenous steroid treat-
ments modulate the expression of relaxant EP(2) and contractile FP
receptors in the rat uterus. Biol Reprod 2000;62:5339.
[37] Geisert RD, Brenner RM, Moffatt J, Harney JP, Yellin T, Bazer FW.
Changes in oestrogen receptor protein, mRNA expression and
localization in the endometrium of cyclic and pregnant gilts. Reprod
Fertil Dev 1993;5:24760.
P. Kaczynski, A. Waclawik / Theriogenology 79 (2013) 784790790
... Acting on the endometrium, E2 induces mechanisms altering the secretion of prostaglandin F2α (PGF2α) in an endocrine (into utero-ovarian venous drainage) to exocrine (into uterine lumen) manner, which protects the corpora lutea from the luteolytic action of PGF2α [2]. Moreover, E2 regulates prostaglandin (PGE2 and PGF2α) synthesis and signaling in the endometrium [10][11][12]. It favors endometrial synthesis of PGE2, which is transported to the ovaries to exert a luteoprotective effect [10,13,14]. ...
... A stimulating effect of PGF2α on luteal angiogenesis has been described in bovines [36,37]. Our previous studies revealed that E2 regulates the expression of the endometrial PTGFR gene and protein in the pig, and indicated the involvement of PGF2α-PTGFR signaling in angiogenic changes within the endometrium [11,12,38]. Likewise, in the present study, we found a stimulating effect of an intrauterine infusion of E2 on luteal PTGFR expression in vivo. ...
... Total RNA was isolated from luteal tissue fragments (approx. 50 mg) collected from gilts after in vivo experiment and from gilts on day 12 of the estrous cycle and pregnancy and reverse-transcribed as described previously [11]. Concentration and integrity of isolated RNA was determined by Agilent 2100 Bioanalyzer (Agilent Technologies, Santa Clara, CA, USA) and NanoDrop spectrophotometer (Thermo Fisher Scientific; Waltham, MA, USA). ...
Article
Full-text available
Citation: Kaczynski, P.; Baryla, M.; Goryszewska, E.; Waclawik, A. Estradiol-17β Regulates Expression of Luteal DNA Methyltransferases and Genes Involved in the Porcine Corpus Luteum Function In Vivo. Int. J. Mol. Sci. 2021, 22, 3655. https:// Abstract: The corpus luteum (CL) is a temporary endocrine gland vital for pregnancy establishment and maintenance. Estradiol-17β (E2) is the major embryonic signal in pigs supporting the CL's function. The mechanisms of the luteoprotective action of E2 are still unclear. The present study aimed to determine the effect of E2 on luteal expression of factors involved in CL function. An in vivo model of intrauterine E2 infusions was applied. Gilts on day 12 of pregnancy and the estrous cycle were used as referential groups. Concentrations of E2 and progesterone were elevated in CLs of gilts receiving E2 infusions, compared to placebo-treated gilts. Estradiol-17β stimulated luteal expression of DNA-methyltransferase 1 (DNMT1), but decreased expression of DNMT3B gene and protein, as well as DNMT3A protein. Similar results for DNMT3A and 3B were observed in CLs on day 12 of pregnancy compared to day 12 of the estrous cycle. Intrauterine infusions of E2 altered luteal expression of the genes involved in CL function: PTGFR, PTGES, STAR, HSD17B1, CYP19A1, and PGRMC1. Our findings indicate a role for E2 in expression regulation of factors related to CL function and a novel potential for E2 to regulate DNA methylation as putative physiological mechanisms controlling luteal gene expression.
... The observed changes in the expression of the endometrial chemerin system under the influence of prostaglandins could be explained by variations in the expression of prostaglandin receptors during the analyzed pregnancy periods. The concentrations of PGE2 and PGF2α receptors fluctuate during pregnancy in mice and during the estrous cycle and early gestation in pigs [31][32][33]. In pigs, differences were observed in the endometrial content of PGF2α receptor (PTGFR) protein during the estrous cycle and gestation [32]. ...
... The concentrations of PGE2 and PGF2α receptors fluctuate during pregnancy in mice and during the estrous cycle and early gestation in pigs [31][32][33]. In pigs, differences were observed in the endometrial content of PGF2α receptor (PTGFR) protein during the estrous cycle and gestation [32]. Estradiol and PGE2, the main factors responsible for the maternal recognition of pregnancy, stimulate the endometrial expression of PGE2 receptors PTGER2 and PTGER4 [33]. ...
... Furthermore, due to a fact that PGs are secreted in a pulsatile manner, and their secretion patterns during early pregnancy are similar, the PGE2:PGF2α ratio is another agent, which should be taken into consideration [38]. Beside its luteoprotective effects, the redirection of prostaglandin secretion also promotes endometrial reconstruction and modulation of the angiogenesis process during the peri-implantation period [32,39,40]. The chemerin system seems to be involved in the stimulation of angiogenesis. ...
Article
Full-text available
Chemerin belongs to the group of adipocyte-derived hormones known as adipokines, which are responsible mainly for the control of energy homeostasis. Adipokine exerts its influence through three receptors: Chemokine-like receptor 1 (CMKLR1), G protein-coupled receptor 1 (GPR1), and C-C motif chemokine receptor-like 2 (CCRL2). A growing body of evidence indicates that chemerin participates in the regulation of the female reproductive system. According to the literature, the expression of chemerin and its receptors in reproductive structures depends on the local hormonal milieu. The aim of this study was to investigate the in vitro effect of prostaglandins E2 (PGE2) and F2α (PGF2α) on chemerin and chemerin receptor (chemerin system) mRNAs (qPCR) and proteins (ELISA, Western blotting) in endometrial tissue explants collected from early-pregnant gilts. Both PGE2 and PGF2α significantly influenced the expression of the chemerin gene, hormone secretion, and the expression of chemerin receptor genes and proteins. The influence of both prostaglandins on the expression of the chemerin system varied between different stages of gestation. This is the first study to describe the modulatory effect of PGE2 and PGF2α on the expression of the chemerin system in the porcine uterus during early gestation.
... RNA isolation, reverse transcription, and analysis of gene expression of all samples were performed as described previously [22,28]. The mRNA content of PROKR1, PGF2α synthase (AKR1C4; also known as AKR1CL1 or PGFS), microsomal prostaglandin E synthase-1 (PTGES; mPGES-1), PTGS2, prostaglandin-E2 9reductase (CBR1), and VEGFA was analyzed using Power SYBR Green (Life Technologies, Carlsbad, CA) with oligonucleotide primers (1 μM; Supplementary Table S1). ...
... The protein levels of PROK1, PROKR1, AKR1C4, PTGES, MAPK1/3, and p-(phospho-) MAPK1/3 were determined by Western blot analyses as described previously [28,31], with some modifications. Briefly, endometrial tissue samples were homogenized in homogenization buffer (50 mM/L Tris-HCl, pH 8.0; 150 mM/L NaCl; and 1 mM/L EDTA) supplemented with protease inhibitor cocktail (Sigma-Aldrich) and phosphatase inhibitor cocktail (Sigma-Aldrich; only in samples destined for protein phosphorylation studies). ...
... Our results are consistent with previous findings, indicating that the expression of PGF2α synthase (AKR1C4) [72] is elevated in the porcine endometrium during pregnancy and/or implantation [73,74]. Furthermore, our group recently reported that PGF2α participates in pregnancy establishment in the pig by promoting angiogenesis and regulating the expression of genes involved in endometrial remodeling and embryo-maternal communication [22,25,28,75]. ...
Article
Pregnancy establishment in mammals, including pigs, requires proper communication between embryos and the maternal reproductive tract. Prokineticin 1 (PROK1) has been described as a secretory protein with pleiotropic functions and as a novel tissue-specific angiogenic factor. However, despite the studies performed mainly on human cell lines and in mice, the function of PROK1 in the endometrium during early pregnancy is still not fully elucidated. We hypothesized that PROK1 contributes to pregnancy establishment in the pig. The present study is the first to report that the expression of PROK1 and its receptor (PROKR1) is elevated in the porcine endometrium during implantation and the early placentation period. PROK1 protein was detected mainly in luminal epithelial cells, glandular epithelial cells and blood vessels in the endometrium. Using the porcine in vivo model of unilateral pregnancy, we revealed that conceptuses induced the endometrial expression of PROK1 and PROKR1. Moreover, the embryonic signal, estradiol-17β, as well as progesterone, stimulated the endometrial expression of PROK1 and PROKR1. We also evidenced that PROK1–PROKR1 signaling supports endometrial angiogenesis in the pig. The PROK1-stimulated proliferation of primary porcine endometrial endothelial (PEE) cells involved PI3K/AKT/mTOR, MAPK, cAMP and NFKB signaling pathways. Furthermore, PROK1 via PROKR1 promoted the formation of capillary-like structures by PEE cells. PROK1 also stimulated VEGFA and PGF2α secretion, which in turn, may indirectly support angiogenic changes within endometrial tissue. In summary, our study suggests that PROK1 acts as an embryonic signal mediator that regulates endometrial angiogenesis and secretory function during implantation and early placentation in pigs.
... The fold difference was calculated by dividing the absorbance obtained for PROK1treated cells by the absorbance obtained for control-treated cells.Real-time RT-PCR. Isolation of RNA, reverse transcription, and real-time PCR for samples from Experiment 1 and 2 were performed as described previously8,25,54,55 . The mRNA expression of PROKR1, VEGFA, FLT1, KDR, LIF, LIFR, WNT4, WNT5A and WNT7A were analyzed using Power SYBR Green (Life Technologies, Carlsbad, CA, USA) with oligonucleotide primers (1 μM; SupplementaryTable 1). ...
Article
Full-text available
Successful pregnancy establishment in mammals depends on proper embryo-maternal communication. Prokineticin 1 (PROK1) is a secretory protein that exerts pleiotropic functions in various tissues. Despite the studies that have primarily been performed with human cell lines and mice, the function of PROK1 in trophoblasts has still not been fully elucidated. Hence, the aim of this study was to establish the role of PROK1 in trophoblasts during implantation and placentation. Prokineticin 1 mRNA was elevated in porcine trophoblasts during implantation and the early placentation period. Furthermore, we reveal that PROK1–PROKR1 signaling induces the expression of genes involved in the regulation of angiogenesis, immunological response, trophoblast cell adhesion, invasion, and proliferation, as well as stimulating phosphorylation of MAPK and PTK2. Ingenuity Pathway Analysis identified the aforementioned and also other functions associated with PROK1-regulated genes/proteins, such as cell-to-cell contact, epithelial tissue differentiation, Ca2+ release, lipid synthesis, and chemotaxis. We also showed evidence that PROK1 acting via PROKR1 increased trophoblast cell proliferation and adhesion. The PROK1-stimulated cell proliferation was mediated by PI3K/AKT/mTOR, MAPK, and cAMP, whereas adhesion was mediated by MAPK and/or PI3K/AKT signaling pathways. Concluding, our study suggests that PROK1 plays a pleiotropic role in trophoblast function during implantation and early placentation.
... In the present study, we demonstrated that the synergistic action of PGE2 with E2 in vivo increased the gene expression of SLCO2A1, which serves as a prostaglandin transporter. Our recent reports indicated a vital role for prostaglandins (PGE2 and PGF2α) in the establishment and development of pregnancy [1,22,[41][42][43][44]. The stimulating effect of E2 acting simultaneously with PGE2 on the expression of SLCO2A1 is the more interesting result, especially when we take into consideration our description of the role of E2 and PGE2 in the mechanism of the PGE2-positive feedback loop in the porcine endometrium [16]. ...
Article
Successful pregnancy establishment in mammals depends on numerous interactions between embryos and the maternal organism. Estradiol-17β (E2) is the primary embryonic signal in the pig, and its importance has been questioned recently. However, E2 is not the only molecule of embryonic origin. In pigs, prostaglandin E2 (PGE2) is abundantly synthesized and secreted by conceptuses and endometrium. The present study aimed to determine the role of PGE2 and its simultaneous action with E2 in changes in porcine endometrial transcriptome during pregnancy establishment. The effects of PGE2 and PGE2 acting with E2 were studied using an in vivo model of intrauterine hormone infusions, and were compared to the effects of E2 alone and conceptuses’ presence on day 12 of pregnancy. The endometrial transcriptome was profiled using gene expression microarrays followed by statistical analyses. Downstream analyses were performed using bioinformatics tools. Differential expression of selected genes was verified by quantitative PCR. Microarray analysis revealed 2413 differentially expressed genes (DEGs) in the endometrium treated simultaneously with PGE2 and E2 (P < 0.01). No significant effect of PGE2 administered alone on endometrial transcriptome was detected. Gene ontology annotations enriched for DEGs were related to multiple processes such as: focal adhesion, vascularization, cell migration and proliferation, glucose metabolism, tissue remodeling, and activation of immune response. Simultaneous administration of E2 and PGE2 induced more changes within endometrial transcriptome characteristic to pregnancy than infusion of E2 alone. The present findings suggest that synergistic action of estradiol-17β and PGE2 resembles the effects of pregnancy on endometrial transcriptome better than E2 alone.
... RNA isolation, reverse transcription, and analysis of the gene expression of all samples were performed, as described previously [28,34]. The mRNA expression of PROKR1, PGR-B, PGR-AB, ESR1, ESR2, FLT1, KDR, LIF, LIFR, WNT4, WNT5A, and WNT7A were analyzed by real-time RT-PCR using Power SYBR Green (Life Technologies, Carlsbad, CA, USA) with oligonucleotide primers (1 μM; Table 1). ...
Article
Acquisition of endometrial receptivity for embryo implantation is one of the crucial processes during pregnancy and is induced mainly by progesterone and enhanced by conceptus signals. Prokineticin 1 (PROK1) is characterized as a secretory protein with diverse functions in various tissues, including the reproductive tract. PROK1, with its receptor PROKR1, are up-regulated in the porcine endometrium during implantation and in women's receptive endometrium and decidua. However, the function of PROK1 in embryo-maternal communication has still not been fully elucidated. Hence, we hypothesize that PROK1 is involved in endometrial receptivity development and implantation in pigs. In this study, using the porcine in vivo model of intrauterine infusions of estradiol-17β (E2) and prostaglandin E2 (PGE2), we revealed that these hormones elevated endometrial expression of PROK1 and PROKR1 mRNA, respectively. Moreover, E2, acting synergistically with PGE2, increased PROKR1 protein expression. We also evidenced that PROK1-PROKR1 signaling induced expression of following genes and/or proteins CCN2, CDH13, FGF2, NFATC2, ANGPT1, ANGPT2, CDH1, MUC4, SPP1, IFNG, IL6, LIF, LIFR, TNF, TGFB3, and FGF9, as well as phosphorylation of PTK2 and secretion of IL6 and IL11 by endometrial explants in vitro. Ingenuity Pathway Analysis revealed that functions associated with the PROK1-regulated genes/proteins include cell-to-cell contact, cell attachment, migration and viability, differentiation of epithelial tissue, leukocyte migration, inflammatory response, angiogenesis, and vasculogenesis. Summarizing, our study suggests that PROK1 acts pleiotropically as an embryonic signal mediator that regulates endometrial receptivity by increasing the expression of the genes and proteins involved in implantation and pregnancy establishment in pigs.
... All qPCRs were conducted at an annealing temperature of 60 • C and dissociation curves consisting of single peaks were generated. The relative abundance of mRNA transcripts for candidate genes was quantified: BCL-2associated X protein (BAX) (Zhao et al., 2014), B-cell lymphoma 2 (BCL2), platelet endothelial cell adhesion molecule (CD31), Doublesex and Mab-3 Related Transcription Factor 1 (DMRT1), GATA Binding Protein 4 (GATA4), hypoxia inducible factor 1 alpha subunit (HIF1A) (Oliver et al., 2011), insulin like protein 3 (INSL3), KI67, tumour suppressor protein 53 (P53), prostaglandin F2 α receptor (PTGFR) (Kaczynski and Waclawik, 2013), secreted phosphoprotein 1 (SPP1) (Hernández et al., 2013), and vascular endothelial growth factor A (VEGFA) (Oliver et al., 2011). Two reference genes were used: TATA box binding protein 1 (TBP1) (Nygard et al., 2007) and Topoisomerase II Beta (TOP2B) (Erkens et al., 2006). ...
Article
It is estimated that intra-uterine growth restricted piglets represent 25% of the total number of piglets born. Growth restricted female piglets have impaired reproductive performance postnatally, however, when during gestation this phenotype arises is not known. With this study, the aim was to improve the understanding of foetal ovarian development in normal and small foetuses throughout gestation. Female Large White X Landrace foetuses were obtained at gestational day (GD) 45, 60 and 90 (n = 5-6 litters/GD). Histological analysis of GATA4 stained foetal ovaries at GD60 and 90 indicated there were fewer primary follicles (P ≤ 0.05) in the foetuses weighing the least compared to those with a weight similar to the mean for the litter (CTMLW) at GD90. Plasma oestradiol concentrations were less in the foetuses with lesser weights compared with greater weight foetuses at GD90 (P ≤ 0.05). The RNA was extracted from ovaries of the lesser weight and CTMLW foetuses at GD45, 60 and 90 and qPCR was performed to quantify relative abundance of 12 candidate mRNAs for which encoded proteins modulate ovarian function and development. Gestational changes in relative abundances of CD31, PTGFR, SPP1 and VEGFA mRNA transcripts were observed. Relative abundance of KI67 (P = 0.066) and P53 (P ≤ 0.05) was less in ovaries of the lesser weight compared to CTMLW foetuses at GD60. There was a lesser relative abundance of PTGFR mRNA transcript in ovaries from the foetuses with lesser weight compared to CTMLW foetuses at GD45 and 60 (P ≤ 0.05). These findings indicate that postnatal differences in reproductive potential of growth restricted females are programmed early in gestation. It is hoped that further investigation will improve the understanding of the relationship between prenatal reproductive development and postnatal reproductive performance.
Article
In women of childbearing age, extensive decidualization, shedding and remodeling of the endometrium during the menstrual cycle are fundamental for successful pregnancy. The role of prostaglandins (PGs) in menstruation has long been proposed in humans, and the rate-limiting enzyme cyclooxygenase was shown to play a key role in endometrial breakdown and shedding in a mouse menstrual-like model in our previous study. However, the specific types of PGs involved and their respective roles remain unclear. Therefore, our objective was to investigate the mechanism through which PGs regulate endometrial disintegration. In this study, the microscopy was observed by HE; the protein levels of prostaglandins E1 (PGE1), prostaglandins E2 (PGE2), prostaglandin F2α (PGF2α) and Prostaglandin I2 (PGI2) were detected by ELISA; the mRNA level of Pfgfr2, Vascular Endothelial Growth Factor(Vegf), Angiostatin and Hypoxia inducible factor-1α (Hif1α) were examined by real-time PCR; PTGFR Receptor (PTGFR), VEGF, Angiostatin and HIF-1α protein levels were investigated by western blotting; the locations of protein were observed by Immunohistochemistry; HIF-1α binding PTGFR promoter was detected by Chromatin Immunoprecipitation (ChIP) and real-time PCR. We found that the concentrations of PGE1, PGE2, and PGF2α all increased significantly during this process. Furthermore, Ptgfr mRNA increased soon after Progesterone (P4) withdrawal, and PTGFR protein levels increased significantly during abundant endometrial breakdown and shedding processes. PTGFR inhibitors AL8810 significantly suppressed endometrial breakdown and shedding, promoted Angiostatin expression, and reduced VEGF-A expressions and vascular permeability. And HIF-1α and PTGFR were mainly located in the luminal/gland epithelium, vascular endothelium, and pre-decidual zone. Interestingly, HIF-1α directly bound to Ptgfr promoter. Moreover, a HIF-1α inhibitor 2-methoxyestradiol (2ME) significantly reduced PTGFR expression and suppressed endometrial breakdown which was in accord with PTGFR inhibitor’s effect. Similar changes occurred in human stromal cells relevant to menstruation in vitro. Our study provides evidence that PGF2α/PTGFR plays a vital role in endometrial breakdown via vascular changes that are regulated by HIF-1α during menstruation.
Article
Full-text available
Pregnancy establishment in mammals, including pigs, requires coordinated communication between developing conceptuses (embryos with associated membranes) and the maternal organism. Porcine conceptuses signalize their presence by secreting multiple factors, of which estradiol-17β (E2) is considered the major embryonic signal initiating the maternal recognition of pregnancy. During this time, a limited supply of prostaglandin (PGF2α) to the corpora lutea and an increased secretion of luteoprotective factors (e.g., E2 and prostaglandin E2 [PGE2]) lead to the corpus luteum's maintained function of secreting progesterone, which in turn primes the uterus for implantation. Further, embryo implantation is related to establishing an appropriate proinflammatory environment coordinated by the secretion of proinflammatory mediators including cytokines, growth factors, and lipid mediators of both endometrial and conceptus origin. The novel, dual role of PGF2α has been underlined. Recent studies involving high-throughput technologies and sophisticated experimental models identified a number of novel factors and revealed complex relationships between these factors and those already established. Hence, it seems that early pregnancy should be regarded as a sequence of processes orchestrated by pleiotropic factors that are involved in redundancy and compensatory mechanisms that preserve the essential functions critical for implantation and placenta formation. Therefore, establishing the hierarchy between all molecules present at the embryo-maternal interface is now even more challenging.
Article
Prostaglandins (PG) have primary functions in the reproductive tract, however, the mechanism of regulation of PG secretion in the endometrium is unclear. Estrogen as a predominant regulator of uterine functions during the mammalian estrous cycle and effects of estrogen on synthesis of PG and function in uterine tissues of cattle are not fully understood. In this study, there was evaluation of the concentration- and time-effects of 17β-estradiol on PG synthesis in endometrial explants of cattle, focusing on the secretion of prostaglandin E2 (PGE2) and prostaglandin F2α (PGF2α) as well as relative abundance of mRNA transcript and protein for both the enzymes responsible for PGE2 and PGF2α synthesis, including prostaglandin-endoperoxide synthase 1 and 2 (PTGS1, PTGS2), PGE2 synthase (PGES), PGF2α synthase (PGFS), and carbonyl reductase (CBR1), and the receptors responsible for downstream PGE2 (PTGER2, PTGER4) and PGF2α (PTGFR) signaling. Results indicated that 17β-estradiol increased PGE2 and PGF2α production at concentrations ranging from 10⁻¹¹ to 10⁻⁸ M. Furthermore, abundances of PTGS1, PTGS2, PGES, PGFS, PTGER2, PTGER4, and PTGFR mRNA transcripts and protein were greater immediately after 17β-estradiol treatment at almost all the concentrations, while these CBR1 abundances were less as a result of treatments with 17β-estradiol. These data support the hypothesis that estradiol modulates the synthesis and function of PG in the endometrium of cattle.
Article
Full-text available
A functional cDNA clone for mouse prostaglandin (PG) F receptor was isolated from a mouse cDNA library using polymerase chain reaction based on the sequence of cloned prostanoid receptors, and cross-hybridization screening. The mouse PGF receptor consists of 366 amino acid residues with putative seven transmembrane domains. The sequence revealed the highest homology to the EP1 subtype of PGE receptor and thromboxane (TX) A2 receptor. Ligand binding studies using membranes of COS cells transfected with the cDNA revealed specific [3H]PGF2 alpha binding. The binding was displaced with unlabeled PGs in the order of PGF2 alpha = 9 alpha, 11 beta PGF2 > PGF 1 alpha > PGD2 > STA2 (a stable TXA2 agonist) > PGE2 > iloprost (a stable PGI2 agonist). PGF2 alpha increased inositol trisphosphate formation in a concentration-dependent manner in COS cells expressing PGF receptor. RNA blot and in situ hybridization analyses demonstrated that the PGF receptor transcripts are abundantly expressed in luteal cells of corpus luteum and in a lesser amount in kidney, heart, stomach, and lung.
Article
Full-text available
Prostaglandins regulate many physiological functions, including reproduction, by binding to specific plasma membrane receptors. In this study we evaluated the regulation of PGF(2α) (FP) and PGE (EP3 subtype) receptors in ovine corpora lutea. In the first study, tissue distribution of FP and EP3 receptors was evaluated in 13 ovine tissues. FP receptor mRNA was present in 100-fold higher concentration in corpora lutea than in other tissues. Similarly, [3H]PGF(2α) binding was much greater in luteal plasma membranes than in membranes from other tissues. In contrast, EP3 receptor mRNA was more uniformly distributed, with high concentrations in adrenal medulla, inner myometrium, kidney medulla and heart. The distribution of [3H]PGE1 binding was generally similar to EP3 mRNA, with the exception that ovarian stroma, endometrium and outer myometrium had high [3H]PGE1 binding but low concentrations of EP3 receptor mRNA. The second study evaluated the action of PGF(2α) on luteal mRNA encoding FP and EP3 receptors. Ewes had PGF(2α) or saline infused into the ovarian artery and corpora lutea were removed at 0, 1, 4, 12 and 24 h. FP receptor mRNA decreased by 50% at 12 and 24 h after infusion with PGF(2α), whereas EP3 mRNA was unchanged. Treatment of large luteal cells with PGF(2α), phorbol didecanoate (protein kinase C activator), or ionomycin (calcium ionophore) decreased FP receptor mRNA after 24 h (P < 0.05). Glyceraldehyde 3-phosphate dehydrogenase mRNA was not changed by any treatment. These results show that EP3 receptors are expressed in many tissues and expression is not regulated by PGF(2α). In contrast, FP receptors are primarily expressed in corpora lutea and expression is inhibited by PGF(2α).
Article
Full-text available
Ovarian progesterone induces essential changes leading to a temporary state of uterine receptivity for conceptus implantation. Estrogens secreted by the porcine conceptus on days 11 and 12 of pregnancy provide the initial signal for maternal recognition of pregnancy and maintenance of a functional corpus luteum (CL) for continued production of progesterone. As prostaglandins F(2)(α) (PGF(2)(α)) and E(2) (PGE(2)) exert opposing actions on the CL, a tight control over their synthesis and secretion is critical either for the initiation of luteolysis or maintenance of pregnancy. One of the supportive mechanisms by which conceptus inhibits luteolysis is changing PG synthesis in favor of luteoprotective PGE(2). Conceptus PGE(2) could be amplified by PGE(2) feedback loop in the endometrium. In pigs, as in other species, implantation and establishment of pregnancy is associated with upregulation of expression of proinflammatory factors, which include cytokines, growth factors, and lipid mediators. The conceptus produces inflammatory mediators: interferon γ and interferon δ, interleukins IL1B and IL6, and PGs, which probably activate inflammatory pathways in the endometrium. The endometrium responds to these embryonic signals by enhancing further progesterone-induced uterine receptivity. Understanding the mechanisms of pregnancy establishment is required for translational research to increase reproductive efficiencies and fertility in humans and animals.
Article
Full-text available
Oxytocin (OXT) and tumor necrosis factor α (TNF) have been implicated in the control of luteolysis by stimulating endometrial secretion of luteolytic prostaglandin F(2α) (PGF(2α)). Nevertheless, OXT concentration in porcine uterine lumen increases markedly on days 11-12 of pregnancy, and TNF is expressed in endometrium during pregnancy. The objective of the study was to determine the effect of OXT and TNF on expression of the enzymes involved in PG synthesis: PG-endoperoxide synthase 2 (PTGS2), PGE(2) synthase (mPGES-1) and PGF synthase, and PGE(2) receptor (PTGER2), as well as on PG secretion by endometrial luminal epithelial cells (LECs) on days 11-12 of the estrous cycle and pregnancy. LECs isolated from gilts on days 11-12 of the estrous cycle (n=8) and pregnancy (n=7) were treated with OXT (100  nmol/l) and TNF (0.6  nmol/l) for 24  h. OXT increased PTGS2 mRNA and mPGES-1 protein contents, as well as PGE(2) secretion but only on days 11-12 of pregnancy. TNF stimulated PTGS2 and mPGES-1 mRNA, as well as mPGES-1 protein expression and PGE(2) release on days 11-12 of pregnancy and the estrous cycle. In addition, expressions of PTGER2 and PTGER4 were determined in corpus luteum (CL). Abundance of PTGER2 mRNA and PTGER4 protein in CL was upregulated on day 14 of pregnancy versus day 14 of the estrous cycle. This study indicates that TNF and OXT regulate PGE(2) synthesis in LECs during early pregnancy. PGE(2) secreted by LECs, after reaching ovaries, could have a luteoprotective effect through luteal PTGER2 and PTGER4, or may directly promote uterine function and conceptus development.
Article
A protein determination method which involves the binding of Coomassie Brilliant Blue G-250 to protein is described. The binding of the dye to protein causes a shift in the absorption maximum of the dye from 465 to 595 nm, and it is the increase in absorption at 595 nm which is monitored. This assay is very reproducible and rapid with the dye binding process virtually complete in approximately 2 min with good color stability for 1 hr. There is little or no interference from cations such as sodium or potassium nor from carbohydrates such as sucrose. A small amount of color is developed in the presence of strongly alkaline buffering agents, but the assay may be run accurately by the use of proper buffer controls. The only components found to give excessive interfering color in the assay are relatively large amounts of detergents such as sodium dodecyl sulfate, Triton X-100, and commercial glassware detergents. Interference by small amounts of detergent may be eliminated by the use of proper controls.
Article
Polyvinyl catheters were inserted into the right and left utero-ovarian veins (UOV) and saphenous vein (SV) and artery (SA)_of six nonpregnant (O) and five pregnant (P) gilts on day 11 after onset of estrus. Beginning on day 12, UOV blood samples were collected at 15-min intervals from 0800 to 1100 hr and 2000 to 2300 hr, and single samples were taken at 1200 and 2400 hrs. Peripheral blood (SA or SV) was sampled at 0800, 1200, 2000 and 2400 hr until gilts returned to estrus () or day 24 or pregnancy. UOV plasma PGF concentrations (ng/ml; n = 1929) were measured by RIA. Status (P O) by day interactions were detected (P<.01) but variances among treatments were heterogenous (P<.01). Cuvilinear day trends were detected for PGF in 0 gilts (P<.01) but not P gilts. PGF peaks, defined as concentrations greater than two SD above the mean concentration for each gilt, occurred with greater frequency ((ifχ2 = 16.4; P>.01)) in 0 than P gilts; and mean peak levels () were 5.0 ± .27 and 3.84 ± .13 ng/ml, respectively.Progesterone concentrations were maintaiend in pregnant pigs and were indicative of luteal maintenance. Systematic differences in day trends of utero-ovarian venous plasma estradiol were detected between O and P pigs. These differences may be of paramount physiological importance and are discussed.
Article
Homeobox A10 (HOXA10) gene expression was demonstrated in the endometrium of adult porcine uteri, however there is little information concerning the role of this gene in the pig. Objectives of the present study were to examine: 1) the expression of HOXA10 in the endometrium of cyclic and early pregnant gilts; 2) the effect of estradiol (E(2)) and progesterone (P(4)) on HOXA10 expression in porcine luminal epithelial (LE) and stromal (ST) cells in vitro; 3) the effect of E(2) and conceptus-exposed medium (CEM) on HOXA10 and prostaglandin endoperoxide synthase (PTGS2) gene expression and prostaglandin (PG) E(2) secretion from LE and ST cells in a co-culture model. The abundance of HOXA10 mRNA was increased on day 15 of pregnancy in comparison to day 15 of the estrous cycle. Moreover, increased HOXA10 mRNA level was detected in ST cells after E(2) and P(4) treatment. E(2) stimulated the expression of HOXA10 in LE cells cultured on collagen and pre-treated with steroids, but not in LE on plastic surfaces. Addition of CEM to LE cells cultured in collagen-coated inserts of the co-culture system resulted in elevated HOXA10 and PTGS2 gene expression and PGE(2) secretion in these cells, but not in ST cells cultured in basal compartments. ST cells directly treated with E(2) or CEM showed higher levels of HOXA10 and PTGS2 expression. Blocking of estrogen receptors with ICI-182,780 did not influence the stimulatory effect of CEM. We conclude that HOXA10 expression in the porcine endometrium is closely related to the implantation process and stimulated by conceptus products. Moreover, the co-culture system of LE and ST cells is a promising model for the study of endometrial response to conceptus-derived factors.
Article
In ruminants, both the endometrium and the conceptus (embryo and associated extraembryonic membranes) trophectoderm synthesizes and secretes prostaglandins (PG) during early pregnancy. In mice and humans, PGs regulate endometrial function and conceptus implantation. In Study One, bred ewes received intrauterine infusions of vehicle as a control (CX) or meloxicam (MEL), a PG synthase (PTGS) inhibitor from Days 8-14 postmating, and the uterine lumen was flushed on Day 14 to recover conceptuses and assess their morphology. Elongating and filamentous conceptuses (12 cm to >14 cm in length) were recovered from all CX-treated ewes. In contrast, MEL-treated ewes contained mostly ovoid or tubular conceptuses. PTGS activity in the uterine endometrium and amounts of PGs were substantially lower in uterine flushings from MEL-treated ewes. Receptors for PGE2 and PGF2 alpha were present in both the conceptus and the endometrium, particularly the epithelia. In Study Two, cyclic ewes received intrauterine infusions of CX, MEL, recombinant ovine interferon tau (IFNT), or IFNT and MEL from Days 10-14 postestrus. Infusion of MEL decreased PGs in the uterine lumen and expression of a number of progesterone-induced endometrial genes, particularly IGFBP1 and HSD11B1. IFNT increased endometrial PTGS activity and the amount of PGs in the uterine lumen. Interestingly, IFNT stimulation of many genes (FGF2, ISG15, RSAD2, CST3, CTSL, GRP, LGALS15, IGFBP1, SLC2A1, SLC5A1, SLC7A2) was reduced by co-infusion with MEL. Thus, PGs are important regulators of conceptus elongation and mediators of endometrial responses to progesterone and IFNT in the ovine uterus.
Article
The aim was to evaluate expression of genes involved in the biosynthesis of prostaglandins (PTG), Prostaglandin H Synthase-1 (PTGS1) and PTGS2, PGF synthase (PTGFS), and PGE synthase (PTGES), PGF receptor (PTGFR), PGE receptors (PTGER2 and PTGER4), prostaglandin transporter (SLCO2A1) and hydroxyprostaglandin dehydrogenase-15 (HPGD). Endometrial biopsies were obtained from mares on day of ovulation (d0, n=4), late diestrus (LD, n=4), early luteolysis (EL, n=4) and after luteolysis (AL, n=4) during the cycle. Stages of the cycle were confirmed by plasma progesterone concentrations measured daily and ultrasound examinations. Biopsies were also taken on days 14 (P14; n=4), 15 (P15, n=4), 18 (P18, n=4) and 22 (P22; n=4) of pregnancy. Relative mRNA expressions were quantified using real-time RT-PCR. A mixed model was fitted on the normalized data and least significant difference test (α=0.05) was employed. Expression of PTGS1 mRNA was low throughout the estrous cycle and early days of pregnancy, but upregulated on P18 and P22. PTGS2 expression was increased on EL, but it was suppressed by pregnancy on P15, P18, and P22. PTGFS expression was upregulated in both cyclic and pregnant mares compared to d0 and its level was the highest on LD. PTGFR expression was transiently increased on LD and EL and was suppressed during early pregnancy. Both PTGES and PTGER2 expressions were increased on LD, EL, and early pregnancy, but were decreased after the luteolysis in cyclic mares as they remained high on P18 and P22. PTGER4 expression did not change throughout the cycle and early pregnancy. Levels of HPGD and SLCO2A1 were significantly increased only on P22. In conclusion, PTGS2 expression increases around the time of luteolysis and concurrent upregulation of PTGFS and PTGES indicates that equine endometrium has increased capability of PTG production around the time of luteolysis. However, pregnancy reduces PTGS2 expression, but maintains the high levels of PTGES during early pregnancy along with PTGER2 while PTGFR expression was suppressed. These findings suggest that possible luteotrophic action of PGE₂ is required in early equine pregnancy. PTGS1 is only upregulated later in the early pregnancy suggesting that it is not involved in luteolysis, but could be the main PTGS enzyme at this time during early pregnancy. An increase in HPGD and SLCO2A1 levels on P22 indicates a tight regulation of PTG action by pregnancy.