ArticlePDF Available

Regulation of Insulin-Like Growth Factor 2 by Oocyte-Secreted Factors in Primary Human Granulosa Cells

Authors:

Abstract and Figures

Context Human granulosa cells (hGCs) produce and respond to insulin-like growth factor 2 (IGF2) but whether the oocyte participates in IGF2 regulation in humans is unknown. Objective To determine the role of oocyte-secreted factors (OSFs) such as growth differentiation factor 9 (GDF9) and bone morphogenetic protein 15 (BMP15) in IGF2 production by hGCs. Design Primary human cumulus GCs in culture. Setting University infertility center. Patients or Other Participants GCs of women undergoing in vitro fertilization. Intervention(s) Cells treated with GDF9 and BMP15 in the presence of vehicle, FSH, dbcAMP, or SMAD inhibitors. Main Outcome Measure(s) Quantification of mRNA, protein, promoter activity, and DNA methylation. Results FSH-stimulation of IGF2 (protein and mRNA) was significantly potentiated by the GDF9 and BMP15 (G+B) combination (P<0.0001) in a concentration-dependent manner showing a maximal effect at five ng/ml each. However, GDF9 or BMP15 alone or in combination (G+B) have no effect on IGF2 in the absence of FSH. FSH stimulated IGF2 promoter 3 activity, but G+B had no effect on promoter activity. G+B potentiated IGF2 stimulation by cAMP. SMAD3 inhibitors inhibited G+B enhancement of IGF2 stimulation by FSH (P<0.05) but had no effect on FSH induction. Moreover, inhibition of insulin-like growth factor receptor partially blocked G+B potentiation of FSH actions (P<0.009). Conclusions For the first time, we show that the oocyte actively participates in the regulation of IGF2 expression in hGCs, an effect that is mediated by the specific combination of G+B via SMAD2/3, which in turn target mechanisms downstream of the FSH receptor.
Content may be subject to copyright.
doi:10.1210/clinem/dgz057 J Clin Endocrinol Metab, January 2020, 105(1):327–335 https://academic.oup.com/jcem 327
Abbreviations: AMH, anti-Müllerian hormone; ANOVA, analysis of variance; BMP15,
bone morphogenetic protein 15; ELISA, enzyme-linked immunosorbent assay; FSH,
follicle-stimulating hormone; GDF9, growth differentiation factor 9; hGC, Human
granulosa cell; IGF2, insulin-like growth factor 2; IVF, in vitro fertilization; LSD, least
significant difference; OSF, oocyte-secreted factor; qPCR, quantitative real-time poly-
merase chain reaction; SEM, standard error of the mean
ISSN Print 0021-972X ISSN Online 1945-7197
Printed in USA
© Endocrine Society 2019. All rights reserved. For permissions, please e-mail: journals.
permissions@oup.com
Received 11 July 2019. Accepted 20 September 2019.
First Published Online 7 October 2019.
CLINICAL RESEARCH ARTICLE
Regulation of Insulin-Like Growth Factor 2 by Oocyte-
Secreted Factors in Primary Human GranulosaCells
Elie Hobeika,1 Marah Armouti,2 MicheleA. Fierro,1 Nichola Winston,1
Humberto Scoccia,1 AlberuniM. Zamah,3 and Carlos Stocco2
1Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology,
University of Illinois at Chicago College of Medicine, Chicago, Illinois 60612; 2Department of Physiology
and Biophysics, University of Illinois at Chicago, Chicago, Illinois 60612; and 3Division of Reproductive
Endocrinology and Infertility, Department of Obstetrics and Gynecology, the University of Chicago
Medicine, Chicago, Illinois 60637
ORCiD number: 0000-0001-7961-6681 (Carlos Stocco).
Context: Human granulosa cells (hGCs) produce and respond to insulin-like growth factor 2
(IGF2) but whether the oocyte participates in IGF2 regulation in humans is unknown.
Objective: To determine the role of oocyte-secreted factors (OSFs) such as growth
differentiation factor 9 (GDF9) and bone morphogenetic protein 15 (BMP15) in IGF2 production
by hGCs.
Design: Primary human cumulus GCs in culture.
Setting: University infertility center.
Patients or Other Participants: GCs of women undergoing in vitro fertilization.
Intervention(s): Cells treated with GDF9 and BMP15 in the presence of vehicle, follicle-
stimulating hormone (FSH), dibutyryl cyclic-AMP (dbcAMP), or mothers against decapentaplegic
homolog (SMAD) inhibitors.
Main Outcome Measure(s): Quantification of mRNA, protein, promoter activity, and DNA
methylation.
Results: FSH stimulation of IGF2 (protein and mRNA) was significantly potentiated by the GDF9
and BMP15 (G+B) combination (P<0.0001) in a concentration-dependent manner showing a
maximal effect at 5ng/mL each. However, GDF9 or BMP15 alone or in combination (G+B) have
no effect on IGF2 in the absence of FSH. FSH stimulated IGF2 promoter 3 activity, but G+B had
no effect on promoter activity. G+B potentiated IGF2 stimulation by cAMP. SMAD3 inhibitors
inhibited G+B enhancement of IGF2 stimulation by FSH (P<0.05) but had no effect on FSH
induction. Moreover, inhibition of insulin-like growth factor receptor partially blocked G+B
potentiation of FSH actions (P<0.009).
Conclusions: For the first time, we show that the oocyte actively participates in the regulation
of IGF2 expression in hGCs, an effect that is mediated by the specific combination of G+B via
SMAD2/3, which in turn target mechanisms downstream of the FSH receptor. (J Clin Endocrinol
Metab 105: 327–335, 2020)
Downloaded from https://academic.oup.com/jcem/article-abstract/105/1/327/5582040 by lib-electronic@uic.edu user on 05 June 2020
Folliculogenesis is a long process lasting several
months in humans that transforms a primordial fol-
licle into a dominant preovulatory follicle (1). The suc-
cess of folliculogenesis depends on a close interaction
between the two main components of the follicle, the
granulosa cells (GCs) and the oocyte. The active role of
the oocyte in folliculogenesis was described in the early
1970s, when it was observed that oocyte ablation leads
to impaired folliculogenesis and follicle luteinization
(2). Later, Nekola etal. (3) conrmed these ndings and
showed that GCs cultured near oocytes appeared to be
less luteinized than those cultured without oocytes. It
was not until two decades later that landmark studies
identied two oocyte-specic growth factors (OSFs),
growth differentiation factor 9 (GDF9) and bone mor-
phogenetic protein 15 (BMP15) (4, 5). The inuence of
these factors on the process of folliculogenesis is now
well accepted.
GDF9 and BMP15 have a high degree of homology
in their sequence and structure. They have similar ex-
pression patterns and functions in the ovary (6–8).
Moreover, recent evidence suggests that GDF9 and
BMP15 form heterodimers, which mediate some of
their actions (9, 10). However, the role of GDF9 and
BMP15 in the control of folliculogenesis has been elu-
cidated exclusively using animal models and cell lines.
These reports showed that GDF9 is a critical player in
the follicular development of mice and sheep, whereas
BMP15 is not essential for fertility in mice but critical
in sheep (4, 5, 8). However, GDF9 and BMP15 function
in the human ovary and in human primary ovarian cells
remains unexplored, mainly due to the lack of appro-
priate experimental approaches.
We previously validated the use of cumulus cells
obtained from patients undergoing in vitro fertiliza-
tion (IVF) as a proxy of undifferentiated GCs to study
follicle-stimulating hormone (FSH) actions in humans
(11, 12). Also, we examined the interaction between
FSH, GDF9, and BMP15 on GC function and showed
that, whereas FSH inhibits anti-Müllerian hormone
(AMH) production in primary human GCs, the com-
bination of GDF9 and BMP15 (GB) potentiates the pro-
duction of AMH (13). The interaction between FSH and
OSFs is not always antagonistic. For instance, our most
recent report shows that GDF9 and BMP15 potentiate
FSH stimulation of aromatase and estrogen production,
two hallmarks of GC differentiation (14).
Previous studies demonstrated an essential role of
the insulin-like growth factor (IGF) system on the in-
duction of aromatase and estradiol synthesis in human
GCs (11, 15, 16). Interactions between FSH and IGFs
have also been shown to upregulate the production of
estradiol and progesterone in several species such as ro-
dent (17), porcine (18), and bovine (19), beyond that
of either factor alone. However, the IGF system differs
signicantly between rodents and humans, making the
use of animal models unsuitable for studies aiming to
elucidate the regulation of the IGF system in humans.
For instance, while IGF1 is mostly expressed in mouse
GCs, IGF2 is the only IGF expressed in human GCs (11,
16, 20). We have also demonstrated that FSH inhibits
IGF1 expression in rodent GCs, whereas FSH stimu-
lates IGF2 expression in human GCs (11, 15, 16). These
ndings were conrmed in a recent report showing
that IGF2 expression in human GCs increases approxi-
mately 64-fold as follicles progress from the small an-
tral to the preovulatory stage, a process that is entirely
regulated by FSH (21). However, the mechanisms con-
trolling the expression of IGF2 in human GCs remains
unexplored. Here, we examined the interaction between
GDF9, BMP15, and FSH on the regulation of IGF2 in
human primary cumulus GCs. We tested the hypothesis
that GDF9 and BMP15 interact with FSH to stimulate
the expression of IGF2 and that this effect mediates, at
least in part, the enhancing effect of OSFs on aromatase
and estradiol production.
Materials and Methods
Human cumulus cell culture
Human cumulus cells were collected from patients under-
going IVF at the University of Illinois IVF Center under an
Institutional Review Board-exempt protocol. No patient
information was collected for reporting. After controlled
ovarian stimulation, mature follicles were aspirated from
women undergoing IVF. The cumulus–oocyte complexes were
then removed from the follicular aspirates, and the cumulus
cells were separated from the oocyte manually. The cumulus
cells from all follicles for a single patient were then pooled and
transported immediately to the laboratory where they were
dispersed by hyaluronidase digestion (80IU/μL) and then cen-
trifuged at 500× g for 2 minutes. Cells were incubated at room
temperature in red blood cell lysis buffer for 5 minutes to elim-
inate contaminating erythrocytes, centrifuged again at 500×
g for 2 minutes. Cells were suspended in 0.4mL of serum-free
and phenol red-free DMEM/F12-0.25% BSA (Sigma-Aldrich)
media supplemented with penicillin (50IU/mL), streptomycin
(50g/mL), and sodium bicarbonate (1.2g/L; Sigma-Aldrich).
Cells were then cultured on plates precoated with BD Matrigel
(BD Biosciences) at a density of 6× 104/mL. Cells were cultured
for at least 48 hours, followed by treatment with various com-
binations of hormones and signaling inhibitors. Treatments
included human recombinant FSH (Serono), GDF9 (R&B),
BMP 15 (R&B), inhibitors of SMAD2/3 (SB431542, Tocris),
SMAD 3 (SIS3; Cayman Chemical Company), or SMAD1/5/8
(LDN-193189, Selleck Chemicals), IGF1R inhibitor AEW
(NVP-AEW451) (Calbiochem), and dbcAMP (Sigma). Cells
of different individuals were not pooled together.
328 Hobeika et al. GDF9 and BMP15 Enhance FSH-Induced IGF2 Expression J Clin Endocrinol Metab, January 2020, 105(1):327–335
Downloaded from https://academic.oup.com/jcem/article-abstract/105/1/327/5582040 by lib-electronic@uic.edu user on 05 June 2020
Polymerase chain reaction
Total RNA was isolated using TRIzol Reagent (Invitrogen)
as recommended in the manufacturer’s protocol. Total RNA
(1 µg) was reverse-transcribed using anchored oligo-dT pri-
mers (Integrated DNA Technologies), and reverse transcriptase
from Moloney murine leukemia virus (Invitrogen) at 37°C for
2 hours. The resulting cDNA was diluted to a nal concentra-
tion of 10ng/µL. Quantitative real-time polymerase chain re-
action (qPCR) was performed using intron-spanning primers
specic for the detection of ribosomal protein L19 (RPL19)
and promoter 3-specic IGF2 transcripts. The number of
copies for each gene was calculated using a standard curve
made with a serial dilution of the respective cDNA (22). Once
the number of copies of the gene of interest was obtained,
the relative expression is calculated as the ratio between the
copy number of the gene of interest and the copy number of
the housekeeping gene Rpl19. Primer sequences are available
upon request.
Promoter activityassay
The IGF2p3–Luc reporter was cloned into a lentivirus
reporter vector using the method described previously (14).
Lentiviruses containing this construct were generated as de-
scribed (14). Empty plasmids were used as controls. Cells were
infected with lentivirus and after overnight incubation treated
with FSH, GDF9, BMP15, or their combination. At 48 hours,
luciferase activity was determined in 50μL of lysates as previ-
ously described (14).
Promoter 3 methylation
We examined the methylation status of the 37 CpG sites
found in the IGF2 promoter 3 using bisulte sequencing.
Cumulus cells were treated with vehicle or G+B in the pres-
ence or absence of FSH for 48 hours before genomic DNA iso-
lation. An aliquot of DNA was treated with sodium bisulte
and puried using Wizard DNA clean-up system (Promega).
To quantify CpG methylation, IGF2 promoter 3 was ampli-
ed using primers specic for bisulte-treated DNA. Then,
next-generation sequencing libraries were prepared from the
original PCR amplicon using primers that introduce Illumina
adaptors. Samples were then sequenced using Illumina
NextSeq. Athreshold of 98% conversion of no-CpG C to
uracil was set for the sample to be included in the analysis. The
sequences were aligned to the IGF2-P3 sequence and 5 mC
percentage calculated.
Enzyme-linked immunosorbentassay
Enzyme-linked immunosorbent assay (ELISA) for IGF2
protein secretion was performed on the supernatant of human
GCs treated with the different combinations of FSH, GDF9,
and BMP15 using the Quantikine® ELISA kit for Human
IGF2 (R&D Systems).
Statistical analysis
Each experiment was run at least in duplicate, and data for
continuous variables are presented as mean values±standard
error of the mean (SEM). Statistical comparisons of mean
values between groups were performed with paired t-tests,
and multiple comparisons were performed with one-way ana-
lysis of variance (ANOVA) with repeated measures followed
by Bonferroni adjustment, Fisher’s least signicant difference
(LSD) test or Tukey’s multiple comparisons test, where appro-
priate. Differences were considered to be statistically signi-
cant if P<0.05.
Results
The combination of GDF9 and BMP15 potentiates
FSH induction ofIGF2
We previously demonstrated that the combination of
GDF9 and BMP15 potentiates the stimulatory effect of
FSH on IGF2 expression (14). To further evaluate the
impact of GDF9 and BMP15 on the expression of IGF2
mRNA, primary hGCs were treated with increasing
concentrations of GDF9 and BMP15 in the presence
of 50 ng/mL of FSH. The concentration of FSH was
initially chosen based on that used in previous reports
(13, 14). As previously shown, FSH signicantly in-
creased IGF2 mRNA transcripts when compared with
nontreated cells using the t-test (P <0.05) (Fig. 1A). This
stimulatory effect of FSH on IGF2 was signicantly
augmented by the addition of the combination GDF9
and BMP15 (G+B) to the media (P<0.01). As shown in
Fig. 1A, IGF2 mRNA levels were signicantly increased
in human GCs treated with G+B at concentrations of
2.5 and 10 ng/mL in the presence of FSH compared
with other treatments and the untreated control cells.
Thus, treatment with FSH and either GDF9 or BMP15
at concentrations of 10ng/mL, or treatment with 10ng/
mL G+B in the absence of FSH did not stimulate IGF2
compared with FSH-only treated cells. To maintain con-
sistency with previous reports, a dose of 5ng/mL for
each GDF9 and BMP15 was used in the remainder of
the experiments (13, 14).
Next, we examined whether G+B was able to en-
hance the effect of increasing concentrations of FSH. As
shown in Fig. 1B, although G+B cotreatment enhanced
the effect of FSH, increasing concentrations of FSH did
not further increase IGF2 expression, suggesting that
the stimulation of IGF2 appears to be maximal in the
presence of FSH and G+B.
GDF9 and BMP15 potentiate FSH stimulation of
IGF2 proteinlevels
Next, we examined whether the effect of G+B on
IGF2 mRNA levels translates into an increase in IGF2
protein. IGF2 was measured in the supernatant of cul-
tures treated as indicated above using ELISA. In con-
cordance with ndings at the mRNA levels, cotreatment
of FSH with G+B potentiated the stimulatory effect of
FSH on IGF2 protein expression (Fig. 2). IGF2 concen-
tration in the supernatant of cells treated with FSH and
G+B cotreatment was increased 1.9-fold compared with
doi:10.1210/clinem/dgz057 https://academic.oup.com/jcem 329
Downloaded from https://academic.oup.com/jcem/article-abstract/105/1/327/5582040 by lib-electronic@uic.edu user on 05 June 2020
the control group (66.67±5.5 vs 33.99±0.6 pg/mL,
P<0.01) and 1.5-fold compared with the group treated
with FSH alone (66.67 ± 5.5 vs 43.15 ± 0.4 pg/mL,
P < 0.05). A paired t-test comparing the FSH-treated
group with the untreated control group showed a stat-
istically signicant increase in IGF2 protein production
(43.15±0.4 vs 33.99±0.6pg/mL, P<0.01). Taken to-
gether, these ndings indicate a robust positive effect of
the GDF9 and BMP15 combination on the stimulatory
effect of FSH on IGF2 protein production.
GDF9 and BMP15 potentiate cAMP stimulation
ofIGF2
Next, we determined whether G+B interacts with
cAMP, the primary second messenger of the FSH re-
ceptor, to stimulate IGF2. As shown in Fig. 3, treat-
ment with dbcAMP (a cAMP analog) alone stimulated
IGF2 mRNA levels compared with the control group
(P<0.05). However, IGF2 mRNA expression was sig-
nicantly higher in cells treated with GDF9, BMP15,
and dbcAMP than in cells treated with dbcAMP alone
Figure 1. The combination of FSH, GDF9, and BMP15 (GB)
potentiate FSH-induced IGF2 expression in a dose-dependent manner.
A. Primary human cumulus cells were treated for 48 hours with
vehicle (C), GDF9 (G: 10ng/mL), BMP15 (B: 10ng/mL), GDF9 and
BMP15 combined (GB: 0.6, 2.5, and 10ng/mL for each of G and
B), in the presence or absence of follicle-stimulating hormone (FSH)
(F: 50ng/mL). IGF2 mRNA levels were determined by quantitative
real-time polymerase chain reaction (qPCR) and expressed relative
to Rpl19. Columns represent the mean±SEM (standard error of the
mean). Columns with different letters differ significantly by one-way
analysis of variance (ANOVA) analysis with Fisher’s least significant
difference (LSD) test, a–b P<0.05, a–c P<0.01, b–c P<0.05,
n=9. Control and FSH groups were compared by t test; *P<0.05,
n=9. B. Primary human cumulus cells were treated for 48 hours
with vehicle (C), GDF9 (G: 5ng/mL), BMP15 (B: 5ng/mL), in the
presence or absence of FSH (F: 5, 25, 100ng/mL). IGF2 mRNA levels
were determined by qPCR and expressed relative to Rpl19. Columns
represent the mean±SEM. Columns with different letters differ
significantly by one-way ANOVA analysis with Fisher’s LSD test, a–b
P<0.05, a–c P<0.01, b–c P<0.05, n=5.
Figure 2. The combination of GDF9 and BMP15 potentiates follicle-
stimulating hormone (FSH)-induced IGF2 expression and protein
synthesis. IGF2 protein levels were quantified by enzyme-linked
immunosorbent assay. Columns represent the mean±standard error
of the mean, columns with different letters differ significantly by
one-way analysis of variance analysis with repeated measures and
Bonferroni correction a–b P<0.001. One-way paired t-test was used
to measure the difference between C and F, (**) for P<0.01, n=3.
Figure 3. The potentiation of FSH actions by GDF9 and BMP15 (GB)
occurs downstream of cAMP. Primary human cumulus cells were
treated for 48 hours with vehicle (C) or the combination of GDF9
and BMP15 (GB: 5ng/mL) in the presence or absence of dbcAMP
(cAMP: 2mM) and the absence of FSH.mRNA levels were determined
by quantitative real-time polymerase chain reaction and expressed
relative to Rpl19. One-way ANOVA analysis with repeated measures
and Bonferroni correction. Columns represent the mean±SEM,
columns with different letters differ significantly, a–b P<0.05, a–c
P<0.01, b–c P<0.05, n=5.
330 Hobeika et al. GDF9 and BMP15 Enhance FSH-Induced IGF2 Expression J Clin Endocrinol Metab, January 2020, 105(1):327–335
Downloaded from https://academic.oup.com/jcem/article-abstract/105/1/327/5582040 by lib-electronic@uic.edu user on 05 June 2020
(P<0.05) or the control (C) group (P<0.01). Thus, as
in the case of FSH, the stimulatory effect of dbcAMP
was potentiated by the presence of G+B in the media,
suggesting that the crosstalk between FSH and G+B oc-
curs downstream of the FSH receptor.
GDF9 and BMP15 increase IGF2 transcripts
derived from the promoter 3 but have no effect
on promoter activity
IGF2 gene transcription is driven by four different
promoters (23). The activity of each promoter gener-
ates IGF2 mRNA with promoter-specic untranslated
exons upstream of the protein-coding exons 7 to 9 that
are common to all transcripts. We have demonstrated
that IGF2 expression in human GCs is controlled by
promoters 3 and 4, of which only promoter 3 is regu-
lated by FSH (16). For this reason, the results listed
in Figs. 1 and 3 were performed using primers that
amplify transcripts derived from promoter 3. To fur-
ther examine the regulation of promoter 3 by G+B, we
cloned 511bp of the IGF2 promoter 3 into a lentiviral
reporter plasmid (IGF2p3–LUC). To determine whether
G+B regulates IGF2p3–LUC, cumulus cells were plated
in 48-well plates and immediately infected with the
lentivirus for 24 hours. At this time, cells were treated
with vehicle, FSH, G+B, or FSH plus G+B. Luciferase
activity was quantied 48 hours after the addition of
FSH. FSH treatment alone increased the activity of
the IGF2p3–LUC construct compared with the control
group (P<0.05) (Fig. 4A). In contrast to the ndings
observed on promoter 3 transcript levels, treatment
with FSH plus G+B did not increase the activity of
the IGF2p3–LUC construct above the levels observed
with FSH alone. This lack of agreement between the
observed regulation of promoter 3 transcripts and the
promoter 3 luciferase reporter by G+B suggests alterna-
tive mechanisms by which GDF9 and BMP15 regulate
IGF2 expression.
IGF2 mRNA upregulation in cancer cells correlates
with demethylation of CpG islands in the genomic re-
gion corresponding to promoter 3, which contains
37 CpG islands (24, 25). Therefore, we examined the
methylation status of all CpG sites in this genomic re-
gion using bisulte sequencing. Cumulus cells were
treated with vehicle or G+B in the presence or absence
of FSH for 48 hours before genomic DNA isolation
and bisulte sequencing. The results demonstrated that
treatment with G+B in the presence or absence of FSH
had no effect on the percentage of methylation of the
IGF2 promoter 3 (data not shown), suggesting that pro-
moter 3 methylation is unlikely to mediate the effects
of G+B.
To examine other possible mechanisms for the regula-
tion of IGF2 by G+B, we examined the expression of the
H19 gene. H19 and IGF2 form a reciprocally imprinted
cluster (IGF2/H19) located on human chromosome 11
(26). It is well known that IGF2 and H19 expression
is inversely regulated via a differentially methylated
region upstream of the H19 gene (27). Therefore, we
quantied the mRNA levels of H19 in cells treated with
vehicle or G+B in the presence or absence of FSH. The
results demonstrated that treatment with G+B and FSH
strongly stimulated the expression of H19 reaching
levels that were signicantly higher than those found in
cells treated with FSH or GB alone (Fig. 4B).
Figure 4. Effect of GDF9 and BMP15 (GB) on IGF2 promoter 3
activity. A. Cells were infected with lentivirus carrying the IGF2p3
LUC reporter and after overnight incubation treated for 48 hours
with vehicle (C), follicle-stimulating hormone (FSH), FSH+GB, or
GB. Luciferase activity was quantified and expressed relative to the
control. Columns represent the mean±SEM, columns with different
letters differ significantly, n=4. B. H19 transcripts were measured in
cumulus cells treated with increasing doses of G+B and FSH (50ng/
mL) for 48 hours. Expression of H19 was determined by quantitative
real-time polymerase chain reaction and expressed relative to
Rpl19. Columns represent the mean±standard error of the mean.
Columns with different letters differ significantly by one-way analysis
of variance analysis followed by Tukey’s multiple comparisons test
(n=5).
doi:10.1210/clinem/dgz057 https://academic.oup.com/jcem 331
Downloaded from https://academic.oup.com/jcem/article-abstract/105/1/327/5582040 by lib-electronic@uic.edu user on 05 June 2020
GDF9 and BMP15 regulate IGF2 via
SMAD signaling
GDF9 and BMP15 activate SMAD2/3 and
SMAD1/5/8 signaling pathways, respectively (28–30).
To investigate the mechanism of G+B-induced IGF2 ex-
pression in the presence of FSH, we used inhibitors of
SMAD2/3 (SB431542), SMAD3 (SIS3), or SMAD1/5/8
(LDN-193189). The concentration of SMAD inhibi-
tors used was based on previous publications in mouse
and humans GCs (10, 30). The inhibitors were added
to the media 1 hour before the addition of FSH or FSH
plus GB. Forty-eight hours later, cells were harvested
for IGF2 mRNA determination. The results show that
inhibition of SMAD1/5/8 had no signicant effects on
the induction of IGF2 mRNA levels by FSH plus G+B
(Fig. 5). However, the potentiation of IGF2 expression
by G+B in the presence of FSH was inhibited by the add-
ition of SB431542 and SIS3 (P=0.018 and P=0.007,
respectively).
IGF1R activity is necessary for GDF9 and BMP15
potentiation of IGF2 expression
In human GCs, we previously reported that FSH
stimulation of IGF2 requires IGF1R activity (16). To
determine whether the stimulatory effect of G+B on
IGF2 also requires the IGF1R, human GCs were treated
with NVP-AEW451, a known inhibitor of IGF1R ac-
tivity, at a concentration of 1μM for one hour before
the addition of FSH, G+B, or both to the media. IGF2
mRNA levels were quantied 48 hours after the add-
ition of FSH. The potentiation of FSH plus G+B on
IGF2 mRNA was signicantly decreased by the pres-
ence of NVP-AEW451 in the media when compared
with cells treated with FSH plus G+B alone (P= 0.02)
(Fig. 6). However, G+B was still able to potentiate the
effect of FSH on IGF2 mRNA levels in the presence of
NVP-AEW451(P=0.008, ANOVA).
Discussion
We have shown for the rst time that GDF9 and
BMP15, acting in coordination with FSH, maximally
stimulate IGF2 in primary human GCs. The enhancing
effect of GDF9 and BMP15 occurs downstream of
the FSH receptor and is inhibited by SMAD2/3 and
SMAD3 pathway inhibitors. Only the combination of
GDF9 and BMP15 enhances IGF2 expression. These
ndings support the hypothesis that GDF9 and BMP15
form heterodimers (9, 10). However, GDF9:BMP15
heterodimers have not been detected in either animal
or human models. On the other hand, a recent report
suggested that monomers of BMP15 and GDF9 acti-
vate cell-surface receptors initiating a synergistic ef-
fect without the need for heterodimer formation (31).
Therefore, additional experiments are needed to deter-
mine the mechanism of the collaborative effect between
GDF9 and BMP15 on the regulation ofGCs.
Figure 5. GDF9 and BMP15 regulate IGF2 via SMAD signaling.
Primary human cumulus cells were treated for 48 hours with vehicle
(C) or the combination of GDF9 and BMP15 (GB: 5ng/mL) in the
presence or absence of FSH (50ng/mL), SMAD2/3 inhibitor SB431542
(SB: 1µM), SMAD3 inhibitor SIS3 (SIS: 5µM), or SMAD1/5/8 inhibitor
LDN-193189 (LDN: 100nM). IGF2 mRNA levels were determined by
qPCR and expressed relative to Rpl19. t-Test was used to evaluate
the difference between mRNA expression in the F+GB with and
without the different SMAD inhibitors. Columns represent the
mean±standard error of the mean, columns with different letters
differ significantly, a–b P<0.05, a–c P<0.01, b–c P<0.05, n=8.
Figure 6. The combination of GDF9 and BMP15 potentiates FSH
stimulation of IGF2 mRNA partially through IGF1R signaling. Primary
human cumulus cells were treated for 48 hours, as described in
Fig. 2A in the presence or absence of the IGF1R inhibitor NVP-
AEW451(1μM). mRNA levels were determined by qPCR and
expressed relative to Rpl19. One-way ANOVA analysis with repeated
measures and Bonferroni correction indicated a global P=0.008
for the group. Columns represent the mean±SEM, columns with
different letters differ significantly. At-test revealed that the addition
of NVP-AEW451inhibits GDF9 and BMP15 potentiation of IGF2
mRNA (**P=0.02), n=4.
332 Hobeika et al. GDF9 and BMP15 Enhance FSH-Induced IGF2 Expression J Clin Endocrinol Metab, January 2020, 105(1):327–335
Downloaded from https://academic.oup.com/jcem/article-abstract/105/1/327/5582040 by lib-electronic@uic.edu user on 05 June 2020
The use of human cumulus GCs collected from cu-
mulus–oocyte complexes isolated from women under-
going IVF has been adopted by other groups and us
to investigate human GC biology (11, 13–16, 32). We
demonstrated that cumulus cells have distinct properties
compared with mural cells such as lack of luteinization
and the ability to respond to FSH and IGFs (12). The use
of human cells is highly relevant because of the specic
characteristics of the IGF system in the human ovary
that are not conserved in other species (see earlier) (11,
16, 20).
IGF2 plays a central role in the differentiation of
human GCs. Our group was the rst to show that FSH
enhances IGF2 mRNA expression in human GCs (16)
and showed that IGF2 is necessary for FSH-induced
upregulation of steroidogenic enzymes such as Cyp19a1,
Cyp11a1, and Star (11). Together, these ndings and the
current report support the hypothesis that the IGF2
system is also involved in the selection of the dominant
follicle. Thus, considering that IGF2 is vital for GC
proliferation and that OSFs increase IGF2 expression,
it is reasonable to propose that a healthy oocyte will
secrete higher quantities of GDF9 and BMP15, hence
sensitizing GCs to FSH. An oocyte producing high levels
of GDF9 and BMP15 will enhance the survival of its
surrounding GCs in the presence of low levels of FSH,
leading to its selection from the cohort.
We proved previously and conrmed in this report
that FSH stimulates IGF2 expression by increasing pro-
moter 3-derived transcripts in a v-akt murine thymoma
viral oncogene homolog 1 (AKT) activation dependent
manner (16). However, the molecular mechanisms in-
volved in the transcriptional activation of IGF2 in hu-
mans are unknown. We show here for the rst time that
FSH increases IGF2 transcription using a reporter vector
in which luciferase activity is controlled by 511bp of
the genomic region of human P3 promoter. The mech-
anisms and factors participating in the stimulation of
promoter 3 activity by FSH remain to be determined. Of
note, in silico analysis of the promoter showed the pres-
ence of binding sites for specicity protein 1 (SP1), early
growth response protein 1 (ERG1), activation protein 2
(AP2), andCCAAT enhancer binding protein beta (C/
EBPb). However, this analysis revealed the absence of
binding sites for cAMP/CREB, which mediates FSH ac-
tions, suggesting an atypical regulation of IGF2 expres-
sion by FSH in humanGCs.
Inhibition of SMAD2/3 activation abolishes IGF2
stimulation by G+B, suggesting a major role of SMAD2/3
in IGF2 regulation. Upon phosphorylation by type 1 re-
ceptor kinase, Smads form heteromeric complexes with
co-Smads and translocate into the nucleus to activate
gene transcription. Additionally, a well-documented
mechanism of R-Smad regulation is through phosphor-
ylation of the linker region found between the MH1 and
MH2 domains (33), which profoundly affects SMAD
signaling. Among the kinases targeting this region is
AKT. We have previously demonstrated that AKT is es-
sential for the induction of IGF2 by FSH (16). Thus,
in human GCs, the activation of SMAD by G+B could
be further enhanced by FSH via activation of AKT.
We speculate that this mechanism could play a central
role in mediating the crosstalk between the signaling
activated by oocyte-secreted factors and FSH. Future
studies will be needed to test this hypothesis.
How GDF9 and BMP15 regulate the expression of
P3 transcripts is also unclear. Our ndings suggest that
combined these factors enhance promoter 3 IGF2 tran-
script expression in the presence of FSH. Strikingly, our
promoter studies showed that GDF9 and BMP15 do not
enhance the stimulation of the P3-Luc reporter by FSH.
Thus, GDF9 and BMP15 may regulate the steady-state
levels of IGF2 by affecting gene transcription indirectly,
or by regulating the stability of the IGF2 mRNA. For
the rst of these possibilities, we considered two pos-
sible mechanisms: (1) changes occur in the methyla-
tion status of promoter 3, or (2) methylation changes
occur upstream of the H19 gene (27), which is known
to upregulate IGF2 and reduce H19 expression. Our re-
sults suggest that the methylation of promoter 3 is not
affected by GDF9+BMP15 treatment. Moreover, IGF2
and H19 are regulated similarly by GDF9 and BMP15,
indicating that methylation on the imprinting-control
region (ICR) region upstream of the H19 gene is most
probably not affected by these factors. Therefore, the
evidence points to the idea that GDF9 and BMP15 are
not involved in the regulation of IGF2/H19 methylation.
However, further studies will be required to evaluate
this possibilityfully.
Changes in mRNA stability may also play a role in
the regulation of IGF2 mRNA by GDF9 and BMP15.
For instance, it will be of great interest to determine if
GDF9 and BMP15 regulate the expression of mRNA
binding proteins known as IGF2 mRNA-binding pro-
teins (IMPs). IMPs have been shown to participate in
the post-transcriptional regulation of IGF2 (34). Are-
cent report demonstrated that the expression of all
IMP isoforms is relatively constant during human fol-
licular development (21). This report also showed a
64-fold increase in IGF2 expression from small antral
to preovulatory follicle maturation (21). Moreover, we
observed previously that inhibition of IGF1 receptor ac-
tivity abolishes IGF2 induction by FSH (16). However,
the lack of IGF1R activity does not prevent the
doi:10.1210/clinem/dgz057 https://academic.oup.com/jcem 333
Downloaded from https://academic.oup.com/jcem/article-abstract/105/1/327/5582040 by lib-electronic@uic.edu user on 05 June 2020
enhancement of IGF2 expression by GDF9 and BMP15.
Taken together, our ndings and previous reports sug-
gest that IGF2 regulation in human GCs may involve
several interdependent mechanisms, which agrees with
our hypothesis that IGF2 plays a central role in follicle
maturation and selection in humans.
A limitation of our study includes the under-represen-
tation of women with diminished ovarian reserve or
low oocyte yield at the time of oocyte retrieval, which
would yield a suboptimal number of GCs. Thus, the
possibility of a variable response of GCs from patients
with different etiologies of infertility cannot be ruled
out. However, the strength of the study is that GCs from
different patients were not pooled together, suggesting
that the mechanisms activated by OSFs on the regula-
tion of human GCs are conserved despite human subject
variability.
In conclusion, GDF9 and BMP15 play a signicant
role in follicle development. In addition to their part-
nership with FSH in the regulation of aromatase, we
present evidence of their synergism in the stimulation of
IGF2. This in itself sensitizes the follicle to FSH-induced
aromatase expression, which is of utmost importance
in follicular growth in humans. This report adds to
our understanding of the unique role that OSFs play in
folliculogenesis. Furthermore, these ndings can be used
in efforts to develop new strategies to improve in vitro
maturation of follicles.
Acknowledgments
Financial Support: NIH grant R56HD086054 (C.S.).
Additional Information
Correspondence: Carlos Stocco, 835 S Wolcott Ave, Chicago,
Illinois, 60612. E-mail: costocco@uic.edu.
Disclosure Summary: The authors declare that there is no
conict of interest that could be perceived as prejudicing the
impartiality of the research reported.
References
1. EricksonGF, ShimasakiS. The physiology of folliculogenesis: the
role of novel growth factors. Fertil Steril. 2001;76(5):943–949.
2. el-Fouly MA, Cook B, Nekola M, Nalbandov AV. Role
of the ovum in follicular luteinization. Endocrinology.
1970;87(2):286–293.
3. NekolaMV, NalbandovAV. Morphological changes of rat fol-
licular cells as inuenced by oocytes. Biol Reprod. 1971;4(2):
154–160.
4. DongJ, AlbertiniDF, NishimoriK, KumarTR, LuN, MatzukMM.
Growth differentiation factor-9 is required during early ovarian
folliculogenesis. Nature. 1996;383(6600):531–535.
5. GallowaySM, McNattyKP, CambridgeLM, etal. Mutations in
an oocyte-derived growth factor gene (BMP15) cause increased
ovulation rate and infertility in a dosage-sensitive manner. Nat
Genet. 2000;25(3):279–283.
6. McIntosh CJ, Lun S, Lawrence S, Western AH, McNatty KP,
Juengel JL. The proregion of mouse BMP15 regulates the co-
operative interactions of BMP15 and GDF9. Biol Reprod.
2008;79(5):889–896.
7. PauliniF, MeloEO. The role of oocyte-secreted factors GDF9 and
BMP15 in follicular development and oogenesis. Reprod Domest
Anim. 2011;46(2):354–361.
8. Yan C, Wang P, DeMayoJ, etal. Synergistic roles of bone mor-
phogenetic protein 15 and growth differentiation factor 9 in
ovarian function. Mol Endocrinol. 2001;15(6):854–866.
9. MottersheadDG, SugimuraS, Al-MusawiSL, etal. Cumulin, an
oocyte-secreted heterodimer of the transforming growth factor-β
family, is a potent activator of granulosa cells and improves oo-
cyte quality. J Biol Chem. 2015;290(39):24007–24020.
10. PengJ, LiQ, WigglesworthK, etal. Reply to Mottershead etal.:
GDF9:BMP15 heterodimers are potent regulators of ovarian
functions. Proc Natl Acad Sci U S A. 2013;110(25):E2258.
11. BaumgartenSC, ConvissarSM, FierroMA, WinstonNJ, ScocciaB,
StoccoC. IGF1R signaling is necessary for FSH-induced activa-
tion of AKT and differentiation of human Cumulus granulosa
cells. J Clin Endocrinol Metab. 2014;99(8):2995–3004.
12. StoccoC, BaumgartenSC, ArmoutiM, etal. Genome-wide inter-
actions between FSH and insulin-like growth factors in the regu-
lation of human granulosa cell differentiation. Hum Reprod.
2017;32(4):905–914.
13. Convissar S, Armouti M, Fierro MA, Winston NJ, Scoccia H,
Zamah AM, Stocco C. Regulation of AMH by oocyte-specic
growth factors in human primary cumulus cells. Reproduction.
2017;154(6):745–753.
14. HobeikaE, ArmoutiM, KalaH, etal. Oocyte-secreted factors synergize
with FSH to promote aromatase expression in primary human cu-
mulus cells. J Clin Endocrinol Metab. 2019;104(5):1667–1676.
15. Zhou P, BaumgartenSC, WuY, etal. IGF-I signaling is essential
for FSH stimulation of AKT and steroidogenic genes in granulosa
cells. Mol Endocrinol. 2013;27(3):511–523.
16. Baumgarten SC, Convissar SM, Zamah AM, Fierro MA,
WinstonNJ, ScocciaB, StoccoC. FSH regulates IGF-2 expression
in human granulosa cells in an AKT-dependent manner. J Clin
Endocrinol Metab. 2015;100(8):E1046–E1055.
17. AdashiEY, Resnick CE, BrodieAM, SvobodaME, VanWykJJ.
Somatomedin-C-mediated potentiation of follicle-stimulating
hormone-induced aromatase activity of cultured rat granulosa
cells. Endocrinology. 1985;117(6):2313–2320.
18. Maruo T, Hayashi M, Matsuo H, Ueda Y, Morikawa H,
Mochizuki M. Comparison of the facilitative roles of insulin
and insulin-like growth factor Iin the functional differentiation
of granulosa cells: in vitro studies with the porcine model. Acta
Endocrinol (Copenh). 1988;117(2):230–240.
19. Spicer LJ, Chamberlain CS, Maciel SM. Inuence of
gonadotropins on insulin- and insulin-like growth factor-I (IGF-
I)-induced steroid production by bovine granulosa cells. Domest
Anim Endocrinol. 2002;22(4):237–254.
20. HernandezER, RobertsCT Jr, LeRoithD, AdashiEY. Rat ovarian
insulin-like growth factor I(IGF-I) gene expression is granulosa
cell-selective: 5’-untranslated mRNA variant representation and
hormonal regulation. Endocrinology. 1989;125(1):572–574.
21. Bøtkjær JA, Pors SE, Petersen TS, et al. Transcription prole
of the insulin-like growth factor signaling pathway during
human ovarian follicular development. J Assist Reprod Genet.
2019;36(5):889–903.
22. StoccoC. In vivo and in vitro inhibition of cyp19 gene expression
by prostaglandin F2alpha in murine luteal cells: implication of
GATA-4. Endocrinology. 2004;145(11):4957–4966.
23. Sussenbach JS, Steenbergh PH, Jansen E, et al. Structural and
regulatory aspects of the human genes encoding IGF-I and -II.
Adv Exp Med Biol. 1991;293:1–14.
334 Hobeika et al. GDF9 and BMP15 Enhance FSH-Induced IGF2 Expression J Clin Endocrinol Metab, January 2020, 105(1):327–335
Downloaded from https://academic.oup.com/jcem/article-abstract/105/1/327/5582040 by lib-electronic@uic.edu user on 05 June 2020
24. Raizis AM, Eccles MR, Reeve AE. Structural analysis of the
human insulin-like growth factor-II P3 promoter. Biochem J.
1993;289 (Pt 1):133–139.
25. Eriksson T, FriskT, GraySG, etal. Methylation changes in the
human IGF2 p3 promoter parallel IGF2 expression in the pri-
mary tumor, established cell line, and xenograft of a human
hepatoblastoma. Exp Cell Res. 2001;270(1):88–95.
26. ReikW, WalterJ. Genomic imprinting: parental inuence on the
genome. Nat Rev Genet. 2001;2(1):21–32.
27. Davis TL, Yang GJ, McCarrey JR, Bartolomei MS. The H19
methylation imprint is erased and re-established differentially on
the parental alleles during male germ cell development. Hum Mol
Genet. 2000;9(19):2885–2894.
28. MooreRK, OtsukaF, ShimasakiS. Molecular basis of bone mor-
phogenetic protein-15 signaling in granulosa cells. J Biol Chem.
2003;278(1):304–310.
29. Mazerbourg S, Klein C, Roh J, et al. Growth differentiation
factor-9 signaling is mediated by the type I receptor, activin
receptor-like kinase 5. Mol Endocrinol. 2004;18(3):653–665.
30. Liu C, Yuan B, Chen H, et al. Effects of MiR-375-BMPR2 as
a key factor downstream of BMP15/GDF9 on the Smad1/5/8
and Smad2/3 signaling pathways. Cell Physiol Biochem.
2018;46(1):213–225.
31. HeathDA, PitmanJL, McNattyKP. Molecular forms of ruminant
BMP15 and GDF9 and putative interactions with receptors.
Reproduction. 2017;154(4):521–534.
32. Pogrmic-MajkicK, SamardzijaD, Stojkov-MimicN, etal. Atrazine
suppresses FSH-induced steroidogenesis and LH-dependent
expression of ovulatory genes through PDE-cAMP signaling
pathway in human cumulus granulosa cells. Mol Cell Endocrinol.
2018;461:79–88.
33. Kamato D, Burch ML, Piva TJ, et al. Transforming growth
factor-β signalling: role and consequences of Smad linker
region phosphorylation. Cell Signal. 2013;25(10):2017–
2024.
34. HammerNA, HansenTv, ByskovAG, etal. Expression of IGF-II
mRNA-binding proteins (IMPs) in gonads and testicular cancer.
Reproduction. 2005;130(2):203–212.
doi:10.1210/clinem/dgz057 https://academic.oup.com/jcem 335
Downloaded from https://academic.oup.com/jcem/article-abstract/105/1/327/5582040 by lib-electronic@uic.edu user on 05 June 2020
... Furthermore, GDF9 is a factor secreted by oocytes, which can promote the development of primordial follicles and regulate the quality and developmental ability of eggs [63,64]. GDF9 mutation can affect ewe fertility by affecting follicles and oocytes [85]. ...
Article
Full-text available
Reproductive traits hold considerable economic importance in pig breeding and production. However, candidate genes underpinning the reproductive traits are still poorly identified. In the present study, we executed a genome-wide association study (GWAS) and runs of homozygosity (ROH) analysis using the PorcineSNP50 BeadChip array for 585 Yorkshire pigs. Results from the GWAS identified two genome-wide significant and eighteen suggestive significant single nucleotide polymorphisms (SNPs) associated with seven reproductive traits. Furthermore, we identified candidate genes, including ELMO1, AOAH, INSIG2, NUP205, LYPLAL1, RPL34, LIPH, RNF7, GRK7, ETV5, FYN, and SLC30A5, which were chosen due to adjoining significant SNPs and their functions in immunity, fertilization, embryonic development, and sperm quality. Several genes were found in ROH islands associated with spermatozoa, development of the fetus, mature eggs, and litter size, including INSL6, TAF4B, E2F7, RTL1, CDKN1C, and GDF9. This study will provide insight into the genetic basis for pig reproductive traits, facilitating reproduction improvement using the marker-based selection methods.
... OSFs secreted by oocytes stimulate granulosa cell proliferation, a process that requires the involvement of TGF-β, GDF9-I type I receptors (ALK4/5/6), and BMP-II receptors on the cell membrane and the activation of the SMAD2/3 or SMAD1/5/8 pathways (Dragovic et al., 2007;Hobeika et al., 2019;Hobeika et al., 2020). OSFs, especially GDF-9, have long been regarded as the main determinants of granulosa cell differentiation (Elvin et al., 1999;Su et al., 2004). ...
Article
Full-text available
During folliculogenesis, the oocyte and surrounding cumulus cells form an ensemble called the cumulus-oocyte complex (COC). Due to their interdependence, research on the COC has been a hot issue in the past few decades. A growing body of literature has revealed that intercellular communication is critical in determining oocyte quality and ovulation. This review provides an update on the current knowledge of COC intercellular communication, morphology, and functions. Transzonal projections (TZPs) and gap junctions are the most described structures of the COC. They provide basic metabolic and nutrient support, and abundant molecules for signaling pathways and regulations. Oocyte-secreted factors (OSFs) such as growth differentiation factor 9 and bone morphogenetic protein 15 have been linked with follicular homeostasis, suggesting that the communications are bidirectional. Using advanced techniques, new evidence has highlighted the existence of other structures that participate in intercellular communication. Extracellular vesicles can carry transcripts and signaling molecules. Microvilli on the oocyte can induce the formation of TZPs and secrete OSFs. Cell membrane fusion between the oocyte and cumulus cells can lead to sharing of cytoplasm, in a way making the COC a true whole. These findings give us new insights into related reproductive diseases like polycystic ovary syndrome and primary ovarian insufficiency and how to improve the outcomes of assisted reproduction.
... We have previously reported that FSH actions require the input of the IGF system Hobeika et al., 2020;Baumgarten et al., 2017;Baumgarten et al., 2015;Baumgarten et al., 2014;Baumgarten et al., 2014). This evidence led us to examine whether the FSH-induced stimulation of GATA4 phosphorylation is also affected by the activation of the IGF1R. ...
Article
Preovulatory granulosa cell (GC) differentiation is essential for the maturation and release of oocytes from the ovary. We have previously demonstrated that follicle-stimulating hormone (FSH) and insulin-like growth factors (IGFs) closely interact to control GC function. Similarly, we showed that GATA4 mediates FSH actions and it is required for preovulatory follicle formation. This report aimed to determine in vivo the effect of FSH on GATA4 phosphorylation and to investigate whether FSH and IGF1 interact to regulate GATA4 activity. In rat ovaries, treatment with equine chorionic gonadotropin (eCG) increased the phosphorylation of GATA4, which was confined to the nucleus of GCs. Using primary rat GCs, we observed that GATA4 phosphorylation at serine 105 increases the transcriptional activity of this transcription factor. Like FSH, IGF1 stimulated GATA4 phosphorylation at serine 105. Interestingly, GATA4 phosphorylation was significantly higher in cells cotreated with FSH and IGF1 when compared to FSH or IGF1 alone, suggesting that IGF1 augments the effects of FSH on GATA4. It was also found that the enhancing effect of IGF1 requires AKT activity and is mimicked by the inhibition of glycogen synthase kinase-3 β (GSK3β), suggesting that AKT inhibition of GSK3β may play a role in the regulation of GATA4 phosphorylation. The data support an important role of the IGF1/AKT/GSK3β signaling pathway in the regulation of GATA4 transcriptional activity and provide new insights into the mechanisms by which FSH and IGF1 regulate GC differentiation. Our findings suggest that GATA4 transcriptional activation may, at least partially, mediate AKT actions in GCs.
... A number of recent studies have shown that GDF9 and BMP15 modulate FSH-induced secretion of a number of granulosa and cumulus cell-secreted proteins and hormones including other TGF-β members (26,(64)(65)(66)(67), which suggests that the oocyte itself is actively engaged in the control of follicle development. It is therefore noticeable that the associations between BMP15 and the concentrations of other TGF-β members show positive associations in the non-PCO group that turn into negative associations in the PCO group. ...
Article
Full-text available
Context The oocyte-secreted factors GDF9 and BMP15 play essential roles in follicle development and oocyte maturation, and aberrant regulation might contribute to the pathogenesis of PCOS. Objective Are there measurable differences in concentrations of GDF9, BMP15, and the GDF9/BMP15 heterodimer in small antral follicle fluids from women with and without polycystic ovaries (PCO)? Design & Setting Follicle fluids (n=356) were collected from 4-11 mm follicles in unstimulated ovaries of 87 women undergoing ovarian tissue cryopreservation for fertility preservation. Patients Twenty-seven women with PCO were identified and 60 women without PCO-like characteristics (non-PCO women) were matched according to age and follicle size. Main outcome measures Intrafollicular concentrations of GDF9, BMP15, GDF9/BMP15 heterodimer, AMH, inhibin-A and -B, total inhibin, activin-B and -AB, and follistatin were measured using ELISA-assays. Results The detectability of GDF9, BMP15, and the GDF9/BMP15 heterodimer were 100%, 94.4%, and 91.5% respectively, and concentrations were significantly negatively correlated with increasing follicle size (P<0.0001). GDF9 was significantly higher in women with PCO (PCO: 4230±189 pg/ml (mean±SEM), n=188; non-PCO: 3498±199 pg/ml, n=168; P<0.03), whereas BMP15 was lower in women with PCO (PCO: 431±40 pg/ml, n=125; non-PCO: 573±55 pg/ml, n=109; P=0.10), leading to a significantly higher GDF9:BMP15 ratio in women with PCO (P<0.01). Significant positive associations between BMP15 and AMH, activins, and inhibins in non-PCO women switched to negative associations in women with PCO. Conclusions Intrafollicular concentrations of GDF9 and BMP15 varied inversely in women with PCO reflecting an aberrant endocrine environment. An increased GDF9:BMP15 ratio may be a new biomarker for PCO.
... The media was replenished every 24 h. To mimic the effect of LH and cAMP in vivo, we used hCG (10 IU/ml; LIVZON, China) and dibutyryl-cAMP (db-cAMP, 1 mM; HY-B0764A, MedChemExpress, USA) respectively to stimulate cells and further cultured for stated hours in vitro according to previous studies (21). KGN cell line was cultured in DMEM/F12 medium as mentioned above. ...
Article
Full-text available
An association between endometriosis and luteinized unruptured follicle syndrome (LUFs) has long been identified. Although inactivating mutation of luteinizing hormone/choriogonadotropin receptor (LHGCR) results in LUFs, whether LHCGR contributes to promoting LUFs in endometriosis remains elusive. To investigate the effect of LHCGR signaling in the development of endometriosis-associated LUFs and dissect the underlying mechanism in vivo mouse endometriosis model was established to measure the effect on ovarian folliculogenesis. In vitro cultures of primary human GCs collected from patients undergoing in vitro fertilization were performed and treated with human chorionic gonadotropin (hCG), dibutyryl cyclic-AMP (db-cAMP), LHCGR or CCAAT/enhancer binding protein-α (C/EBPα) small interfering RNA to identify the potential mechanisms. KGN cell line was used to investigate the mechanistic features of transcriptional regulation. Results showed an increased incidence of LUFs was observed in mice with endometriosis. The expression of LHCGR was decreased in the GCs of endometriosis mice. In in vitro cell models, LHCGR signaling increased the expression of C/EBPα and cyclooxygenase-2(COX-2), while inhibiting C/EBPα mitigated the induced COX-2 expression. Mechanically, C/EBPα bounded to the promoter region of COX-2 and increased the transcriptional activity under the stimulation of hCG or db-cAMP. Taken together, this study demonstrated that the LHCGR signaling was reduced in GCs of endometriosis and resulted in a decrease in gonadotropin-induced COX-2 expression. Our study might provide new insights into the dysfunction of GCs in endometriosis.
... The growth and development of follicles from the primordial resting phase to growing follicles are under the control of pituitary gonadotropin and local ovarian factors such as GDF-9, and oocyte-specific factors such as BMP15 (Hobeika et al., 2020). It was demonstrated that GDF-9 and BMP15 are expressed in the human oocytes during early follicular development (Vatanparast et al., 2020). ...
Article
Full-text available
Research question : Can the low-temperature transport time of removed human ovarian tissue be prolonged until cryopreservation? Design Fresh ovarian cortex from nine premenopausal patients was either slow-programmed frozen immediately or stored at 4℃ for 24h or 48h before slow-freezing. The fresh and frozen-thawed biopsies were evaluated by follicle counting via calcein-staining, histologic analyses via HE, and apoptosis via TUNEL. The fresh cortex was assessed by ROS and TAC assay for oxidative stress detection. The frozen-thawed cortex biopsies were also evaluated by qPCR for mRNA expression of BCL-2, BAX, TNFa, HIF-1a, BMP15, and GDF9, western-blot for detection of BCL-2, BMP15, GDF9, and CASPASE3. The frozen-thawed cortex was cultured in-vitro for four-days, AMH and glucose were assessed in the supernatant, and ROS and TAC assay detected cortex's oxidative stress. Results :In the fresh cortex, no significant differences between the three groups. In the frozen-thawed cortex, no significant differences between the three groups regarding follicle viability, TUNEL, mRNA expression of TNFa, HIF-1a, and BMP15. GDF9 mRNA and BAX/BCL-2 were lower and higher at 48h than at 0h, respectively. However, the protein expression of BCL-2, CASPASE3, GDF9, and BMP15 were not different. In the cultured cortex, ROS, TAC, and glucose uptake were not different in the three groups. Conclusion : Ovarian tissue transportation was validated for up to 24h in the practical procedure. This study showed that 4-8℃ transportation for 24h or 48h does not seem to damage the ovarian tissue. However, ovarian tissue transportation above 48h still needs to be further studied for conclusions to be made.
Article
Full-text available
Background The last phase of folliculogenesis is driven by follicle‐stimulating hormone (FSH) and locally produced insulin‐like growth factors (IGFs), both essential for forming preovulatory follicles. Methods This review discusses the molecular crosstalk of the FSH and IGF signaling pathways in regulating follicular granulosa cells (GCs) during the antral‐to‐preovulatory phase. Main findings IGFs were considered co‐gonadotropins since they amplify FSH actions in GCs. However, this view is not compatible with data showing that FSH requires IGFs to stimulate GCs, that FSH renders GCs sensitive to IGFs, and that FSH signaling interacts with factors downstream of AKT to stimulate GCs. New evidence suggests that FSH and IGF signaling pathways intersect at several levels to regulate gene expression and GC function. Conclusion FSH and locally produced IGFs form a positive feedback loop essential for preovulatory follicle formation in all species. Understanding the mechanisms by which FSH and IGFs interact to control GC function will help design new interventions to optimize follicle maturation, perfect treatment of ovulatory defects, improve in vitro fertilization, and develop new contraceptive approaches.
Article
The cargo carried by extracellular vesicles (EVs) plays an important physiological role in their corresponding target organs or target tissue cells. Extracellular vesicles are classified into large extracellular vesicles (LEVs) and small extracellular vesicles (SEVs) according to their diameters. Since different subtypes contain different contents, their roles are also different. In this study, the morphology and size of LEVs were analyzed by transmission electron microscopy and nanoparticle size, and the marker proteins of LEVs (CD63, GP96, TSG101, ALB) were identified by western blot, and high-purity LEVs were obtained. Through the uptake of extracellular vesicles by purified ovarian granulosa cells and the determination of granulosa cell viability, cell apoptosis, and steroid hormone production, the result indicated that LEVs significantly enhanced cell viability (P < 0.05), reduced the rate of granulosa cell apoptosis (P < 0.05). Meanwhile, LEVs promoted the secretion of estradiol in granulosa cells (P < 0.05). This study provides a reference for the in-depth study of the function of follicular fluid extracellular vesicle subtypes and the research on the regulation of extracellular vesicles on follicle and oocyte development.
Article
Full-text available
Secreted frizzled-related protein-4 (SFRP4) belongs to a family of soluble ovarian-expressed proteins that participate in female reproduction, particularly in rodents. In humans, SFRP4 is highly expressed in cumulus cells. However, the mechanisms that stimulate SFRP4 in cumulus cells have not been examined. We hypothesise that oocyte-secreted factors such as growth differentiation factor 9 (GDF9) and bone morphogenetic protein 15 (BMP15) are involved in the regulation of SFRP4. Human cumulus cells were collected from patients undergoing fertility treatments and treated with GDF9 or BMP15 or their combination in the presence of follicle-stimulating hormone (FSH) or vehicle. FSH treatment significantly decreased SFRP4 mRNA levels when compared with nontreated cells. However, SFRP4 mRNA levels were increased significantly by GDF9 plus BMP15 in a concentration-dependent manner in the presence or absence of FSH. The combination of GDF9 plus BMP15 also increased SFRP4 protein levels and decreased the activity of the β-catenin/TCF-responsive promoter significantly. GDF9 plus BMP15 inhibited steroidogenic acute regulatory protein and luteinising hormone/choriogonadotrophin (LH/hCG) receptor stimulation by FSH, while treatment with SFRP4 blocked the stimulatory effect of FSH on these genes. The evidence demonstrates that GDF9 and BMP15 act in coordination to stimulate SFRP4 expression and suggests that SFRP4 mediates the anti-luteinising effects of the oocyte in human cumulus cells.
Article
Granulosa cells (GCs) play important roles in the regulation of ovarian functions, and in vitro culture is a relevant model for the study of steroidogenesis in ovarian follicles. Thus, growth factors secreted by the oocyte, like Growth and Differentiation Factor 9 (GDF9) and Bone Morphogenetic Protein 15 (BMP15), play an important part in the luteinization of granulosa cells. The aim of this work was to express GDF9 and BMP15 genes in bovine GCs in vitro and evaluate their effects on the luteinization process. Samples of culture medium and GCs transfected with GDF9 and BMP15 were obtained for 21 consecutive days to analyze the steroidogenic hormones’ concentration (progesterone (P4) and estradiol (E2)) and the expression of STAR, GDF9, BMP15 and their respective receptors (ALK5/TGFBR1, ALK6/BMPR1B and BMPR2). The results demonstrated an inhibitory effect of GDF9 and BMPF15 on P4 secretion in bovine GCs cultured in vitro. Moreover, our study demonstrated the entire expression of their respective receptors (TGFBR1, BMPR1B and BMPR2) and the inhibition of the steroidogenic marker, STAR gene. This work sheds light on a novel biological function of BMP15 and GDF9 in bovine GCs physiology, which could elucidate a non‐described biological role for GDF9 and BMP15 in bovine granulosa cells’ metabolism.
Article
Full-text available
Purpose The IGF signaling cascade exerts important regulatory functions in human ovarian folliculogenesis. The scope of this study was to evaluate the transcription profile of insulin-like growth factor (IGF) genes during human ovarian follicle development and to analyze follicle fluid levels of key IGF proteins. Methods Gene expression profiling was performed with microarray gene analysis. The analysis was assessed from ovarian follicles and granulosa cells (GCs) obtained from isolated stage-specific human ovarian follicles, including preantral follicles, small antral follicles, and preovulatory follicles. Numerous genes involved in the IGF signaling pathway was evaluated and key genes were validated by qPCR from GCs. Protein levels of various IGF components of human follicular fluid (FF) were measured by ELISA and time-resolved immunofluorometric assays (TRIFMA). Results The gene expression levels of PAPPA, IGF2, IGF receptors and intracellular IGF-activated genes increased with increasing follicle size. This was especially prominent in the late preovulatory stage where IGF2 expression peaked. Protein levels of intact IGF binding protein-4 decreased significantly in FF from large preovulatory follicles compared with small antral follicles concomitant with higher protein levels of PAPP-A. The IGF modulators IGF-2 receptor, IGFBPs, stanniocalcins, and IGF-2 mRNA binding proteins were all observed to be expressed in the different follicle stages. Conclusions This study confirms and highlights the importance of PAPP-A regulating bioactive IGF levels throughout folliculogenesis and especially for the high rate of granulosa cell proliferation and expression of key ovarian hormones important in the last part of the follicular phase of the menstrual cycle.
Article
Full-text available
Context The role of growth differentiation factor 9 (GDF9) and bone morphogenetic protein 15 (BMP15) on aromatase regulation is poorly understood in humans. Objective Determine GDF9 and BMP15 effects on follicle-stimulating hormone (FSH) stimulation of estradiol production in primary human cumulus granulosa cells (GCs). We hypothesized that the combination of GDF9 and BMP15 potentiates FSH-induced aromatase expression. Design Primary human cumulus GCs in culture. Setting University infertility center. Patients or Other Participants GCs of 60 women undergoing in vitro fertilization were collected. Intervention(s) Cells were treated with GDF9 and/or BMP15 (GB) in the presence or absence of FSH, dbcAMP, or SMAD inhibitors. Main Outcome Measure(s) Promoter activity, mRNA, protein levels and phosphorylation, and estradiol levels were quantified. Results FSH and GB treatment increased CYP19A1 promoter activity, mRNA, and protein levels as well as estradiol when compared to cells treated with FSH only. GB treatment potentiated cAMP stimulation of aromatase and IGF2 stimulation by FSH. GB effects were inhibited by SMAD3 inhibitors and by IGF1 receptor inhibitors. GB, but not FSH, stimulate SMAD3 phosphorylation. Conclusion The combination of GDF9 and BMP15 potently stimulates the effect of FSH and cAMP on CYP19a1 promoter activity and mRNA/protein levels. These effects translate into an increase in estradiol production. This potentiation seems to occur through activation of the SMAD2/3 and SMAD3 signaling pathway and involves, at least in part, the effect of the IGF system.
Article
Full-text available
Background/aims: Bone morphogenetic protein 15 (BMP15) and growth differentiation factor 9 (GDF9), which are secreted by oocytes, are important regulators of follicular growth and development and ovarian function. These two factors can regulate the proliferation and apoptosis of cumulus cells via modulation of the Smad signaling pathway. Studies have shown that BMP15 and GDF9 can affect the level of miR-375, whereas the target gene of miR-375 is BMPR2, the type II receptor of BMP15 and GDF9. However, whether or how the BMP15/ GDF9-miR-375-BMPR2 pathway affects the proliferation and apoptosis of bovine cumulus cells through regulation of the Smad signaling pathway remains unclear. Methods: In this study, cumulus cells were first obtained from cumulus-oocyte complexes (COCs). Appropriate concentrations of BMP15 and GDF9 were added during the in vitro culture process. Cell Counting Kit-8 (CCK-8) analyses and flow cytometry were used to determine the effects of BMP15/GDF9 on bovine cumulus cells proliferation and apoptosis. Subsequently, miR-375 mimics, miR-375 inhibitor and BMPR2 siRNA were synthesized and used for transfection experiments. Western Blot analysis was used to detect changes before and after transfection in the expression levels of the BMP15/GDF9 type I receptors ALK4, ALK5 and ALK6; the phosphorylation levels of Smad2/3 and Smad1/5/8, which are key signaling pathway proteins downstream of BMP15/GDF9; the expression levels of PTX3, HAS2 and PTGS2, which are key genes involved in cumulus cells proliferation; and Bcl2/Bax, which are genes involved in apoptosis. Results: The addition of 100 ng/mL BMP15 or 200 ng/mL GDF9 or the combined addition of 50 ng/mL BMP15 and 100 ng/mL GDF9 effectively inhibited bovine cumulus cell apoptosis and promoted cell proliferation. BMP15/GDF9 negatively regulated miR-375 expression and positively regulated BMPR2 expression. High levels of miR-375 and inhibition of BMPR2 resulted in increased expression of ALK4 and decreased expression of PTX3, HAS2 and PTGS2, whereas miR-375 inhibition resulted in the opposite results. BMP15 and GDF9 significantly activated the levels of p-Smad2/3 and p-Smad1/5/8, whereas miR-375 inhibited the levels of p-Smad2/3 and p-Smad1/5/8 by negatively regulating BMPR2 and also led to apoptosis. Conclusion: BMP15 and GDF9 have synergistic effects and can act through miR-375 to affect the expression levels of type I receptor ALK4 and type II receptor BMPR2 and the activation of Smad signaling pathway, which subsequently affected the proliferation, spread and apoptosis of cumulus cells.
Article
Full-text available
Study question: Is the genome-wide response of human cumulus cells to FSH and insulin-like growth factors (IGFs) comparable to the response observed in undifferentiated granulosa cells (GCs)? Summary answer: FSH actions in human cumulus cells mimic those observed in preantral undifferentiated GCs from laboratory animals, and approximately half of the regulated genes are dependent on the simultaneous activation of the IGF1 receptor (IGF1R). What is known already: Animal studies have shown that FSH and the IGFs system are required for follicle growth and maturation. In humans, IGF levels in the follicular fluid correlate with patients' responses to IVF protocols. The main targets of FSH and IGFs in the ovary are the GCs; however, the genomic mechanisms involved in the response of GCs to these hormones are unknown. Study design, size, duration: Human cumulus cells isolated from IVF patients were cultured for 48 h in serum-free media in the presence of vehicle, FSH, IGF1R inhibitor or their combination. Participants/materials, setting, methods: Discarded cumulus cells were donated to research by reproductive-aged women undergoing IVF due to non-ovarian etiologies of infertility at a university-affiliated clinic. The effect of FSH and/or IGF1R inhibition on cumulus cell function was evaluated using Affymetrix microarrays, quantitative PCR, western blot, promoter assays and hormone level measurements. Main results and the role of chance: The findings demonstrate that human cumulus cells from IVF patients respond to FSH with the expression of genes known to be markers of the preantral to preovulatory differentiation of GCs. These results also demonstrate that ~50% of FSH-regulated genes require IGF1R activity and suggest that several aspects of follicle growth are coordinately regulated by FSH and IGFs in humans. This novel approach will allow for future mechanistic and molecular studies on the regulation of human follicle maturation. Large scale data: Data set can be accessed at Gene Expression Omnibus number GSE86427. Limitations, reasons for caution: Experiments were performed using primary human cumulus cells. This may not represent the response of intact follicles. Wider implications of the findings: Understanding the mechanisms involved in the regulation of GC differentiation by FSH and IGF in humans will contribute to improving treatments for infertility. Study funding/competing interest(s): The project was financed by the National Instituted of Health grant number R56HD086054 and R01HD057110 (C.S.). The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript. We have no competing interests to declare.
Article
Full-text available
Growth differentiation factor 9 (GDF9) and bone morphogenetic protein 15 (BMP15) are oocyte-specific growth factors with central roles in mammalian reproduction, regulating species-specific fecundity, ovarian follicular somatic cell differentiation and oocyte quality. In the human, GDF9 is produced in a latent form, the mechanism of activation being an open question. Here, we produced a range of recombinant GDF9 and BMP15 variants, examined their in silico and physical interactions, and their effects on ovarian granulosa cells (GC) and oocytes. We found that the potent synergistic actions of GDF9 and BMP15 on GC can be attributed to the formation of a heterodimer, which we have termed cumulin. Structural modelling of cumulin revealed a dimerization interface identical to homodimeric GDF9 and BMP15, indicating likely formation of a stable complex. This was confirmed by generation of recombinant heterodimeric complexes of pro/mature domains (pro-cumulin) and covalent mature domains (cumulin). Both pro-cumulin and cumulin exhibited highly potent bioactivity on GC, activating both SMAD2/3 and SMAD1/5/8 signaling pathways, and promoting proliferation and expression of a set of genes associated with oocyte-regulated GC differentiation. Cumulin was more potent than pro-cumulin, pro-GDF9, pro-BMP15 or the two combined on GC. However, on cumulus-oocyte complexes, pro-cumulin was more effective than all other growth factors at notably improving oocyte quality as assessed by subsequent day 7 embryo development. Our results support a model of activation for human GDF9 dependant on cumulin formation through heterodimerization with BMP15. Oocyte-secreted cumulin is likely to be a central regulator of fertility in mono-ovular mammals. Copyright © 2015, The American Society for Biochemistry and Molecular Biology.
Article
The regulation of AMH production by follicular cells is poorly understood. The purpose of this study was to determine the role of the oocyte-secreted factors, growth differentiation factor 9 (GDF9) and bone morphogenetic protein 15 (BMP15), on AMH production in primary human cumulus cells. Cumulus cells from IVF patients were cultured with a combination of GDF9, BMP15, recombinant FSH, and specific signaling inhibitors. Stimulation with GDF9 or BMP15 separately had no significant effect on AMH mRNA levels. In contrast, simultaneous stimulation with GDF9 and BMP15 (G+B) resulted in a significant increase in AMH mRNA expression. Increasing concentration of G+B (0.6, 2.5, 5 and 10 ng/ml) stimulated AMH in a dose-dependent manner, showing a maximal effect at 5 ng/ml. Western blot analyses revealed an average 16-fold increase in AMH protein levels in cells treated with G+B when compared to controls. FSH co-treatment decreased the stimulation of AMH expression by G+B. The stimulatory effect of G+B on the expression of AMH was significantly decreased by inhibitors of the SMAD2/3 signaling pathway. These findings show for the first time that AMH production is regulated by oocyte-secreted factors in primary human cumulus cells. Moreover, our novel findings establish that the combination of GDF9 + BMP15 potently stimulates AMH expression.
Article
Atrazine (ATR) alters female reproductive functions in different animal species. Here, we analyzed whether ATR disturbs steroidogenic and ovulatory processes in hormone-stimulated human cumulus granulosa cells and mechanism of its action. Results showed that treatment of human cumulus granulosa cells with 20 μM ATR for 48 h resulted in lower FSH-stimulated estradiol and progesterone production. ATR reduced mRNA levels of aromatase (CYP19A1), steroidogenic acute regulatory protein (STAR) and luteinizing hormone/choriogonadotropin receptor (LHCGR). Addition of hCG 48 h after FSH and ATR treatment did not trigger maximal expression of the ovulatory genes amphiregulin (AREG) and epiregulin (EREG). Mechanistic experiments showed that ATR activated cPDE and decreased cAMP level. Addition of total PDE and specific PDE4 inhibitors, IBMX and rolipram, prevented ATR's action on CYP19A1 and STAR mRNA expression in FSH-stimulated human cumulus granulosa cells. This study suggests that ATR alters steroidogenesis and ovulatory process in human cumulus granulosa cells jeopardizing female reproduction.
Article
Bone morphogenetic factor 15 (BMP15) and growth differentiation factor 9 (GDF9) are oocyte-secreted factors with demonstrable effects on ovarian follicular development and ovulation-rate. However, the molecular forms of BMP15 and GDF9 produced by oocytes remains unclear. The aims herein, using Western blotting (WB) procedures with specific monoclonal antibodies (mabs), were to identify the molecular forms of BMP15 and GDF9 synthesised and secreted by isolated ovine (o) and bovine (b) oocytes in vitro. The mabs were known to recognise the biological forms of BMP15 or GDF9 since they had previously been shown to inhibit their bioactivities in vitro and in vivo. Using recombinant variants of oBMP15 and oGDF9, including a cysteine mutant form of oBMP15 (S356C) and a human (h) BMP15:GDF9 heterodimer (cumulin), it was established that the mabs were able to identify monomeric, dimeric, promature and higher molecular weight forms of BMP15 and GDF9 and cumulin (GDF9 mab only). After using non-reducing, reducing and reducing + cross-linking conditions, the major oocyte-secreted forms of o and b BMP15 and GDF9 were the cleaved and uncleaved monomeric forms of the promature proteins. There was no evidence for dimeric or heterodimeric forms of either mature BMP15 or GDF9. From in silico modelling studies using TGFB, Activin or BMP crystal templates, and both present and previously published data, a model is proposed to illustrate how the monomeric forms of BMP15 and GDF9 may interact with the Type II and Type I, BMP cell-surface receptors to initiate the synergistic actions of these growth factors.
Article
IGF2 is highly expressed in the granulosa cells of human dominant ovarian follicles; however, little is known about the regulation of the IGF2 gene or the interaction of IGF2 and FSH during follicle development. To examine the mechanisms involved in the regulation of the IGF2 gene by FSH and the interplay between FSH and IGF2 during granulosa cell differentiation. Design, Setting, Patients, and Interventions: Cumulus granulosa cells were separated from cumulus-oocyte-complexes isolated from the follicular aspirates of IVF patients and cultured for in vitro studies. Protein and mRNA levels of IGF2 and CYP19A1 (aromatase) were quantified using Western blot and quantitative real-time PCR. IGF2 promoter-specific activation was determined by the amplification of alternative exons by PCR. Cell proliferation was assessed after treatment with FSH and/or IGF2. FSH significantly enhanced IGF2 expression after 8 hours of treatment and at low doses (1 ng/ml). Reciprocally, IGF2 synergized with FSH to increase cell proliferation and the expression of CYP19A1. When IGF2 activity was blocked, FSH was no longer able to stimulate CYP19A1 expression. Determination of IGF2 promoter usage in human cumulus cells showed that the IGF2 gene is driven by promoters P3 and P4. However, FSH exclusively increased P3 promoter derived transcripts. Moreover, the FSH-induced stimulation of P3-driven IGF2 transcripts was blocked by co-treatment with inhibitors of AKT or IGF1R. The inhibitory effect of the IGF1R inhibitor on FSH-induced IGF2 mRNA accumulation was reversed by overexpression of a constitutively active AKT construct. FSH is a potent enhancer of IGF2 expression in human granulosa cells. In return, IGF2 activation of the IGF1R and AKT is required for FSH to stimulate CYP19A1 expression and proliferation of granulosa cells. These findings suggest a positive loop interaction between FSH and IGF2 that is critical for human granulosa cell proliferation and differentiation.
Article
Context: FSH is routinely administered to in vitro fertilization patients to induce follicle maturation. During this process, granulosa cells differentiate and acquire specific functional characteristics that are required to coordinate ovulation and oocyte maturation. Objective: The objective of the study was to gain insight into the molecular mechanisms regulating human granulosa cell differentiation. Design, Setting, Patients, and Interventions: Cumulus and mural granulosa cells were isolated from the follicular aspirates of in vitro fertilization patients and analyzed immediately or cultured in serum-free media in the presence of FSH, IGFs, or an inhibitor of type I IGF receptor (IGF1R) activity. Main outcome: We quantified the mRNA and protein levels of steroidogenic enzymes, components of the IGF system, and gonadotropin receptors; measured 17β-estradiol levels; and examined the activation of intracellular signaling pathways to assess the granulosa cell differentiation as well as the FSH and IGF actions in both cumulus and mural cells. Results: In freshly isolated cells, LH receptor (Lhr) and steroidogenic acute regulator (Star) were expressed at lower levels in cumulus than mural cells, whereas FSH receptor (Fshr) and anti-Müllerian hormone (Amh) were expressed at higher levels in cumulus than mural cells. In vitro, the expression of Igf2, the differentiation markers Lhr, Star, and Cyp19a1 (aromatase) as well as 17β-estradiol production remained low in untreated cumulus cells but increased significantly after FSH treatment. Strikingly, this stimulatory effect of FSH was abolished by the inhibition of IGF1R activity. FSH-induced activation of v-akt murine thymoma viral oncogene homolog 3 (AKT) required IGF1R activity, and overexpression of constitutively active AKT rescued the induction of differentiation markers and 17β-estradiol production by FSH in the presence of the IGF1R inhibitor. Conclusions: The cumulus cell response to FSH resembles the differentiation of preantral to preovulatory granulosa cells. This differentiation program requires IGF1R activity and subsequent AKT activation.