ArticlePDF Available

In human granulosa cells from small antral follicles, androgen receptor mRNA and androgen levels in follicular fluid correlate with FSH receptor mRNA

Authors:

Abstract and Figures

Human small antral follicles (diameter 3-9 mm) were obtained from ovaries surgically removed for fertility preservation. From the individual aspirated follicles, granulosa cells and the corresponding follicular fluid were isolated in 64 follicles, of which 55 were available for mRNA analysis (24 women). Expressions of androgen receptor (AR) mRNA levels in granulosa cells, and of androstenedione and testosterone in follicular fluid, were correlated to the expression of the FSH receptor (FSHR), LH receptor (LHR), CYP19 and anti-Müllerian Hormone-receptor II (AMHRII) mRNA in the granulosa cells and to the follicular fluid concentrations of AMH, inhibin-B, progesterone and estradiol. AR mRNA expression in granulosa cells and the follicular fluid content of androgens both showed a highly significant positive association with the expression of FSHR mRNA in granulosa cells. AR mRNA expression also correlated significantly with the expression of AMHRII, but did not correlate with any of the hormones in the follicular fluid. These data demonstrate an intimate association between AR expression in immature granulosa cells, and the expression of FSHR in normal small human antral follicles and between the follicular fluid levels of androgen and FSHR expression. This suggests that follicular sensitivity towards FSH stimulation may be augmented by stimulation of androgens via the AR.
Content may be subject to copyright.
ORIGINAL RESEARCH
In human granulosa cells from small
antral follicles, androgen receptor
mRNA and androgen levels in
follicular fluid correlate with FSH
receptor mRNA
M.E. Nielsen1, I.A. Rasmussen1, S.G. Kristensen2, S.T. Christensen3,
K. Møllga
˚rd4, E. Wreford Andersen 5, A.G. Byskov2, and
C. Yding Andersen2,*
1
The Fertility Clinic, Odense University Hospital, Odense, Denmark
2
Laboratory of Reproductive Biology, Section 5712, The Juliane Marie
Centre for Women, Children and Reproduction, University Hospital of Copenhagen, University of Copenhagen, Blegdamsvej 9,
Rigshospitalet, Copenhagen DK-2100, Denmark
3
Department of Cell and Developmental Biology, August Krogh Institute, University of
Copenhagen, Copenhagen, Denmark
4
Department of Cellular and Molecular Medicine, The Panum Institute, University of Copenhagen,
Copenhagen, Denmark
5
Department of Biostatistics, University of Copenhagen, Copenhagen, Denmark
*Correspondence address: Tel: +45-35455822; Fax: +45-35455824; E-mail: yding@rh.dk
Submitted on April 17, 2010; resubmitted on August 24, 2010; accepted on August 26, 2010
abstract: Human small antral follicles (diameter 3– 9 mm) were obtained from ovaries surgically removed for fertility preservation.
From the individual aspirated follicles, granulosa cells and the corresponding follicular fluid were isolated in 64 follicles, of which 55 were
available for mRNA analysis (24 women). Expressions of androgen receptor (AR) mRNA levels in granulosa cells, and of androstenedione
and testosterone in follicular fluid, were correlated to the expression of the FSH receptor (FSHR), LH receptor (LHR), CYP19 and anti-
Mu
¨llerian Hormone-receptor II (AMHRII) mRNA in the granulosa cells and to the follicular fluid concentrations of AMH, inhibin-B,
progesterone and estradiol. AR mRNA expression in granulosa cells and the follicular fluid content of androgens both showed a highly
significant positive association with the expression of FSHR mRNA in granulosa cells. AR mRNA expression also correlated significantly
with the expression of AMHRII, but did not correlate with any of the hormones in the follicular fluid. These data demonstrate an intimate
association between AR expression in immature granulosa cells, and the expression of FSHR in normal small human antral follicles and
between the follicular fluid levels of androgen and FSHR expression. This suggests that follicular sensitivity towards FSH stimulation may
be augmented by stimulation of androgens via the AR.
Key words: AR / FSHR / LHR mRNA expression / steroids / small human antral follicles
Introduction
During folliculogenesis androgens serve an important function as sub-
strate for granulosa cell estradiol synthesis, and perform, in addition, a
number of trophic functions during follicular growth, especially in
earlier developmental stages prior to follicular selection. Studies in
rhesus monkeys almost a decade ago showed that androgens, adminis-
tered in pellets that provide slow release for 3–10 days, stimulate
growth in follicles until the early antral stage (Vendola et al., 1998,
1999a,b). Proliferation of granulosa cells was significantly increased
and the number of growing follicles augmented after exposure to tes-
tosterone or dihydrotestosterone treatment (Vendola et al., 1998,
1999a,b). Furthermore, androgen receptor (AR) gene expression in
granulosa cells from these primate ovaries, evaluated by in situ hybri-
dazation, correlated positively with proliferation and negatively with
apoptosis (Weil et al., 1998).
Subsequent studies demonstrated interactions between FSH and
androgens in the follicular development and revealed that follicles
demonstrate a highly significant positive correlation between the
FSH receptor (FSHR) and AR mRNA levels when comparing the
mean expression from each group of follicles in each individual
monkey (Weil et al., 1999). Furthermore, testosterone administration
that resulted in supra-physiological levels augmented granulosa
cell FSHR expression and it was suggested that androgens promote
&The Author 2010. Published by Oxford University Press on behalf of the European Society of Human Reproduction and Embryology. All rights reserved.
For Permissions, please email: journals.permissions@oup.com
Molecular Human Reproduction, Vol.17, No.1 pp. 63– 70, 2011
Advanced Access publication on September 14, 2010 doi:10.1093/molehr/gaq073
by guest on December 27, 2015http://molehr.oxfordjournals.org/Downloaded from
follicular growth by sensitizing granulosa cells to FSH action (Weil
et al., 1999).
In the ovary, the AR seemed predominantly to be expressed by the
granulosa cells and in particular by the smaller follicles (rats: Harlow
et al., 1986;Tetsuka and Hillier, 1997; marmoset: Hillier et al.,
1997). However, in humans and sheep, the pattern of AR expression
in granulosa cells appears to be regulated differently from that in rat
and monkey, since granulosa cell AR immunostaining was reported
to be stronger in pre-ovulatory follicles than in smaller follicles
(human: Horie et al., 1992, sheep: Campo et al., 1985). A recent
study in mouse that used granulosa cells and oocyte-specific AR
knockout mice found that such mice had premature ovarian failure
and were subfertile, with longer oestrous cycles and fewer ovulated
oocytes (Sen and Hammes, 2010). It was concluded that the AR
appears to promote pre-antral follicle growth and prevent follicular
atresia and are essential for normal follicular development and fertility
(Sen and Hammes, 2010).
At the clinical level, the above observations may explain the
response women with hyper-androgenism often show when treated
with exogenous FSH administration for ovulation induction. These
women may react with an exaggerated follicular response, suggesting
that their granulosa cells are especially sensitive to FSH stimulation
(Farhi and Jacobs, 1997;Thessaloniki ESHRE/ASRM-Sponsored
PCOS Consensus Workshop Group, 2008). Indeed, a number of
studies have shown that granulosa cells from PCOS women during
culture are hyper-responsive to FSH and possess an augmented
capacity for synthesizing estrogens (Mason et al., 1994). In a group
of women who showed a moderate or poor response to ovarian
stimulation with exogenous gonadotrophins the use of locally pro-
duced androgens to augment sensitivity to FSH stimulation and to
increase the number of retrieved mature oocytes has recently been
attempted (Løssl et al., 2006). Prior to controlled ovarian stimulation
with exogenous gonadotrophins local ovarian androgen production
was enhanced by pre-treating women with a bolus of hCG, thereby
stimulating local androgen production and by co-administering an
aromatase inhibitor to prevent the conversion of androgens to estro-
gens (Løssl et al., 2006). Compared with a control group the androgen
pretreatment group developed significantly more embryos following
ovarian stimulation and IVF treatment, supporting the concept that
androgens may also enhance follicular responsiveness and FSHR
expression in humans. However, since the AR appears to be mainly
expressed in pre-ovulatory follicles in women (Horie et al., 1992), it
is unclear at what developmental stage FSH sensitivity is enhanced
and whether it is feasible to augment follicular responsiveness to
smaller follicles.
Human small antral follicles are scarce material and not many
studies have been performed on such individual small follicles. In the
present study, we took advantage of the presence of normal small
antral follicles in ovaries surgically removed for fertility preservation
by cryopreservation of the ovarian cortex. The antral follicles
located in the medually part cannot sustain freezing and are normally
discarded in this procedure. In order to characterize human antral fol-
licles and evaluate whether AR expression played similar roles in
humans as found in primates and other species, the aim of the
present study was to determine AR mRNA expression in granulosa
cells of normal small human antral follicles and to correlate that
with the mRNA expression of FSHR, LH receptor (LHR), AMH
receptor II (AMHRII) and aromatase (CYP19). In addition, the hormo-
nal characteristics of the corresponding follicular fluid were also
evaluated.
Materials and Methods
Patients and collection of small antral
follicular fluid
Granulosa cell and follicular fluid samples were obtained from individual
small antral follicles present in ovaries surgically removed for fertility pres-
ervation. Cryopreservation of the ovarian cortex was offered as means of
fertility preservation to patients in whom the appropriate treatment posed
a high risk of eliminating the ovarian follicle pool.
A total of 64 follicles were obtained from 24 women aged 15– 37 years
(median 29 years) at various times during their menstrual cycle. Diagnosis
for ovarian cryopreservation included mammary cancer (9), Hodgkin’s and
non-Hodgkin’s disease (5), Ewing and oestogenic sarcoma (3), lymphoma
(2), rectum cancer (1), cervical cancer (2), medulla blastoma (1) and der-
matomyositis (1). None of the women received GnRH agonists and two
women received oral contraception. All ovaries appeared normal by (i)
visual inspection in connection with the cryopreservation procedure and
(ii) by subsequent microscopic evaluation of histological sections from a
small piece of the ovarian cortex.
The follicles were collected immediately after recovery of the ovary
prior to, or during, isolation of the ovarian cortex. Each antral follicle
visible on the surface of the ovary or observed during preparation of
cortex was aspirated with a 1 ml syringe with a 26-gauge needle
(Becton Dickinson, Brøndby, Denmark). The granulosa cells of each
small follicle were isolated from the follicular fluid by centrifugation
(2000g, 2 min). After aspirating the follicular fluid from the granulosa
cells in the pellet, each ampoule of granulosa cells and follicular fluid
were snap-frozen in liquid nitrogen and stored at 808C until RNA puri-
fication or until measurement of hormones.
One to eight antral follicles were collected per patient (one follicular
fluid, five patients; two follicular fluids, eight patients; three follicular fluids,
seven patients; four follicular fluids, two patients; six follicular fluids, one
patient; eight follicular fluids, one patient). The volume of each follicle
was estimated from the volume in the syringe and only those with a
volume from 30 to 350 ml corresponding to 3 9 mM in diameter
were included in the study. The ethical committee of the municipalities
of Copenhagen and Frederiksberg approved the project. Results from
this material have been used in a previous publication (Nielsen et al.,
2010).
RNA purification
Total RNA was purified under RNase-free conditions at room tempera-
ture using Tri Reagent (Sigma-Aldrich, St Louis, MO, USA) in combination
with the RNAeasy Mini Kit (Qiagen, Hilden, Germany). Each cell sample
was lysed in 1 ml Tri Reagent according to the Tri Reagent protocol and
incubated at room temperature for 5 min. Following addition of 200 ml
1-bromo-3-chloropropane (Sigma-Aldrich), the cell sample solutions
were shaken vigorously for 15 s and incubated at room temperature for
5 min. Following centrifugation at 15 000gfor 15 min at 48C, the
mixture separated into three phases and the upper aqueous phase con-
taining the RNA was transferred to a clean, RNase-free Eppendorf tube.
The following steps of RNA purification was carried out according to
the RNeasy Mini Kit protocol. The final elution step was repeated and
the purified total RNA was subsequently stored at 2608C.
The samples were analysed for total RNA quality and level of degra-
dation using an Agilent 2100 Bioanalyzer and RNA 6000 Pico LabChip
64 Nielsen et al.
by guest on December 27, 2015http://molehr.oxfordjournals.org/Downloaded from
according to the manufacturer’s instructions (RNA 6000 Pico assay kit,
Agilent Technologies, Waldbronn, Germany). RNA samples which did
not show two peaks representing 18 and 28 s rRNA were excluded.
Total RNA quantity was evaluated using the Nanodrop
w
ND1000 Spec-
trophotometer (NanoDrop Technologies, Wilmington, DE, USA).
DNA synthesis and RTPCR analysis
First-strand cDNA was synthesized using the High-Capacity cDNA Reverse
Transcription Kit (Applied Biosystems, Carlsbad, CA, USA) according to the
manufacturer’s manual. A master mix containing 2.0 ml10×RT Buffer,
0.8 ml25×dNTP Mix (100 mM), 2.0 mlof10×RT random primers,
1.0 ml of MultiScribeTM Reverse Transcriptase (50 U/ml), 1.0 mlofRNase
Inhibitor and 3.2 ml of nuclease-free (Diethylpyrocarbonate, DEPC) water
was prepared and for each 20 ml of reaction 10 ml of master mixture was
added to an equal volume of total RNA (concentrations ranging from 5
to 10 ng/ml). All steps were performed on ice. Samples were centrifuged
briefly at 12 000gand then incubated at room temperature for 10 min, fol-
lowed by 378C for 2 h and finally, the reaction was stopped at 858Cfor5s.
First-strand cDNA was stored at 2208C until real-time PCR analysis.
Evaluation of gene expression levels was achieved by relative quantifi-
cation (RQ) real-time PCR analysis using TaqMan
w
technology (Applied
Biosystems). FSHR, LHR, CYP19a1 and AMH-r2 TaqMan
w
Gene Expres-
sion Assays (pre-designed) as well as the Endogenous Control Assays
for human b-actin and glyceraldehyde 3-phosphatdehydrogenase
(GAPDH) were purchased from Applied Biosystems (Assay id-no.:
b-actin:#4326315E; GAPDH:#4333764F; AR:#Hs00171172_m1 FSHR:#
Hs00174865_m1; LHR:#Hs00174885_m1; AMHRII:#Hs00179718_m1;
CYP19a1:#Hs00903413_m1). Sample triplicates were prepared according
to the manufacturer’s instructions. A total reaction volume of 25 mlwas
prepared on ice containing 12.5 ml TaqMan
w
Universal PCR Master Mix,
No AmpErase
w
UNG(2X), 1.25 ml20XTaqMan
w
Gene Expression
Assay Mix, 1.7 –1.85 ml undiluted cDNA and RNase-free water up to
25 ml. The samples were then centrifuged at 2800gat 48Cfor5min.
cDNA was amplified using the 7500 Real-time PCR system (Applied
Biosystems) under the following thermal cycling conditions: 958C for
10 min, 45 cycles of 958C for 15 s and 608C for 1 min. The data were sub-
sequently analysed using human b-actin and GAPDH as housekeeping
genes and the SDS Software for Relative Quantification (Applied Biosys-
tems). The calculation of the expression levels of each individual gene
was carried out according to the Comparative CTMethod for RQ as
described by the manufacturer (Relative Quantification of Gene
Expression: User Bulletin #2, Applied Biosystems). The expression levels
were related to the housekeeping genes human b-actin and GAPDH.
The expression level for each gene was calculated according to human
b-actin and GAPDH separately, and the DDC
T
values were compared. If
no deviation was observed, the housekeeping gene expression was con-
sidered to be constant. As another test for variation in housekeeping
gene expression, the ratios between the RQ values for the five target
genes calculated according to b-actin or GAPDH, were compared. On
the basis of these two tests, the housekeeping genes were considered
to remain sufficient constantly expressed. Often, expression levels are
expressed as 2DDCTwhen compared with the expression of a calibrator
or another target gene. Normalized expression levels can also be
expressed as 2DDCT, for example, when expression levels are not
related to a calibrator. The expression levels in this study are therefore
presented as 2DCTand normalized to GAPDH (Applied Biosystems
Support; Schmittgen and Livak, 2008).
Hormone measurements
Estradiol and progesterone were measured using commercially available
RIA kits (DSL-43100 & DSL-3400; Diagnostic System Laboratories,
Webster, TX, USA). Samples for both assays were diluted 1:50 in steroid-
free serum just prior to measurement. Androstenedione was measured
using an RIA kit (DSL-3800) with samples being diluted 1:200 in steroid-
free serum and testosterone using an RIA kit (DSL-4000) diluted 1:100
in steroid-free serum.
The steroid-free serum was produced in-house by stripping a pool of
serum with repeated washings of charcoal particles. The charcoal was
removed through several filtering steps ending with a sterile filter to
ensure total removal of charcoal particles. Blank samples were tested
for the absence of steroids prior to the running hormone assays.
AMH was measured using a specific ELISA-kit according to the manufac-
turer’s instructions (DSL-10-14400; Diagnostic System Laboratories). Fol-
licular fluid samples from small antral follicles were diluted either 1:500 or
1:3000 in the zero standard provided by the manufacturer. Inter-assay
variation of a sample containing 7.6 ng AMH/ml was 4.4 % (n¼12) and
intra-assay variation was 3.3% (n¼5) of sample containing 0.45 ng/ml.
Dilution curves of follicular fluid samples proved parallel to the standard
curve.
Inhibin-B was measured using a specific ELISA-kit according the manu-
facturer’s instructions (The Oxford Bio-innovation kit; Biotech-IgG,
Copenhagen, Denmark). Prior to measurement, all follicular fluid
samples irrespective of whether they derived from small antral or pre-
ovulatory follicles were diluted 1:100 or 1:500 in serum obtained from a
pool of five post-menopausal women (who had no inhibin-B activity).
The follicular fluids were pre-treated with SDS, heated, and exposed to
hydrogen peroxide before they were applied to the wells of the plate
and incubated overnight at room temperature. Subsequently, the plates
were washed and incubated with detection antibody for 3 h at room
temperature. Substrate solution was applied and incubated for 1 h. The
amplifier solution was added, and the plates were read at 490 nm
with an ELISA reader with its reference at 620 nm (coefficient of
variation ,7%).
SDSpolyacrylamide gel electrophoresis and
western blot analysis
Cell lysates were prepared by using 0.5% sulphSDS lysis buffer. Lysates
were sonicated and centrifuged at 20 000gto precipitate debris. Protein
concentrations were compared with a BCA protein standard (Pierce Bio-
technology, Rockford, IL, USA) and measured with a Protein assay
(BioRAD Laboratories, CA, USA, DC based on Lowry’s method) ensuring
that equal amounts of protein could be loaded in each lane. Proteins were
analysed by SDS–polyacrylamide gel electrophoresis (PAGE) and western
blot analysis using the XCell and XCell II blot module systems (Novex) as
described (Christensen et al., 2001). Briefly, proteins were separated by
gel electrophoresis under denaturing and reducing conditions on 10%
NuPAGE Bis–Tris gels (NP0303BOX, Invitrogen, Taastrup, Denmark)
using NuPAGE MOPS SDS running buffer (NP0001, Invitrogen) and Fer-
mentas protein standards. Separated proteins were electrophoretically
transferred to nitrocellulose membranes (LC2000, Invitrogen), which
were stained in 1% Ponceau S red solution (P7170, Sigma-Aldrich, Copen-
hagen, Denmark), and incubated for 1 h at room temperature in blocking
buffer [5% non-fat dry milk in TBST (0.01 M Tris–HCl, 0.15 M NaCl, 0.1%
Tween 20, pH 7.4)] before incubation with primary antibodies in blocking
buffer overnight at 48C. The antibodies used were rabbit polyclonal
anti-AR (N-20) (1:200; Santa Cruz Biotechnology, CA, USA), rabbit poly-
clonal anti-AR (N-20) blocking peptide (40:1; Santa Cruz Biotechnology)
and mouse monoclonal anti-b-actin (1:5000; Sigma-Aldrich). The
membranes were washed several times in TBST, incubated for 1 h with
alkaline phosphatase-coupled secondary antibodies diluted 1:5000 in
blocking buffer, and washed in TBST prior to development with
S-bromo-D-chloro-3-indolyl phosphate/nitro blue tetrazolium solution
Androgen receptor and FSHR in human follicles 65
by guest on December 27, 2015http://molehr.oxfordjournals.org/Downloaded from
(KLP, Gaithersburg, MD, USA). The developed blots were scanned and
processed for publication in Adobe Photoshop version 6.
Immunohistochemistry
For immunohistochemistry ovarian medulla tissue containing an antral fol-
licle was dissected free and fixed in Bouin’s fixative. The tissue was pre-
pared for histology. Five micrometre paraffin sections were dewaxed,
rehydrated and washed in TBS (0.05 M Tris, pH 7.6, 0.15 M NaCl) with
0.01% Nonidet P-40 (TBS/Nonidet). Sections were incubated in 0.45%
H
2
O
2
in TBS/Nonidet for 15 min to block endogenous peroxidase
activity, and then in 10% normal goat serum in TBS/Nonidet for 30 min
at room temperature to block non-specific binding. All sections were incu-
bated overnight at 48C with the primary antibody raised against the human
AR (#SC-816, Santa Cruz Biotechnology) diluted 1:50 in 10% goat serum.
The primary antibody was detected using DakoCytomation EnVision+
DualLink System, Peroxidase (DAB+) code K4065, following the manu-
facturer’s instructions. As a negative control, sections were incubated
with Rabbit IgG (X0903 DakoCytomation) instead of the primary anti-
body. As a positive control, sections from fetal testes tissues with
known occurrence of the antigen were used.
Statistics
For evaluation of a possible association between the levels of the
measured substances, normal random effects models were used with a
random effect of women to take into account that results from the
same women may be more similar than results from different women.
AR mRNA expression was grouped into four approximately equally
sized groups (,20, 20 –45, .45 130, .130) and similarly testosterone
concentration was grouped (,45, 45–210, .210 350, .350 nmol/l).
The outcomes were log-transformed to approximate normality leading
to geometric means being calculated in each group on the original scale.
Likelihood ratio tests were conducted for equality over groups as well
as trend tests. P-value of ,0.05 was accepted as statistical significance.
Results
The quality of the extracted total RNA allowed evaluation of the ana-
lysed genes in 55 of the 64 samples. In the remaining nine samples the
concentration of the extracted total RNA was too low or not of
appropriate quality for quantification of the gene expression levels.
Table Ishows the mRNA expression levels of AR divided into four
groups of approximately equal size in relation to mRNA expression of
FSHR, LHR, CYP19 (aromatase) and AMHRII expression. AR mRNA
expression was highly significantly associated with the FSHR
expression (Table II). A significant linear trend was shown between
the mRNA expression of AR and FSHR (P,0.0001; Fig. 1). In
addition, AMHRII mRNA expression was also significantly associated
with the AR mRNA expression (Fig. 1). In contrast, LHR and
CYP19 mRNA expression did not show any significant associations
with AR mRNA expression (Table I).
The correlation between mRNA levels of AR in granulosa cells and
the corresponding follicular fluid levels of hormones showed no signifi-
cant associations (Table II).
.......................................................................................................
.............................................................................................................................................................................................
Table II AR mRNA expression in granulosa cells of small antral follicles in relation to the hormone concentration of the
corresponding follicular fluid.
Androgen receptor mRNA expression [geometric mean (95% CI)] Overall test Test for trend
<20 20–45 >45–130 >130
No. of follicular fluid 11 16 14 14
AMH (ng/ml) 399 (226; 701) 394 (243; 639) 813 (508; 1300) 525 (312; 881) 0.0967 0.1903
Inhibin-B (ng/ml) 58 (34; 99) 59 (38; 94) 62 (40; 97) 52 (32; 86) 0.9346 0.7683
Estradiol (nmol/l) 26 (10; 69) 27 (12; 61) 15 (7; 34) 22 (9; 53) 0.7381 0.5734
Progesterone (nmol/l) 245 (157; 382) 281 (192; 411) 191 (132; 276) 309 (206; 464) 0.2378 0.7244
Androstenedione (nmol/l) 1860 (1336; 2589) 2785 (2091; 3710) 2618 (1984; 3456) 2593 (1894; 3551) 0.0977 0.1904
Testosterone (nmol/l) 139 (93; 207) 189 (133; 268) 178 (126; 252) 226 (155; 329) 0.1494 0.0587
......................................................................................................
.............................................................................................................................................................................................
Table I Estimated level of mRNA expression of FSHR, CYP19, LHR and AMHRII in relation to AR mRNA expression in
granulosa cells of small antral follicles as geometric mean and 95% confidence interval, with a random effect of woman.
Androgen receptor mRNA expression [geometric mean (95% CI)] Overall test Test for trend
<20 20–45 >45–130 >130
No. of follicular fluid 11 16 14 14
FSHR 8 (3; 19) 20 (9; 42) 76 (37; 157) 107 (48; 237) ,0.0001 ,0.0001
CYP19 9 (3; 24) 10 (4; 24) 10 (4; 23) 18 (7; 47) 0.6632 0.2999
LHR 0.33 (0.14; 0.78) 0.21 (0.10; 0.43) 0.53 (0.26; 1.07) 0.76 (0.34; 1.70) 0.1011 0.0485
AMHRII 3.1 (1.6; 5.7) 10.2 (6.1; 17.1) 10.7 (6.4; 17.9) 19.6 (11.3; 33.9) 0.0003 0.0001
Test for same level in all four groups and test for trend over groups.
66 Nielsen et al.
by guest on December 27, 2015http://molehr.oxfordjournals.org/Downloaded from
The diameter of the majority of the follicles was present in a relative
narrow range of around 4–7 mm (estimated from the aspirated
volume) and the AR mRNA expression did not show any significant
correlation to the follicular volume (data not shown).
Table III shows the follicular fluid concentrations of testosterone
divided into four groups of approximately equal size in relation to
the mRNA expression of FSHR, LHR, CYP19 (aromatase) and
AMHRII expression. Intrafollicular concentrations of testosterone
were highly significantly associated with FSHR mRNA expression in
the corresponding granulosa cells, whereas LHR, CYP19 (aromatase)
and AMHRII expression showed no significant associations. As
expected, follicular fluid concentrations of testosterone showed a
highly significant association with androstenedione, but no significant
correlation to levels of inhibin-B.
Intrafollicular concentrations of androstenedione did not show
significant correlations to mRNA expression levels for any of the
evaluated substances except for the FSHR mRNA expression, which
proved to be significant (P,0.05; data not shown).
In patients in whom granulosa cells from more than one follicle were
included, the FSHR mRNA levels were mostly seen to vary as much as
between patients (Fig. 2). Within patient variation reached one order
of magnitude with the remaining variability being related to between
patient variation (Fig. 2). The other granulosa cell measured mRNAs
showed a range similar to that of FSHR.
Figure 3shows a representative result of western blots of granulosa
cells from small human antral follicles and demonstrated that the AR
protein itself was expressed. A similar result was obtained with mul-
tiple samples. Expression levels of AR were higher in granulosa cells
from antral follicles than in medulla tissue from human ovaries. As a
control an extract from undifferentiated human embryonic stem
cells showed no expression. The western blot result is supported by
immunohistochemical staining of histological sections of a human
antral follicle, which showed expression of the AR protein mainly on
the granulosa cells but also some staining on the theca cells.
Discussion
The present study comprises material from human antral follicles with
diameters from 3 to 9 mm obtained from normal ovaries. The data
showed that granulosa cell FSHR gene expression was significantly
associated with the expression of AR on the granulosa cells, and, in
addition, significantly associated with the intrafollicular concentrations
of androgens (i.e. androstenedione and testosterone). This suggests
that the collective effect of androgens acting through the AR
appears to promote gonadotrophins-responsiveness of developing
human follicles. Although the present results should be interpreted
with caution due to their correlative nature, they are in accordance
with a number of previous studies on primates and rodents.
Further, ovaries from rhesus monkeys showed that the AR gene
was most abundantly expressed on granulosa cells in the ovary and
augmented in healthy follicles. It was suggested that androgens stimu-
late early primate follicle development (Weil et al., 1998). Subsequent
Figure 1 Correlation between mRNA expression of the FSHR and
the AR and between FSHR and AMHRII in granulosa cells from
normal small human antral follicles.
.......................................................................................................
.............................................................................................................................................................................................
Table III Follicular fluid concentrations of testosterone in relation to mRNA expression of FSHR, LHR, CYP19 and
AMHRII genes in the corresponding granulosa cells and to other selected follicular fluid hormones.
Testosterone concentration (nmol/l) [geometric mean (95% CI)] Overall test Test for trend
<145 145–210 211 –350 >350
No. of follicular fluid 15 13 14 13
FSHR 24 (11; 49) 13 (6; 29) 98 (46; 211) 105 (44; 250) 0.0030 0.0052
LHR 0.5 (0.2; 1.1) 0.5 (0.2; 1.2) 0.4 (0.2; 0.8) 0.4 (0.2; 0.9) 0.8814 0.5484
CYP19 10 (4; 26) 14 (6; 37) 11 (4; 28) 11 (4; 32) 0.9465 0.9078
AMHRII 6 (3; 12) 16 (8; 31) 11 (5; 21) 9 (4; 20) 0.1393 0.4312
Androstenedione (nmol/l) 1541 (1206; 1970) 2262 (1762; 2904) 3418 (2664; 4385) 3509 (2643; 4660) ,0.0001 ,0.0001
Inhibin-B (ng/ml) 53 (33; 84) 48 (30; 78) 67 (41; 107) 84 (49; 143) 0.3081 0.1048
Androgen receptor and FSHR in human follicles 67
by guest on December 27, 2015http://molehr.oxfordjournals.org/Downloaded from
studies found that exogenously administered androgens promote
FSHR expression in vivo explaining earlier studies on the androgen-
induced enhanced estrogen biosynthesis and partly explained the
amplifying effect that androgens exert on FSH action (Weil et al.,
1999). Finally, a recent study on conditional granulosa cells and
oocyte-specific AR knockout mice saw that increasing androgen and
AR gene expression by small follicles was associated with increased
FSHR expression, which adds to the perception that androgens via
their receptors may stimulate pre-antral follicle growth, prevent
atresia and are essential for normal follicular development (Sen and
Hammes, 2010).
Taken together, the present results confirm and extends previous
studies and suggest that androgens also in humans exert important
functions in small antral follicles and indicate that proper androgen
priming of such follicles enhance FSHR expression and make them
more sensitive to FSH stimulation and subsequent growth.
Previous studies have shown that androgens enhance granulosa cells
expression of FSHR in vitro resulting in an increased aromatase
Figure 2 Concentration of FSHR mRNA in individual granulosa cell samples from small antral follicles given for each individual patient.
Figure 3 AR expression in granulosa cells from small human antral follicles. (A) Western blot analysis of AR (N-20, 110 kDa), blocking peptide
control (BP, 40×of Ab concentration) and b-actin (43 kDa) reactivity on human ovarian medulla tissue, granulosa cells from small human antral
follicles and human embryonic stemcells (hESCs). (Band C) Histological sections of human ovarian tissue showing immunohistochemical staining of
granulosa cells in antral follicles. GC, granulosa cells; BM, basal membran; TC, theca cells.
68 Nielsen et al.
by guest on December 27, 2015http://molehr.oxfordjournals.org/Downloaded from
expression and an augmented conversion of androgen to estrogen
(rat: Hillier and De Zwart, 1981,1982;Daniel and Armstrong,
1984; primate: Harlow et al., 1986). However, in rhesus monkeys
treated with large doses of exogenous androgens, testosterone was
without effect on granulosa cell expression of aromatase and circulat-
ing levels of estradiol became reduced (Weil et al., 1999). The present
study of naive follicles support the in vivo data, since we were also
unable to show an association between neither AR gene expression
and aromatase expression, nor between follicular fluid androgens
and aromatase expression. The fact that androgens and AR gene
expression appear to promote FSHR expression in human follicles
adds another regulatory element to the complex 2-cell two gonado-
trophins regulation of follicular estradiol synthesis, but does suggest
that proper FSH stimulation is a prerequisite for the androgen
action to be effective. It is unlikely that the AR directly regulates the
FSHR gene since no androgen response element on the FSHR gene
promoter has been identified. It may be that the AR or follicular
fluid androgens influence FSHR mRNA stability or may work indirectly,
perhaps by causing enhanced local production of IGF1 or inhibin-B
(Vendola et al., 1999a). The significant association between the
expression of AR and FSHR with AMHRII in the present study
showed that the AMH signalling system also might affect the inter-
action between AR and FSHR expression.
Studies in post-pubertal gilts treated with dihydrotestosterone
demonstrated a dose-dependant increase in ovulation rate (Ca
´rdenas
et al., 2002). The androgen treatment resulted in an increased amount
of FSHR mRNA in pre-ovulatory follicles and it was suggested that
the androgen effect might be caused by an up-regulation of FSHR
expression (Ca
´rdenas et al., 2002). Although an increased number
of pre-ovulatory follicles were not reported in primate studies
(Vendola et al., 1998), the results of the present study suggest an
increased FSH responsiveness of follicles that have a high expression
of AR and contain high concentrations of androgen. Collectively,
these results encourage pursuing clinical studies to evaluate whether
androgen priming of patients who show a poor or inadequate follicular
response following controlled ovarian stimulation can enhance follicu-
lar responsiveness and the number of recruitable follicles.
In fact several studies described administration of dehydroepian-
drosterone (DHEA) to patients in order to increase the systemic
levels of androgens and reported an enhanced harvest of mature
oocytes subsequent to ovarian stimulation (Barad and Gleicher,
2005,2006;So
¨nmezer et al., 2009). Whether androgens augmented
systematically by administration of DHEA or increased locally
by stimulating the ovaries own androgen production (i.e. LH or
hCG stimulation possible in combination with aromatase inhibitors
that block the conversion of androgen to estrogen), it appears to
make follicles more sensitive to FSH. To what extent an enhanced
FSH responsiveness of follicles augment the quality of the enclosed
oocyte and subsequently results in more fit embryos with a higher
implantation potential is not yet clear. Further studies are needed to
define the conditions by which androgen priming may improve clinical
results, but the present study does encourage this approach.
It is further of interest that mRNA expression of AMHRII also was
significantly associated with AR mRNA expression. In a previous study
we found that FSHR was significantly associated with AMHRII mRNA
expression (Nielsen et al., 2010). The present study is unable to
address whether AR and androgens affect AMHRII expression or
whether it occurs via the FSHR or a different signal transduction
pathway, but it does demonstrate that AMH, which occurs in concen-
trations of several hundreds ng/ml (Yding Andersen et al., 2008), and
its receptor AMHRII are likely to be involved in the complicated regu-
lation of follicular growth and development.
This study was unable to demonstrate significant correlations
between the follicular diameter and the AR mRNA expression. This
may relate to the fact that the diameters of a majority of the follicles
included had a relatively narrow range of 4 –7 mm. In addition, the
procedure by which the follicular fluid was aspirated may not have
yielded the total volume of each individual follicle fluid in all cases.
Further, the health status of the follicle may be expected to influence
the results. In order to compensate for these shortcomings future
studies should increase numbers in combination with assessment of
the follicular health status.
In conclusion, using fluid and granulosa cells from normal human fol-
licles with a diameter of 3–9 mm, the present study found a highly sig-
nificant association between the expression of AR on granulosa cells,
and the expression of FSHR, and between the follicular fluid content of
androgens and FSHR expression. These results encourage further
studies to improve results of already-employed strategies to
enhance the outcome of ovarian stimulation by performing androgen
priming prior to the administration of exogenous gonadotrophins.
Authors’ roles
M.E.N. performed all the mRNA purification and quantification and
participated in planning the study, evaluating the results and writing
the paper. I.A.R. assisted M.E.N. and participated in evaluating the
results. S.G.K. performed W.B. and participated in writing the
paper. S.C. performed W.B. and participated in writing the paper.
K.M. performed immunohistochemical staining and participated in
writing the paper. E.W.A. performed all statistical analyses and partici-
pated in writing the paper. A.G.B. participated in planning the study
and in writing the paper. C.Y.A. collected all the small antral follicles,
participated in planning the study, performed hormone analysis of the
follicular fluids, evaluated the results and wrote the paper.
Acknowledgements
The excellent technical assistance by Tiny Roed and Maria Westfall
is gratefully acknowledged. Cryopreservation of ovarian tissue is
performed in a much appreciated collaboration with the following,
Anders Nyboe Andersen, The Fertility Clinic, University Hospital of
Copenhagen, Denmark, Erik Ernst, Skejby University Hospital,
Aarhus, Denmark and Per Emil Rasmussen, Odense University
Hospital, Odense, Denmark.
Conflict of interest: none declared.
Funding
Cryopreservation of ovarian tissue would not have been possible
without support from the Danish Cancer Foundation Grant
(DP05112/R2-A41-09-S2) and the Novo Nordisk Foundation,
which is hereby acknowledged.
Androgen receptor and FSHR in human follicles 69
by guest on December 27, 2015http://molehr.oxfordjournals.org/Downloaded from
References
Barad DH, Gleicher N. Increased oocyte production after treatment with
dehydroepiandrosterone. Fertil Steril 2005;84:756.
Barad D, Gleicher N. Effect of dehydroepiandrosterone on oocyte and
embryo yields, embryo grade and cell number in IVF. Hum Reprod
2006;21:2845–2849.
Campo SM, Carson RS, Findlay JK. Distribution of specific androgen
binding sites within the ovine ovarian follicle. Mol Cell Endocrinol 1985;
39:255–265.
Ca
´rdenas H, Herrick JR, Pope WF. Increased ovulation rate in gilts treated
with dihydrotestosterone. Reproduction 2002;123:527– 533.
Christensen ST, Guerra C, Wada Y, Valentin T, Angeletti RH, Satir P,
Hamasaki T. A regulatory light chain of ciliary outer arm dynein in
Tetrahymena thermophila.J Biol Chem 2001;276:20048–20054.
Daniel SA, Armstrong DT. Site of action of androgens on
follicle-stimulating hormone-induced aromatase activity in cultured rat
granulosa cells. Endocrinology 1984;114:1975–1982.
Farhi J, Jacobs HS. Early prediction of ovarian multifollicular response
during ovulation induction in patients with polycystic ovary syndrome.
Fertil Steril 1997;67:459–462.
Harlow CR, Hillier SG, Hodges JK. Androgen modulation of
follicle-stimulating hormone-induced granulosa cell steroidogenesis in
the primate ovary. Endocrinology 1986;119:1403–1405.
Hillier SG, De Zwart FA. Evidence that granulosa cell aromatase
induction/activationby follicle-stimulating hormone is an androgen
receptor-regulated process in-vitro.Endocrinology 1981;109:1303– 1305.
Hillier SG, de Zwart FA. Androgen/antiandrogen modulation of cyclic
AMP-induced steroidogenesis during granulosa cell differentiation in
tissue culture. Mol Cell Endocrinol 1982;28:347–361.
Hillier SG, Tetsuka M, Fraser HM. Location and developmental regulation
of androgen receptor in primate ovary. Hum Reprod 1997;12:107– 111.
Horie K, Takakura K, Fujiwara H, Suginami H, Liao S, Mori T.
Immunohistochemical localization of androgen receptor in the human
ovary throughout the menstrual cycle in relation to oestrogen and
progesterone receptor expression. Hum Reprod 1992;7:184– 190.
Løssl K, Andersen AN, Loft A, Freiesleben NL, Bangsbøll S, Andersen CY.
Androgen priming using aromatase inhibitor and hCG during
early-follicular-phase GnRH antagonist down-regulation in modified
antagonist protocols. Hum Reprod 2006;21:2593– 2600.
Mason HD, Willis DS, Beard RW, Winston RM, Margara R, Franks S.
Estradiol production by granulosa cells of normal and polycystic
ovaries: relationship to menstrual cycle history and concentrations of
gonadotropins and sex steroids in follicular fluid. J Clin Endocrinol
Metab 1994;79:1355–1360.
Nielsen ME, Rasmussen IA, Fukuda M, Westergaard LG, Andersen CY.
Concentrations of anti-Mullerian hormone in fluid from small human
antral follicles show a negative correlation with CYP19 mRNA
expression in the corresponding granulosa cells. Mol Hum Reprod
2010;10:637–643.
Schmittgen TD, Livak KJ. Analyzing real-time PCR data by the comparative
CT method. Nature Protocols Online 2008;3:1101–1108.
Sen A, Hammes SR. Granulosa cell-specific androgen receptors are critical
regulators of ovarian development and function. Mol Endocrinol 2010;
24:1393–1403.
So
¨nmezer M, Ozmen B, Cil AP, Ozkavukc¸u S, Tas¸c¸i T, Olmus¸H,
Atabekog
˘lu CS. Dehydroepiandrosterone supplementation improves
ovarian response and cycle outcome in poor responders. Reprod
Biomed Online 2009;19:508–513.
Tetsuka M, Hillier SG. Differential regulation of aromatase and androgen
receptor in granulosa cells. J Steroid Biochem Mol Biol 1997;61:
233–239.
Thessaloniki ESHRE/ASRM-Sponsored PCOS Consensus Workshop
Group. Consensus on infertility treatment related to polycystic ovary
syndrome. Hum Reprod 2008;23:462– 477.
Vendola KA, Zhou J, Adesanya OO, Weil SJ, Bondy CA. Androgens
stimulate early stages of follicular growth in the primate ovary. J Clin
Invest 1998;15:2622– 2629.
Vendola K, Zhou J, Wang J, Bondy CA. Androgens promote insulin-like
growth factor-I and insulin-like growth factor-I receptor gene
expression in the primate ovary. Hum Reprod 1999a;14:2328– 2332.
Vendola K, Zhou J, Wang J, Famuyiwa OA, Bievre M, Bondy CA.
Androgens promote oocyte insulin-like growth factor I expression and
initiation of follicle development in the primate ovary. Biol Reprod
1999b;61:353–357.
Weil SJ, Vendola K, Zhou J, Adesanya OO, Wang J, Okafor J, Bondy CA.
Androgen receptor gene expression in the primate ovary: cellular
localization, regulation, and functional correlations. J Clin Endocrinol
Metab 1998;83:2479–2485.
Weil SJ, Vendola K, Zhou J, Bondy CA. Androgen and follicle-stimulating
hormone interactions in primate ovarian follicle development. J Clin
Endocrinol Metab 1999;84:2951–2956.
Yding Andersen C, Rosendahl M, Byskov AG. Concentration of
anti-Mu
¨llerian hormone and inhibin-B in relation to steroids and age in
follicular fluid from small antral human follicles. J Clin Endocrinol Metab
2008;93:2344–2349.
70 Nielsen et al.
by guest on December 27, 2015http://molehr.oxfordjournals.org/Downloaded from
... Previous studies demonstrated that T facilitates follicular recruitment [29], promotes follicular growth and development [30], and increases the expression of insulinlike growth factor 1 (IGF-1) in primate ovaries, crucial for regulating follicular development [30]. It also heightens follicular responsiveness to FSH through androgen receptors [31,32]. In vitro studies also indicate that while T depletion leads to apoptosis in granulosa cells, this process is inhibited by the administration of T [27]. ...
Article
Full-text available
Purpose This study aimed to investigate the changes in serum Anti-Müllerian Hormone (AMH) levels, sex hormone levels, follicle-stimulating hormone (FSH)/luteinizing hormone (LH) ratio in patients with celiac disease (CeD), and their correlation with clinical characteristics and nutrient levels. Methods This cross-sectional study collected clinical and biochemical data from a total of 67 females diagnosed with CeD and 67 healthy females within the reproductive age range of 18–44 years. The study was conducted at a tertiary hospital between September 2016 and January 2024. Both groups underwent comprehensive clinical and laboratory assessments. Serum levels of AMH and sex hormones were quantified using chemiluminescence immunoassay, and their associations with CeD clinical features and nutrient levels were thoroughly analyzed. Results The study included 67 patients and 67 controls with a mean age of 36.7±7.6 years. No statistically significant differences were found between the two groups in mean age, BMI, FSH, LH, E2, P levels, FSH/LH, menstrual irregularities, abortions history, parity, and gravidity (all P>0.05). However, AMH, T, FER, FA, Zn, and Se levels were significantly lower, and PRL levels were higher in the CeD group (all P<0.05). Spearman’s correlation analysis showed that AMH levels were negatively correlated with age, tTG level, disease duration, and Marsh grading (P<0.05). Conclusions This study highlights the association between impaired ovarian function in CeD patients and disease severity and nutrient levels. Early detection and intervention for ovarian function abnormalities are imperative to enhance fertility potential in CeD patients.
... [17], the highly significant of TSH and low value of T3 and T4 may be happen because of the low thyroid function this causes the pituitary gland to secrete TSH [18] and The hypothalamus at the base of the brain produces the thyronin-releasing hormone (TRH), which in turn stimulates the pituitary gland to produce thyroid stimulating hormone (TSH) and the hypothalamus produces somatostatin from the pituitary gland which has the opposite effect on the production of (TSH) and reducing its secretion [21], Symptoms associated with hypothyroidism include irregular menstrual dysfunction due to ovulation problems and prolactin hormone levels with an increased risk of PCOS [22]. and testosterone hormone was no moral difference between the patient and control It can be explained that there are no significant differences between healthy women with polycystic ovary syndrome because they have been taking treatment with metformin pills for a period of not less than three months, which indicates their response to treatment and thus reduce the difference in the level of this hormone between women infected and control [23] addition, the absence of significant differences between the two groups can be explained by a positive correlation between the mRNA levels of the androgen receptor and FSH receptors [24] [25] . ...
Article
Full-text available
This study was done by partially purification of prolidase from serum of patients with polycystic ovary syndrome by Gel filtration technique, and using sephadex G100 gel as a stationary phase. The degree of purification (15.1) fold, enzyme yield (95.5%) and specific activity (0.00176 IU/I), were carried out .Kinetics studies for the partial purified enzyme technique showed optimal concentration of substrate which was 5 mmol/l Km = 0.66ng and Vmax =0.80 mM, while optimum Temperature was (35C°) and optimum pH was (8). The molecular weight of the partial purified enzyme has been determined by gel electrophoresis method , in presence of polyacrylamide gel and sodium dodecyl sulphate (SDS_PAGE) which showed that the approximates molecular weight was (54KD),we found high level of prolactin in the patient with polycystic ovary syndrome which was(24.
... We did however document an increase in the sensibility of the granulosa cells to FSH, with an increase in the expression of the FSH receptors, therefore resulting in an increase of the steroidogenesis. 6,11,15,16 Regarding adverse effects, Rangel's 2021 study reported that the administration of 100mg of DHEA for 16 weeks is safe, as there are no observable metabolic modifications, nor any changes in the menstrual cycle, nor hyperandrogenism indicators. Our study corroborates the absence of adverse effects with the aforementioned dose for a month, suggesting this for safety. ...
... In our study, the posttreatment endometrial thickness in the intervention group exhibited a slight yet significant increase compared to the control group. This finding aligns with previous studies suggesting a correlation between optimal endometrial thickness and improved IVF outcomes [17]. Furthermore, Andreeva et al. [15] reported in their study that extended testosterone pretreatment could potentially enhance IVF treatment in patients with low ovarian response. ...
Article
Full-text available
Objective: The aim of this study was to compare the outcomes of in vitro fertilization (IVF) in patients with a poor ovarian response who used methyltestosterone, versus those using a placebo, in an infertility clinic setting. Methods: This clinical trial included 120 women who had undergone IVF with intracytoplasmic sperm injection due to poor ovarian reserve and infertility. The study took place at the Yas Infertility Center in Tehran, Iran, between January 1, 2018 and January 1, 2019. In the intervention group, 25 mg of methyltestosterone was administered daily for 2 months prior to the initiation of assisted reproductive treatment. The control group was given placebo tablets for the same duration before starting their cycle. Each group was randomly assigned 60 patients. All analyses were performed using SPSS ver. 23 (IBM Corp.). Results: The endometrial thickness in the intervention group was 7.57±1.22 mm, whereas in the control group, it was 7.11±1.02 ( p =0.028). The gonadotropin number was significantly higher in the control group (64.7±13.48 vs. 57.9±9.25, p =0.001). However, there was no significant difference between the two groups in the antral follicular count. The chemical and clinical pregnancy rates in the intervention group were 18.33% and 15% respectively, compared to 8.33% and 6.67% in the control group. The rate of definitive pregnancy was marginally higher in the intervention group (13.3% vs. 3.3%, p=0.05). Conclusion: The findings of this study suggest that pretreatment with methyltestosterone significantly increases endometrium thickness and is associated with an increase in the definitive pregnancy rate.
... All quantitative RT-qPCR reactions were performed on the QuantStudio™ 7 Flex Real-Time PCR system (Thermo Fisher Scientific), according to the supplier's protocol. GAPDH was used as the endogenous control gene for the normalization of linear and circular transcripts [32][33][34][35]. RNU6 was used as the endogenous control for miRNA gene expression data normalization. ...
Article
Full-text available
CircRNAs are a class of non-coding RNAs able to regulate gene expression at multiple levels. Their involvement in physiological processes, as well as their altered regulation in different human diseases, both tumoral and non-tumoral, is well documented. However, little is known about their involvement in female reproduction. This study aims to identify circRNAs potentially involved in reproductive women’s health. Candidate circRNAs expressed in ovary and sponging miRNAs, already known to be expressed in the ovary, were selected by a computational approach. Using real time PCR, we verified their expression and identified circPUM1 as the most interesting candidate circRNA for further analyses. We assessed the expression of circPUM1 and its linear counterpart in all the follicle compartments and, using a computational and experimental approach, identified circPUM1 direct and indirect targets, miRNAs and mRNAs, respectively, in cumulus cells. We found that both circPUM1 and its mRNA host gene are co-expressed in all the follicle compartments and proposed circPUM1 as a potential regulator of PTEN, finding a strong positive correlation between circPUM1 and PTEN mRNA. These results suggest a possible regulation of PTEN by circPUM1 in cumulus cells and point out the important role of circRNA inside the pathways related to follicle growth and oocyte maturation.
Article
Aim: To investigate the impact of letrozole cotreatment progestin-primed ovarian stimulation (PPOS) (Le PPOS) in controlled ovarian stimulation (COS) and the pregnancy outcomes in frozen-thawed embryo transfer cycles. Methods: This retrospective cohort study included women who underwent in vitro fertilization (IVF)/intracytoplasmic sperm injection (ICSI). A total of 2575 cycles were included (1675 in the Le PPOS group and 900 in the PPOS group). The primary outcome was the clinical pregnancy rates. The secondary outcome was the live birth rates. Results: In this study, propensity score matching (PSM) was performed to create a perfect match of 379 patients in each group. After matching, the numbers of oocytes retrieved, mature oocytes, fertilization, and clinical pregnancy rates were more favorable in the Le PPOS group than in the PPOS group (all p < 0.05). The multivariable analysis showed that the clinical pregnancy rate was higher in the Le PPOS than in the PPOS group (odds ratio = 1.46, 95% confidence interval: 1.05-2.04, p = 0.024) after adjusting for potentially confounding factors (age, anti-Müllerian hormone levels, antral follicular count, the type of embryo transferred, number of transferred embryos, body mass index, and follicular stimulating hormone and estradiol levels on starting day). Conclusions: This retrospective study with a limited sample size suggests that the Le PPOS protocol might be an alternative to the PPOS protocol in women undergoing COS and could lead to better pregnancy outcomes. The results should be confirmed using a formal randomized controlled trial.
Article
Full-text available
The optimal strategy for stimulation of young women with a low ovarian reserve is still a challenging issue because the physio-pathogeny of this disorder is often unknown. As androgen production by the ovary plays a crucial role in folliculogenesis, it was tempting to speculate that subtle perturbations in ovarian steroidogenesis might participate to the low responsiveness to gonadotrophins. Indeed, in vitro analysis of human luteinized granulosa cells has recently provided evidence for some enzymatic deficits in steroidogenesis and altered response to gonadotrophins. Therefore, improving androgen environment of women classified in Poseidon Group 3 should be considered. In this clinical situation, the potential benefit of androgen supplementation or stimulation of theca cells by LH-activity products are respectively discussed.
Article
Full-text available
The treatment of infertile women with polycystic ovary syndrome (PCOS) is surrounded by many controversies. This paper describes, on the basis of the currently available evidence, the consensus reached by a group of experts regarding the therapeutic challenges raised in these women. Before any intervention is initiated, preconceptional counselling should be provided emphasizing the importance of life style, especially weight reduction and exercise in overweight women, smoking and alcohol consumption. The recommended first-line treatment for ovulation induction remains the anti-estrogen clomiphene citrate (CC). Recommended second-line intervention, should CC fail to result in pregnancy, is either exogenous gonadotrophins or laparoscopic ovarian surgery (LOS). The use of exogenous gonadotrophins is associated with increased chances for multiple pregnancy and, therefore, intense monitoring of ovarian response is required. LOS alone is usually effective in < 50% of women and additional ovulation induction medication is required under those circumstances. Overall, ovulation induction (representing the CC, gonadotrophin paradigm) is reported to be highly effective with a cumulative singleton live birth rate of 72%. Recommended third-line treatment is in vitro fertilization. More patient-tailored approaches should be developed for ovulation induction based on initial screening characteristics of women with PCOS. Such approaches may result in deviation from the above mentioned first-, second- or third-line ovulation strategies in well-defined subsets of patients. Metformin use in PCOS should be restricted to women with glucose intolerance. Based on recent data available in the literature, the routine use of this drug in ovulation induction is not recommended. Insufficient evidence is currently available to recommend the clinical use of aromatase inhibitors for routine ovulation induction. Even singleton pregnancies in PCOS are associated with increased health risk for both the mother and the fetus.
Article
Androgens have well defined roles in male reproduction and prostate cancer. In contrast, other than the obligatory role of testosterone as an estradiol precursor in steroidogenesis, little is known about androgens and androgen receptor (AR) actions in the female. Interestingly, female mice with global knockout of AR expression were found to have reduced fertility with abnormal ovarian function. However, since ARs are expressed in a myriad of reproductive tissues, including the hypothalamus, pituitary, and various ovarian cells, the role of tissue-specific ARs in regulating female fertility remained unknown. To examine the importance of ovarian ARs in female reproduction, we generated granulosa cell (GC)- and oocyte-specific AR null mice by crossing AR-flox mice with MisRIIcre (granulosa cell-specific) or GDF9cre (oocyte-specific) mice. Relative to wild-type (WT) and heterozygous (AR+/-) mice, GC-specific AR-/- mice had sub-fertility and premature ovarian failure. This sub-fertility was manifested by smaller litter size and fewer litter numbers at 8-9 weeks of age, with disrupted estrous cycling and even more dramatic reduction in fertility by 24-25 weeks. Interestingly, GC-specific AR-/- mice ovulated significantly lower number of oocytes relative to AR+/- mice, possibly explaining the observed reduced litter size and numbers. Furthermore, corresponding with the reduced ovulation, the ovaries of the GC-specific AR-/- mice contained more pre-antral and atretic follicles, with fewer antral follicles and corpora lutea, relative to AR+/- mice. This accumulation of small pre-antral follicles was not due to increased recruitment of primordial follicles, as the percentage of primordial and primary follicles in GC-specific AR-/- and AR+/- mice were similar. These observations suggest that a sub-set of follicles in GC-specific AR-/- mice may be stalled in the pre-antral phase of development, thereby subjecting them to become atretic rather than develop into antral follicles, followed by ovulation and corpora lutea production. In fact, consistent with this observation that pre-antral follicles have difficulty growing and progressing through normal folliculogenesis, in-vitro growth of pre-antral follicles from GC-specific AR-/- mice was significantly slower than follicles from WT animals. Moreover, under macroscopic examination, the uteri of GC-specific AR-/- animals appeared to be normal, suggesting that the observed reproductive defects and sub-fertility are not due to changes in the uterus, but owing exclusively to lack of ARs in the GCs. In contrast to GC-specific AR-/- mice, fertility, estrous cycle, and ovarian morphology of oocyte-specific AR-/- mice were normal, although androgens no longer promoted oocyte maturation in these animals. Since follicular development and overall fertility of the oocyte-specific AR-/- mice were normal, AR expression in oocytes does not appear to be essential for reproduction. In summary, our data show that ARs in GCs, but not oocytes, regulate female fertility, perhaps by promoting pre-antral follicle growth and development, while preventing follicular atresia. In fact, nearly all the reproductive phenotypes observed in the global AR knockout mice can be explained by the lack of AR expression in GCs, suggesting that ovarian actions of androgens can have significant effects on the entire hypothalamic-pituitary-gonadal axis. (platform)
Article
Two different methods of presenting quantitative gene expression exist: absolute and relative quantification. Absolute quantification calculates the copy number of the gene usually by relating the PCR signal to a standard curve. Relative gene expression presents the data of the gene of interest relative to some calibrator or internal control gene. A widely used method to present relative gene expression is the comparative CT method also referred to as the 2−ΔΔCT method. This protocol provides an overview of the comparative CT method for quantitative gene expression studies. Also presented here are various examples to present quantitative gene expression data using this method.
Article
The physiological significance of androgens in female reproduction was unclear until female mice with global knockout of androgen receptor (AR) expression were found to have reduced fertility with abnormal ovarian function. However, because ARs are expressed in a myriad of reproductive tissues, including the hypothalamus, pituitary, and various ovarian cells, the role of tissue-specific ARs in regulating female fertility remained unknown. To examine the importance of ovarian ARs in female reproduction, we generated granulosa cell (GC)- and oocyte-specific AR-knockout (ARKO) mice by crossing AR-flox mice with MisRIIcre (GC-specific) or growth differentiation factor growth differentiation factor-9cre (oocyte-specific) mice. Relative to heterozygous and wild-type mice, GC-specific ARKO mice had premature ovarian failure and were subfertile, with longer estrous cycles and fewer ovulated oocytes. In addition, ovaries from GC-specific knockout mice contained more preantral and atretic follicles, with fewer antral follicles and corpus lutea. Finally, in vitro growth of follicles from GC-specific AR-null mice was slower than follicles from wild-type animals. In contrast to GC-specific AR-null mice, fertility, estrous cycles, and ovarian morphology of oocyte-specific ARKO mice were normal, although androgens no longer promoted oocyte maturation in these animals. Together, our data indicate that nearly all reproductive phenotypes observed in global ARKO mice can be explained by the lack of AR expression in GCs. These GC-specific ARs appear to promote preantral follicle growth and prevent follicular atresia; thus they are essential for normal follicular development and fertility.
Article
Ovaries surgically removed for fertility preservation from a total of 24 women served as a source of human small antral follicles, including the follicular fluid (FF) and the corresponding granulosa cells (GC). The FF was used to evaluate the intrafollicular concentrations of anti-Müllerian hormone (AMH), inhibin-B, estradiol, progesterone, androstenedione and testosterone. In GC mRNA expression of the AMH type II receptor (AMH-r2) was determined and correlated to the mRNA expression of CYP19 (aromatase), FSH-receptor (FSH-r) and LH-receptor (LH-r) and to the hormonal profiles of the corresponding FF. GC and FF from a total of 64 follicles (diameter of 3-9 mm) were evaluated. Concentrations of AMH in FF showed a highly significant inverse correlation with CYP19 mRNA expression in the corresponding GC and with concentrations of estradiol, progesterone and inhibin-B in the FF. However, a small subgroup of follicles exhibited high levels of AMH simultaneously with relative high levels of CYP19 mRNA. In contrast to AMH, mRNA expression of AMH-r2 was significantly positively correlated to the mRNA expression of FSH-r and CYP 19, but failed to correlate to any other measured parameters. These data confirms an intimate correlation between follicular AMH levels, AMH-r2, FSH-r expression and estradiol secretion in the developing human follicle.
Article
The effect of dehydroepiandrosterone (DHEA) supplementation on cycle outcome was assessed in patients with poor ovarian response. In total, 19 poor responder patients who were scheduled to undergo a second intracytoplasmic sperm injection (ICSI)/embryo transfer cycle were enrolled and first ICSI/embryo transfer cycles were taken as the control group. All subjects were given DHEA supplementation (25 mg t.i.d.) for at least 3 months prior to their second ICSI/embryo transfer cycle. In both cycles a fixed dose of rFSH (300 IU/day) and human menopausal gonadotrophin (HMG) (75 or 150 IU/day) along with a flexible gonadotrophin-releasing hormone (GnRH) antagonist protocol were administered. A favourable decrease was noted in mean day 3 serum oestradiol concentrations after DHEA supplementation (75.14 +/- 28.93 versus 43.07 +/- 11.77; P < 0.01). Increased number of >17 mm follicles (3 +/- 0.7 versus 1.9 +/- 1.3; P < 0.05), MII oocytes (4 +/- 1.8 versus 2.1 +/- 1.8; P < 0.05), top quality day 2 (2.2 +/- 0.8 versus 1.3 +/- 1.1; P < 0.05) and day 3 embryos (1.9 +/- 0.8 versus 0.7 +/- 0.6; P < 0.05) were achieved in DHEA-supplemented cycles. Cycle cancellation rates were reduced (5.3% versus 42.1%; P < 0.01), and the pregnancy rate per patient and clinical pregnancy rate per embryo transfer (47.4% versus 10.5%; P < 0.01 and 44.4% versus 0%; P < 0.01) were improved after DHEA supplementation. DHEA supplementation might enhance ovarian response, reduce cycle cancellation rates and increase embryo quality in poor responders.