ArticlePDF Available

Expression and effect of resistin on bovine and rat granulosa cell steroidogenesis and proliferation

Authors:

Abstract and Figures

Resistin, initially identified in adipose tissue and macrophages, was implicated in insulin resistance. Recently, its mRNA was found in hypothalamo-pituitary axis and rat testis, leading us to hypothesize that resistin may be expressed in ovary. In this study, we determined in rats and cows 1) the characterization of resistin in ovary by RT-PCR, immunoblotting, and immunohistochemistry and 2) the effects of recombinant resistin (10, 100, 333, and 667 ng/ml) ± IGF1 (76 ng/ml) on steroidogenesis, proliferation, and signaling pathways of granulosa cells (GC) measured by enzyme immunoassay, [(3)H]thymidine incorporation, and immunoblotting respectively. We observed that resistin mRNA and protein were present in several bovine and rat ovarian cells. Nevertheless, only bovine GC abundantly expressed resistin mRNA and protein. Resistin treatment decreased basal but not IGF1-induced progesterone (P<0.05; whatever the dose) and estradiol (P<0.005; for 10 and 333 ng/ml) production by bovine GC. In rats, resistin (10 ng/ml) increased basal and IGF1-induced progesterone secretion (P<0.0001), without effect on estradiol release. We found no effect of resistin on rat GC proliferation. Conversely, in cows, resistin increased basal proliferation (P<0.0001; for 100-667 ng/ml) and decreased IGF1-induced proliferation of GC (P<0.0001; for 10-333 ng/ml) associated with a decrease in cyclin D2 protein level (P<0.0001). Finally, resistin stimulated AKT and p38-MAPK phosphorylation in both species, ERK1/2-MAPK phosphorylation in rats and had the opposite effect on the AMPK pathway (P<0.05). In conclusion, our results show that resistin is expressed in rat and bovine ovaries. Furthermore, it can modulate GC functions in basal state or in response to IGF1 in vitro.
Content may be subject to copyright.
REPRODUCTION
RESEARCH
Expression and effect of resistin on bovine and rat granulosa cell
steroidogenesis and proliferation
Virginie Maillard, Pascal Froment, Christelle Rame
´, Svetlana Uzbekova, Se
´bastien Elis
and Joe
¨lle Dupont
Unite
´de Physiologie de la Reproduction et des Comportements, UMR85, Equipe Me
´tabolisme et Reproduction,
Institut National de la Recherche Agronomique, F-37 380 Nouzilly, France
Correspondence should be addressed to J Dupont; Email: jdupont@tours.inra.fr
Abstract
Resistin, initially identified in adipose tissue and macrophages, was implicated in insulin resistance. Recently, its mRNA was found in
hypothalamo–pituitary axis and rat testis, leading us to hypothesize that resistin may be expressed in ovary. In this study, we determined
in rats and cows 1) the characterization of resistin in ovary by RT-PCR, immunoblotting, and immunohistochemistry and 2) the effects of
recombinant resistin (10, 100, 333, and 667 ng/ml)GIGF1 (76 ng/ml) on steroidogenesis, proliferation, and signaling pathways of
granulosa cells (GC) measured by enzyme immunoassay, [
3
H]thymidine incorporation, and immunoblotting respectively. We observed
that resistin mRNA and protein were present in several bovine and rat ovarian cells. Nevertheless, only bovine GC abundantly expressed
resistin mRNA and protein. Resistin treatment decreased basal but not IGF1-induced progesterone (P!0.05; whatever the dose) and
estradiol (P!0.005; for 10 and 333 ng/ml) production by bovine GC. In rats, resistin (10 ng/ml) increased basal and IGF1-induced
progesterone secretion (P!0.0001), without effect on estradiol release. We found no effect of resistin on rat GC proliferation.
Conversely, in cows, resistin increased basal proliferation (P!0.0001; for 100–667 ng/ml) and decreased IGF1-induced proliferation of
GC (P!0.0001; for 10–333 ng/ml) associated with a decrease in cyclin D2 protein level (P!0.0001). Finally, resistin stimulated AKT and
p38-MAPK phosphorylation in both species, ERK1/2-MAPK phosphorylation in rats and had the opposite effect on the AMPK pathway
(P!0.05). In conclusion, our results show that resistin is expressed in rat and bovine ovaries. Furthermore, it can modulate GC functions
in basal state or in response to IGF1 in vitro.
Reproduction (2011) 141 467–479
Introduction
Resistin is an adipocyte-derived cytokine that plays an
important role in the development of insulin resistance
and obesity in rodents (Lazar 2007). This finding was
supported by in vitro experiments using cultured cells as
well as animal experiments (Steppan et al. 2001a,Moon
et al. 2003,Banerjee et al. 2004,Kim et al. 2004,
Rangwala et al. 2004,Satoh et al. 2004). Resistin is a
cysteine-rich protein of around 12 kDa that belongs to a
family of polypeptides named resistin-like molecules
(RELMs). These molecules contain three domains: an
N-terminal signal sequence, a variable middle portion,
and a highly constant C-terminal sequence (Holcomb
et al. 2000,Steppan et al. 2001b). Resistin is secreted in
circulation mainly as a homodimer (Banerjee & Lazar
2001,Rajala et al. 2002). Several types of cell can
express resistin. In mice, adipocytes may be the major
source of resistin (Steppan et al. 2001a,2001b), whereas
in humans, resistin mainly come from monocytes and
macrophages (Nagaev & Smith 2001,Savage et al. 2001,
Patel et al. 2003). Beside its effects on glucose
metabolism and insulin sensitivity, resistin regulates a
plethora of various functions through its action on
multiple cell targets in both rodents and humans. Thus,
resistin is able to exert proinflammatory processes in
adiposetissue(Nagaev et al.2006)andvascular
endothelium (Li et al. 2007), promote vascular smooth
muscle cell proliferation (Calabro et al. 2004), and
stimulate in vitro angiogenesis (Di Simone et al. 2006,
Mu et al. 2006,Robertson et al. 2009). Despite much
research on resistin’s action, the receptor(s) mediating its
biological effects has not yet been identified, and little is
known on the intracellular signaling pathways activated
by this protein.
Recently, some data have suggested that resistin could
affect male and female fertility. Indeed, expression of
resistin (mRNA and protein) has been reported in several
reproductive tissues including hypothalamus (Morash
et al. 2002,Tovar et al. 2005,Wilkinson et al. 2005),
pituitary (Morash et al. 2002,Brown et al. 2005), and
testis (Nogueiras et al. 2004). In hypothalamus, resistin
has been shown to inhibit feeding (To v a r et al. 2005,
Vazquez et al. 2008). In pituitary, resistin expression is
q2011 Society for Reproduction and Fertility DOI: 10.1530/REP-10-0419
ISSN 1470–1626 (paper) 1741–7899 (online) Online version via www.reproduction-online.org
regulated in a nutritional-, age-, and gender-specific
manner (Morash et al. 2004). Resistin mRNA expression
increases to a peak level in pituitary of pre-pubertal mice
(Morash et al. 2002,2004). Furthermore, at this stage,
pituitary mRNA resistin levels are strongly stimulated by
corticosteroids (Brown et al. 2005). Administration of
resistin to dispersed rat anterior pituitary cells increases
GH release (Rodriguez-Pacheco et al. 2009). In rat testis,
resistin protein is detectable throughout postnatal
development, and its mRNA is under the control of
several hormones and mediators such as gonadotropins,
leptin, and nutritional status (Nogueiras et al. 2004). In
this organ, resistin is more precisely present in interstitial
Leydig cells and, to a lesser extent, in Sertoli cells within
seminiferous tubules (Nogueiras et al. 2004). Further-
more, in rodent testis, resistin is able to induce in vitro
testosterone secretion in the basal state and in response
to hCG (Nogueiras et al. 2004). In cultured human theca
cells, 17a-hydroxylase activity is increased by resistin in
the presence of forskolin or forskolinGinsulin,
suggesting a role of resistin in stimulation of androgen
production by theca cells (Munir et al. 2005). Moreover,
some studies have shown elevated concentrations of
serum resistin in women with polycystic ovary syndrome
(Panidis et al. 2004,Carmina et al. 2005,Munir et al.
2005,Yilmaz et al. 2009), which is known to be
associated with hyperinsulinemia, hyperandrogenism,
and insulin resistance (Gambineri et al. 2002). In
mammals, insulin as well as insulin-like growth factor
1 (IGF1) is well known to play a key role on the
development of antral follicles, i.e. steroidogenesis and
granulosa cell (GC) proliferation (Silva et al. 2009). We
and others reported that adipokines, such as adiponectin
and leptin, influence GC function (Zachow & Magoffin
1997,Spicer et al. 2000,Chabrolle et al. 2007a,2007b,
2009,Maillard et al. 2010). Some studies showed that
resistin could interfere with some components of IGF1
receptor signaling, such as insulin receptor substrate 1
(IRS1; Barnes & Miner 2009). All these findings led us to
hypothesize that resistin could be expressed in the ovary
and could modulate IGF1 effects on ovarian cells.
The objectives of this study were to investigate in two
different species, bovine and rat, 1) the mRNA and protein
expression of resistin in ovary and 2) the effect of
recombinant resistin with or without IGF1 on prolifer-
ation, steroidogenesis, and different signaling pathways
(AKT, AMPK, and pERK1/2 and p38-MAPK) in GCs in vitro.
Results
Characterization of resistin in bovine and rat ovaries
In bovine ovary, RT-PCR analysis revealed the amplifi-
cation of one cDNA corresponding to a fragment of
resistin (300 bp) in whole ovary, corpus luteum, small and
large follicles, GCs, immature and in vitro matured
cumulus cells, and oocytes (Fig. 1A). More precisely, we
determined by real-time PCR the resistin mRNA levels in
fresh and cultured GCs from small follicles. As shown in
Fig. 1A, resistin mRNA expression was about fivefold
higher in fresh than in cultured GCs. In rat ovary, RT-PCR
analysis also resulted in the amplification of one cDNA,
which corresponds to a fragment of resistin (321 bp) in
whole ovary and corpus luteum, but not in GCs (Fig. 1C).
As shown in Fig. 1C, the expression of rat resistin mRNA is
very low compared with those of rat cyclophilin A in fresh
GCs. Furthermore, it is undetectable in cultured rat GCs.
As shown in Fig. 1B, resistin protein (as the dimer form,
about 23 kDa) was expressed in bovine large and small
follicles, whole ovary, corpus luteum, cumulus cells,
immature and in vitro matured oocytes, and fresh isolated
and cultured GCs. In rat species, immunoblotting of
protein extracts revealed the presence of resistin (as the
dimer form, about 23 kDa) in whole ovary, corpus luteum
and a weak expression in cultured GCs (Fig. 1D).
Immunohistochemistry analyses confirmed the
immunoblot results. Resistin was localized in bovine
primary and antral follicles, and equally expressed
in oocyte and theca, granulosa and cumulus cells
(Fig. 2). In rat ovary, resistin was present more
abundantly in oocyte, theca cells, and corpus luteum
than in GCs (Fig. 3).
Effect of recombinant resistin on basal and
IGF1-induced steroid production by primary GCs
We next investigated whether the supplementation of
various doses of recombinant human (rh) resistin or
recombinant rat (rr) resistinGexogenous rh IGF1
(76 ng/ml) affected the steroidogenesis of primary bovine
and rat GCs respectively. The progesterone and estradiol
secretions were measured by EIA protocols in the culture
medium after 48 h of treatment.
We found that rh resistin decreased the basal
progesterone and estradiol secretions by primary bovine
GCs whatever the dose used (10–667 ng/ml; Fig. 4A
and C). The decrease in progesterone secretion was
significant for the four doses of rh resistin (10, 100, 333,
and 667 ng/ml; about K22%, P!0.05; Fig. 4A), whereas
only the treatment with 10 and 333 ng/ml of resistin
significantly decreased the estradiol production (about
K30%, P!0.005; Fig. 4C). As expected, the pro-
gesterone and estradiol secretions by cultured bovine
GCs were significantly increased by IGF1 treatment
compared with the basal release, by 2.3- and 1.7-fold
respectively (P!0.0001 and P!0.05; Fig. 4B and D). In
the presence of IGF1, no significant effect of rh resistin
treatment was observed on bovine GC steroidogenesis
(Fig. 4B and D). In the rat, the resistin supplementation at
10 ng/ml significantly increased the progesterone
secretion in the presence or not of IGF1 (about 1.9-fold,
P!0.0001 for both; Fig. 5B and A). No significant effect
of rr resistin was found on both basal and IGF1-induced
estradiol secretion by primary rat GCs (Fig. 5B).
468 V Maillard and others
Reproduction (2011) 141 467–479 www.reproduction-online.org
Effect of recombinant resistin on bovine and rat
GC proliferation
We examined whether the treatment with various doses
of recombinant resistin affected the DNA synthesis of
primary GCs. [
3
H]thymidine incorporation by bovine or
rat GCs was measured after 24 h of culture in serum-free
medium in the presence or absence of rh (10, 100, 333,
and 667 ng/ml) or rr (10 and 100 ng/ml) resistin
respectively Grh IGF1 (76 ng/ml). Resistin did not
modify the basal DNA synthesis of rat GCs (data not
shown). Conversely, resistin (100, 333, and 667 ng/ml)
significantly increased the basal DNA synthesis of
bovine GCs by about 1.3-fold (P!0.0001; Fig. 6A).
We observed in the two species that IGF1 significantly
increased [
3
H]thymidine incorporation induced by
about 5.8- and 1.4-fold in bovine and rat GCs
respectively (P!0.0001; Fig. 6B and data not shown
for rat GCs). Whereas no effect of resistin was shown on
rat GC proliferation induced by IGF1 (data not shown),
resistin at 10, 100, and 333 ng/ml doses significantly
decreased [
3
H]thymidine incorporation induced by IGF1
in bovine GCs (Fig. 6B): this decrease in cell proliferation
was more marked for the dose of 10 ng/ml resistin
(K21.2%, P!0.0001). At this latter dose of resistin, we
observed that the protein level of cyclin D2 induced by
IGF1 (76 ng/ml, 3 h) was significantly reduced (K45%,
P!0.0001) compared with the stimulation with IGF1
alone on bovine GCs (Fig. 6C).
Various signaling pathways modulated by recombinant
resistin treatment in bovine and rat GCs
The pattern of AKT, ERK1/2, p38-MAPK, and AMPKa
phosphorylation from 1 to 120 min was analyzed in
primary bovine and rat GCs, which were overnight
serum starved and then supplemented with rh and rr
resistin respectively. In both species, a rapid and
transient increase in phosphorylated AKT (Figs 7Aand8A)
and phosphorylated p38-MAPK (Figs 7C and 8C) was
observed after 1 min of recombinant resistin treatment
mRNA (Bovine) mRNA (rat)
SF LF CL O
Cum
Im
Oo
Im
GC AT
Resistin (300 bp)
Resistin (300 bp)
Actin (188 bp)
Cum
IVM
Oo
IVM
OGCCL AT
Resistin (321 bp)
Actin (188 bp)
Protein (bovine)
γ-Tubulin γ-Tubulin
γ-Tubulin
LF SF hATOCLMGAT kDa
48
~ 23
Oo
Im
Oo
IVM
hAT hATCum fGC cGC
Resistin dimer
Protein (rat)
Resistin dimer
48
~ 23
UT O ATCLcGC
kDa
Resistin dimer
kDa
48
~ 23
0
0.1
0.2
0.3
fGC cGC
***
Ratio resistin
/cycloA
0
0.001
0.002
0.003
0.004
fGC cGC
Ratio resistin
/cycloA
Undetectable
A
BD
C
Figure 1 Expression of resistin in bovine and rat ovaries. (A and C) RT-PCR analysis of the mRNA for resistin in ovary. In the cow (A), the analysis was
conducted in small (SF) and large (LF) follicles, corpus luteum (CL), whole ovary (O), cumulus cells from immature (Cum Im) and 24 h IVM (Cum
IVM) cumulus–oocyte complexes (COC), immature (Oo Im) and 24 h IVM (Oo IVM) oocytes, fresh granulosa cells (GC), and adipose tissue (AT).
Real-time RT-PCR in fresh and cultured bovine granulosa cells from small follicles. Each reaction was run in duplicates, and threshold (C
t
) values for
resistin mRNA were subtracted from that of cyclophilin A and converted from log linear to linear term. The graph represents mean values from three
different pools of granulosa cells(one pool is representative of one bovine ovary containing aboutten small follicles). ***P!0.001. In the rat (C), the
analysis was performed in O, CL, GC, and AT. DNA fragments were electrophoresed in 1.5% agarose gel stained with ethidium bromide for the two
species. The level of rat resistin mRNAwas also determined by real-time quantitative PCR in fresh and cultured rat granulosa cells as described in the
Materials and Methods section. The graph represents mean values from two different pools of granulosa cells (one pool is representative of 20 rate
ovaries). In cultured rat granulosa cells, the mRNA resistin expression was undetectable. (B and D) Detection of resistin in ovary by immunoblotting.
In cow (B), resistin dimer (w23 kDa) was observed in LF, SF, O, CL, mammary gland (MG), AT, Oo Im (nZ50 per lane), Oo IVM (nZ50 per lane),
cumulus cells (Cum, from 60 24-h IVM COC per lane), and fresh (fGC) and 48 h cultured (cGC) granulosa cells from small follicles (!6 mm).
Human adipose tissue (hAT) was used as positive control for the presence of resistin, since the antibody cross-reacts with the human resistin. In rat
(D), resistin dimer (w23 kDa) was detected in 48 h cGC, uterusCoviduct (UT), O, CL, and AT. g-Tubulin protein was used as a loading control in the
two species (nZ3).
Role of resistin in granulosa cells 469
www.reproduction-online.org Reproduction (2011) 141 467–479
(P!0.05 and P!0.1 respectively for cows, and P!0.005
and P!0.05 respectively for rats). Recombinant resistin
significantly and transiently increased phosphorylation
of ERK1/2-MAPK in rat GCs after 1 min (P!0.005;
Fig. 8B), but not in bovine ones (Fig. 7B). Finally,
phosphorylated AMPKawas significantly increased in
cultured bovine GCs by resistin treatment from 1 to
120 min (P!0.05; Fig. 7D), whereas a significant and
transient decrease in AMPKaphosphorylation was found
in primary rat GCs after 1 min of supplementation
(P!0.05; Fig. 8D).
Discussion
In this study, we reported for the first time that resistin
mRNA and protein were present in various structures of
bovine and rat ovaries. Besides, we showed that a 48 h
treatment with rh resistin decreased progesterone and
estradiol secretions by primary bovine GCs. In the rat, rr
resistin induced the progesterone production by GCs,
without effect on estradiol release. Whereas resistin
treatment did not affect the rat GC proliferation, in the
cow it increased the basal proliferation and decreased
the IGF1-induced proliferation of GCs associated with a
decrease in cyclin D2 protein level. Finally, in both
species, recombinant resistin stimulated AKT and p38-
MAPK phosphorylation and had the opposite effect on
the AMPK pathway in GCs. ERK1/2-MAPK phosphoryl-
ation was only affected in rat GCs.
Protein and mRNA expression of resistin have been
described in several tissues of the reproductive axis
(Morash et al. 2002,Nogueiras et al. 2004,Brown et al.
2005,Tov ar et al. 2005,Wilkinson et al. 2005).
Furthermore, resistin mRNA is expressed in the bovine
mammary gland, and its expression in this tissue
decreases during lactation (Komatsu et al. 2003). Our
results of RT-PCR, immunoblotting, and immunohisto-
chemistry showed the presence of resistin in rat and
bovine whole ovary. In the cow, resistin was widely
expressed in small and large follicles, corpus luteum,
oocyte and cumulus, theca and GCs. In their communi-
cation, Jones et al. (2009) also revealed the mRNA
expression of resistin in rat whole ovary. We demon-
strated in addition that resistin mRNA was present in rat
corpus luteum but very weakly in fresh GCs (and
undetectable in cultured GCs), and that resistin protein
was localized in rat oocyte, theca cells, corpus luteum
and weakly present in GCs. Previously, we found a
similar distribution of adiponectin, another adipokine, in
rat (Chabrolle et al. 2007b), chicken (Chabrolle et al.
2007a), and human (Chabrolle et al. 2009) ovarian cells,
whereas it was equally expressed in all structures
(including fresh and cultured GCs) in bovine ovary
(Maillard et al. 2010). If we hypothesize that the
specificity of primary antibodies used in our study is
the same in each species, our results are in favor of a
specific expression of adipokines in the bovine ovary.
Thus, our present data underline an in situ production of
resistin in ovarian cells and a different expression of this
adipokine in rat and bovine GCs. This local production
of resistin in bovine cultured GCs and not in rat cultured
GCs could contribute to ‘species’ differences in
responses including steroidogenesis and proliferation.
Subsequently, we explored the effects of recombinant
resistin on steroidogenesis and proliferation of primary
bovine and rat GCs in the presence or absence of IGF1.
We tested a physiological dose of resistin (10 ng/ml),
referring to published resistin concentrations in human
(Munir et al. 2005,Asimakopoulos et al. 2009,Hansen
et al. 2010) and rodent (Chabrolle et al. 2008,Shankar
et al. 2010) plasma (between around 5 and 20 ng/ml)
and higher resistin supplementations (100, 333, or
667 ng/ml), since resistin concentration in bovine
plasma is still unknown. Moreover, we chose rr resistin
for tests on rat GCs and rh resistin for tests on bovine
cells, since the protein sequence of Bos taurus resistin is
AF AF
Control IgGResistin
50 µM
50 µM
GC
TC
50 µM
50 µM
50 µM
O
O
Cum
Cum
50 µM
GC
TC
PrF
FF
FF
Figure 2 Localization of resistin in bovine ovary by
immunohistochemistry. Resistin was detected in
primary (PrF) and antral (AF) follicles, granulosa
cells (GC), cumulus cells (Cum), theca cells (TC),
and oocyte (Oo). Follicular fluid (FF). Negative
controls included a section incubated with rabbit
IgG (nZ3).
470 V Maillard and others
Reproduction (2011) 141 467–479 www.reproduction-online.org
closer to the sequence of Homo sapiens resistin
(similarity: around 83%) than that of other species.
In this study, in the cow, the basal but not the IGF1-
induced progesterone and estradiol productions by
primary GCs from small follicles were significantly
decreased by resistin without any dose effect (at the
four doses and only at the two doses 10 and 333 ng/ml
respectively), while resistin at the physiological dose
enhanced progesterone but not estradiol release by
cultured rat GCs. As suggested previously, these different
responses observed between the bovine and rat cultured
GCs could be explained by species differences in resistin
protein expression. Leptin, another adipokine, has been
shown to exert an inhibitory effect on steroidogenesis in
rat (Zachow & Magoffin 1997) and bovine (Spicer et al.
2000) cultured GCs. In cultured rat GCs, leptin inhibited
the increase induced by IGF1 in FSH-stimulated
estradiol production. This inhibitory effect of leptin was
specific for estradiol production, since there was no
effect on basal, FSHK,orFSHCIGF1-dependent
progesterone levels (Zachow & Magoffin 1997). In
bovine cultured GCs, leptin had also weak inhibitory
effects on gonadotropin-and/or IGF1-induced steroido-
genesis (Spicer et al. 2000). In this study, resistin did not
significantly regulate the estradiol secretion of primary
rat GCs in the presence of IGF1, and the progesterone
release in this model was not influenced by IGF1
treatment. Further investigations are required to eluci-
date the role of resistin in the presence of IGF1 on GCs of
different species. Other ovarian cells have also been
described to response to a resistin stimulation. Munir
et al. (2005) had demonstrated in primary human
theca cells that recombinant resistin (notably 10 and
100 ng/ml) increased 17a-hydroxylase activity and
CYP17 (steroidogenic enzyme) mRNA expression
induced by forskolin alone or in combination with
insulin. All these data suggest that physiological levels of
resistin could affect ovarian functions.
Resistin has been reported to stimulate proliferation of
different cellular types (Calabro et al. 2004,Ort et al.
2005,Mu et al. 2006,Park et al. 2008). Conversely,
resistin-13 peptide, a fragment of human resistin
possessing some biological activity of the entire
hormone, inhibited the proliferation of the breast cancer
hCG
(resistin)
PMSG
(resistin)
Immature
(resistin) Control (IgG)
TC
GC
O
TC
GC
O
FF
GC
TC
GC CL
CL
CL
CL
GC
CL
CL
CL
CL
Cum
100 µM
100 µM
100 µM 50 µM
25 µM
250 µM
250 µM
250 µM
AB
CDE
HGF
Figure 3 Localization of resistin in rat ovary by immunohistochemistry. Resistin was detected in ovary from immature rat (A), in large follicles in ovary
from rat treated with PMSG for 48 h (C, D, and E) and in corpus luteum (CL) in ovary from rat treated withPMSG for 48 h and then with hCG for 24 h
(F, G, and H). Resistin was localized more precisely in theca cells (TC), CL and oocyte (O), and weakly in cumulus (Cum) and granulosa (GC) cells.
Follicular fluid (FF). Negative controls (B) included a section incubated with rabbit IgG (nZ3).
Role of resistin in granulosa cells 471
www.reproduction-online.org Reproduction (2011) 141 467–479
MDA-MB-231 cells (Pan et al. 2007). Moreover, other
adipokines, such as adiponectin and leptin, were able to
modulate basal or hormone-induced proliferation of GCs
from different species (Spicer et al. 2000,Sirotkin &
Grossmann 2007,Sirotkin et al. 2008,Maillard et al.
2010,Sirotkin & Meszarosova 2010). For these reasons,
we evaluated the potential effects of several doses of
recombinant resistinGIGF1 (76 ng/ml) on the prolifer-
ation of primary bovine and rat GCs for 24 h. As already
shown for adiponectin (Chabrolle et al. 2007b) and leptin
(Duggal et al. 2002) on rat GC proliferation, no effectof rr
resistin (10 and 100 ng/ml) was observed on basal and
IGF1-induced mitosis of rat GCs in our study, suggesting
that adipokines including resistin do not play a role in rat
GC growth. On the other hand, we found in cows that
resistin was able to affect GC proliferation: the basal cell
proliferation was stimulated with high levels of resistin
(100, 333, and 667 ng/ml) but not with the physiological
dose (10 ng/ml), whereas the IGF1-induced proliferation
was significantly decreased with 10, 100, and 333 ng/ml
resistin. This partial inhibitory effect of resistin was
greater for the physiological supplementation. To inves-
tigate the molecular mechanism involved in this
impaired mitotic response of IGF1-stimulated bovine
GCs to resistin, we evaluated the expression of the cyclin
D2 in GCs treated with resistin (10 ng/ml) for 24 h and
then with IGF1 (76 ng/ml) for 3 h. The cyclin D2 is a well-
established marker of mammalian cell proliferation
and more particularly one of the crucial factors of the
G1/S transition of the cell cycle (Sherr 1993,
Sicinski et al. 1996,Sherr & Roberts 1999). We observed
a significantly decrease in cyclin D2 protein expression
induced by IGF1, suggesting that reduction in IGF1-
induced proliferation of bovine GCs by resistin could be
due to in part an inhibition of cyclin D2.
Until now, few data were available on the intracellular
signaling pathways activated by resistin. This hormone
has been shown to modulate phosphorylation of MAPK
(ERK1/2, p38, and JNK; Di Simone et al. 2009,Chen
et al. 2010), AKT (Palanivel et al. 2006), and AMPK
(Satoh et al. 2004) in different cell types. These signaling
pathways have been described to play a role in
steroidogenesis and/or proliferation of GCs in response
to various hormones such as FSH, IGF1, and insulin, as
well as more recently to some adipokines including
leptin and adiponectin (Moore et al. 2001,Kayampilly &
Menon 2004,Tosca et al. 2005,Yu et al. 2005,Ryan
et al. 2008,Kayampilly & Menon 2009). Since the
majority of resistin effects on rat and bovine steroidogen-
esis and proliferation were observed from the physio-
logical level (10 ng/ml) in our study, we then examined
the effect of this dose on phosphorylation of AKT,
ERK1/2, p38-MAPK, and AMPKain bovine and rat
GCs for 1–120 min. We chose these times of stimulation
because some studies showed that other adipokines
including adiponectin or leptin activate rapidly these
signaling pathways in GCs (Chabrolle et al. 2007b,Lin
et al. 2009). Resistin supplementation led to different
and even opposite effects on ERK1/2-MAPK and AMPK
pathways. Indeed, phosphorylation of ERK1/2-MAPK
A
C
0
0.2
0.4
0.6
0.8
1.0
1.2 a
b
a,b
b
a,b b,c
b,c
c
a,b a,b
a
0
0.5
1
1.5
2
2.5
3
Resistin (ng/ml) 0 10 100 333 667 Resistin (ng/ml) 00 10 100 333 667
b
a
b
bb
0
0.2
0.4
0.6
0.8
1.0
1.2
IGF1–+++++
Resistin (ng/ml) 00 10 100 333 667Resistin (ng/ml) 100 100 333 667
IGF1–+++++
b,c
b,c b,c b
c
a
0
0.5
1
1.5
2
2.5
3
B
D
Bovine steroidogenesis
Ratio progesterone secretion/
basal amount
Ratio estradiol secretion/
basal amount
Ratio estradiol secretion/
basal amount
Ratio progesterone secretion/
basal amount
Figure 4 Effect of rh resistin on progesterone (A and B) and estradiol (C and D) secretions by bovine granulosa cells. Progesterone (A and B) and
estradiol (C and D) secretions were measured by EIA protocol in culture medium of granulosa cells after 48 h of culture in enriched McCoy’s 5A
medium (without FBS) with different concentration of rh resistin (10, 100, 333, and 667 ng/ml)GIGF1 (76 ng/ml; B and D). The data are expressed as
the amount of steroids (pg/ml) secreted per 48 h per basal amount. The concentration of progesterone in the culture medium at the basal state (no
resistin or IGF1 treatment) was 998.6G75.5 ng/ml, and the concentration of estradiol was 73.8G9.7 pg/ml. The results, expressed as meansGS.E.M.,
are representative of three to four independent cultures with each condition in quadruplet. Bars with different superscripts are significantly
different (P!0.05).
472 V Maillard and others
Reproduction (2011) 141 467–479 www.reproduction-online.org
was transiently induced in rat GCs after 1 min of resistin
treatment, whereas no effect was observed in bovine
GCs. The activation of ERK1/2-MAPK pathway may
explain in part the increased secretion of progesterone
measured in medium of rat GCs after 48 h of resistin
treatment, since the use of the specific U0126 inhibitor
of ERK1/2-MAPK has been described to significantly
decrease progesterone secretion (Tosca et al. 2005).
Finally, we found that resistin induced a prolonged
phosphorylation of AMPKafrom 1 to 120 min in bovine
GCs, while resistin transiently decreased the protein
quantity of phosphorylated AMPKain rat GCs after 1 min
of supplementation. In previous studies, we showed that
activation of AMPK with 5-aminoimidazole-4-carbox-
amide-1-d-ribonucleoside decreased progesterone
secretion in basal state in both rat and bovine species
(Tosca et al. 2005,2007). In this study, we observed an
inhibitory and a stimulatory effect of resistin treatment
on basal progesterone secretion by bovine and rat GCs
respectively. Thus, we can postulate that opposite
resistin effects on AMPK phosphorylation found in both
species could take part in this difference. Our data also
showed in both species a rapid and transient increase
in phosphorylated AKT and p38-MAPK after 1 min of
recombinant resistin treatment. Similar results have been
shown in rat GCs in response to adiponectin (Chabrolle
et al. 2007b). These two signaling pathways are involved
in the cell survival and apoptosis of GCs (Westfall et al.
2000,Peter & Dhanasekaran 2003). Thus, we can
speculate that resistin through activation of p38-MAPK
and AKT pathways treatment could regulate follicular
atresia. The specific inhibition of each one of these
intracellular signaling pathways could allow us to really
understand the role of resistin on rat and bovine GC
steroidogenesis and survival.
In conclusion, this study shows the presence of resistin
in bovine and rat ovarian cells. More precisely, resistin
expression is species-dependent in GCs. Furthermore, in
these cells, we observed that recombinant resistin can
modulate steroidogenesis and proliferation in basal state
or in response to IGF1 in vitro. Thus, resistin could be a
metabolic signal involved in the reproductive functions.
Further investigations in several species are required to
understand the different molecular mechanisms involved
in resistin effects and its possible interaction with
different hormones (such as FSH, insulin, IGF1, and
other adipokines) on steroidogenesis and proliferation of
cultured ovarian cells. Particularly, it seems important to
measure plasma resistin in different species in order to
compare the ovarian local and plasma concentration of
this adipokine. However, the role of resistin will not be
able to be fully understood until its receptor is identified.
Materials and Methods
Chemicals, hormones, and antibodies
Unless otherwise stated in the text, chemicals were obtained
from Sigma–Aldrich.
Rh and rr resistins produced in Escherichia coli were purchased
from Biovendor Research and Diagnostic Products (Heidelberg,
Germany). Rh IGF1 was obtained from Sigma–Aldrich.
Rabbit polyclonal antibodies to rat resistin and human
resistin were purchased from Chemicon International (Milli-
pore, Guyancourt, France) and Biovendor Research and
Diagnostic Products respectively. Rabbit polyclonal antibodies
to cyclin D2 (C-17), p38a(C-20), ERK2 (C-14), and phospho-
AKT1/2/3 (Ser473)-R were from Santa Cruz Biotechnology
(Euromedex, Souffelweyersheim, France). Rabbit polyclonal
antibodies to phospho-p44/42 MAPK (ERK1/2) (Thr202/Tyr204),
phospho-p38 MAP kinase (Thr180/Tyr182), phospho-AMPKa
(Thr172), AMPKa, and AKT were obtained from Cell Signaling
Technology (Ozyme, Saint Quentin Yvelines, France). Mouse
MABs to vinculin (clone hVIN-1) and g-tubulin (clone GTU-88)
were purchased from Sigma–Aldrich. HRP-conjugated anti-
rabbit and anti-mouse IgG were purchased from Eurobio
(Les Ulis, France).
A
B
IGF1 (76 ng/ml)
Resistin (ng/ml)
Resistin (ng/ml)
0100 0 10 10010 667 667
–– +++–– +
IGF1 (76 n
g
/ml)
0100 0 10 10010 667 667
–– +++–– +
0
0.5
1
1.5
2
2.5
a,b
c
a,b a
b
d
a,d a,d
0
0.2
0.4
0.6
0.8
1.0
1.2
1.4
1.6
1.8
aaa,b a,b
b,c,d
a,c
d
d
2.0
Rat steroidogenesis
Ratio progesterone secretion/
basal amount
Ratio estradiol secretion/
basal amount
Figure 5 Effect of rr resistin on progesterone (A) and estradiol (B)
secretions by rat granulosa cells. Progesterone (A) and estradiol (B)
amounts were determined by EIA protocol in culture medium of
granulosa cells after 48 h of culture in enriched McCoy’s 5A medium
(without FBS) with different concentration of rh resistin (10, 100, and
667 ng/ml)GIGF1 (76 ng/ml). The data are expressed as the amount of
steroids (pg/ml) secreted per 48 h per basal amount. The concentration
of progesterone in the culture medium at the basal state (no resistin or
IGF1 treatment) was 20G6 ng/ml, and the concentration of estradiol
was 42.4G4.9 pg/ml. The results, expressed as meansGS.E.M., are
representative of three to four independent cultures with each
condition in quadruplet. Different letters indicate significant
difference (P!0.05).
Role of resistin in granulosa cells 473
www.reproduction-online.org Reproduction (2011) 141 467–479
Ethics, animals, and tissues samples
All procedures were approved by the Agricultural Agency and
the Scientific Research Agency, and conducted in accordance
with the guidelines for Care and Use of Agricultural Animals in
Agricultural Research and Teaching.
Mature (60-day old) and immature (21-day old) female rats
of the Wistar strain were purchased from Charles River
(L’Arbresle, France). Protein and mRNA characterization of resistin
were performed on ovaries, uterus (Coviduct), and adipose
tissue from mature female rats. For immunohistochemistry
analyses, ovaries were collected from immature (21-day old)
rats that were treated with 25 IU pregnant mares serum
gonadotropin (PMSG) alone for 48 h to induce follicular
growth (nZ6) or from immature rats that received a single
i.p. injection of 25 IU hCG after 48 h of PMSG treatment to
induce ovulation and luteinization (nZ6).
Bovine ovaries, mammary gland, and abdominal adipose
tissue were collected at a local slaughterhouse.
Tissue samples for mRNA and protein characterization were
frozen in liquid nitrogen and stored at K80 8C.
Isolation and culture of GCs and cumulus–oocyte
complexes
Bovine ovaries from a slaughterhouse were transferred to saline
solution until the dissection. Immature female Wistar rats were
injected subcutaneously with diethylstilboestrol (DES,
1 mg/day) for 3 days. On the third day of DES treatment, the
animals were killed, and the ovaries were removed aseptically.
GCs were isolated by puncturing follicles (follicles !6mm
for cow), allowing the expulsion of cells in modified McCoy’s
5A medium, supplemented with androstenedione (0.1 mmol/l).
The composition of the complete medium was previously
described (Maillard et al.2010). After several steps of
centrifugation/washes in fresh medium, recovered cells were
incubated in modified McCoy’s 5A medium with amphotericin
B (2.7 mmol/l; Eurobio) and 10% fetal bovine serum (FBS, PAA
Laboratories, Les Mureaux, France) for 24 or 48 h, followed by
an overnight serum starvation. Then, cells were cultured in the
presence or absence of test reagents for different time according
to the analyzed biological function. Cultures were performed in
water-saturated atmosphere containing 5% CO
2
in air at 37 8C.
Cumulus–oocyte complexes (COC) were aspirated from 3
to 6 mm antral follicles. COC with a compact and complete
cumulus were selected and washed three times in HEPES-
buffered tissue culture medium-199 (TCM-199) with gentamy-
cin (50 mg/l). Groups of 50 COC were matured in vitro in 500 ml
serum-free TCM-199 supplemented with ascorbic acid
(75 mg/ml), L-cysteine (90 mg/ml), epidermal growth factor
(EGF) (10 ng/ml), and fibroblast growth factor (FGF) from
bovine pituitary (mainly FGF2, 2.2 ng/ml), b-glycine
(720 mg/ml), glutamine (0.1 mg/ml), hCG (5 IU/ml), IGF1
(19 ng/ml), insulin (5 mg/ml), mercaptoethanol (0.1 mM),
PMSG (10 IU/ml), pyruvate (110 mg/ml), selenium (5 ng/ml),
and transferrin (5 mg/ml) (Donnay et al. 2004). In vitro
maturation (IVM) was performed for 24 h at 38.8 8Cin
humidified atmosphere consisting of 5% CO
2
and 95% air.
Oocytes and cumulus cells (Cum) were mechanically separated
Cell proliferation and cyclin D2
expression in bovine granulosa cells
IGF1 (76 ng/ml)
a
d
0
10
20
30
40
50
60
70
80
90
bb,c
cc
Resistin (ng/ml)
0
2
4
6
8
10
12
14
16
18
20
0
a
10
a
100
b
333
c
667
Resistin (ng/ml) 0 0 10 100 333 667
–+++++
+
+
+
+
b
A
Resistin (10 ng/ml, 24 h)
IGF1 (76 n
g
/ml, 3 h)
Cyclin D2
Vinculin
Ratio cyclin
D2/vinculin
0
1
2
3
4
5
6
a
a
b
a
kDa
116
34
C
[3H] thymidine incorporation
(CPM × 103)
B
[3H] thymidine incorporation
(CPM × 103)
Figure 6 Effect of rh resistin on cell proliferation (A and B) and cyclin D2
expression (C) in bovine granulosa cells. (A and B) [
3
H]thymidine
incorporation was determined in bovine granulosa cells cultured for
24 h in enriched McCoy’s 5A medium (without FBS) with various doses
of rh resistin (10, 100, 333, and 667 ng/ml)GIGF1 (76 ng/ml) as
described in the Materials and Methods section. The data are expressed
as meanGS.E.M., and the measurement unit of [
3
H]thymidine
incorporation is counts per min (c.p.m.). The results are representative
of three independent cultures with each condition in triplicate.
Bars with different superscripts are significantly different (P!0.05).
(C) Serum-starved granulosa cells were pre-incubated or not for 24 h
with rh resistin (10 ng/ml) and then stimulated or not with IGF1
(76 ng/ml) for 3 h. Protein extracts were separated by electrophoresis
on 12% (w:v) SDS-polyacrylamide gel. After transfer to nitrocellulose
membranes, the proteins were probed with anti-cyclin D2. The blots
were stripped and reprobed with antibodies against vinculin. Bands on
the blots were quantified, and the cyclin D2/vinculin ratio was
calculated. Values represent meansGS.E.M. relative to the basal state
from three independent experiments. Different letters indicate
significant difference (P!0.05).
474 V Maillard and others
Reproduction (2011) 141 467–479 www.reproduction-online.org
and collected after COC aspiration (immature stage, germinal
vesicle stage for oocyte) or after 24 h of IVM (mature stage).
They were then frozen in liquid nitrogen and stored at K80 8C
before RNA or protein analyses.
RNA isolation, RT-PCR, and real-time RT-PCR
As described previously (Maillard et al. 2010), total DNA-free
RNA was extracted from whole ovary, dissected healthy
follicles (small follicles !6mm%large follicles), corpus luteum,
oocytes, cumulus and GCs, and adipose tissue (as positive
control), and was stored at K80 8CuntilRT.
RTand PCR were carried out according to previous protocols
(Maillard et al. 2010). Briefly, total RNA (1 mg or from 50
oocytes) was reverse transcribed from oligo(dT) 15 Primer
(Promega) in a 20 ml final volume of reaction mixture for 1 h at
37 8C. PCR was performed with 2 mM of each specific primer
designed on bovine or rat resistin (Table 1) and purchased from
Invitrogen (Fischer Scientific, Strasbourg, France). The PCR
conditions were 94 8C for 6 min, 58 8C for 1 min, 72 8C
for 1 min, and 72 8C for 7 min for 35 cycles. After visualization
on 1.5% agarose gel stained with ethidium bromide, all
PCR products were sequenced by Genome Express
(Meylan, France).
For real-time RT-PCR, 1 mg total RNA of fresh or cultured GCs
was reverse transcribed in a final volume of 20 ml using RNase
H–MMLV reverse transcriptase (Superscript II, Invitrogen) and
oligo(dT) 15 primers (Promega). cDNA was then diluted to
1:20. A 15 ml master mix containing 10 ml iQ SYBR Green
supermix (Bio-Rad), 0.25 ml forward primer (10 mM), 0.25 ml
reverse primer (10 mM), and 4.5 ml water was then prepared to
perform real-time PCR. Specific sets of primer pairs used are
shown in Table 1. cDNA dilution (5 ml) was added to the PCR
master mix to a final volume of 20 ml. The following PCR
protocol was used on MyiQ Cycler system (Bio-Rad): initial
denaturation (5 min at 95 8C), followed by a three-step
0
0.5
1
1.5
2
2.5
3
Ratio pAKT/AKT
a
b
a,b a,b a,b
Phospho AKT
AKT
60
60
kDa
Ratio pp38/p38
0
0.4
0.8
1.2
1.6
2
a
b
P
<0.1
a,b
a,b a,b
38
38
kDa
Phospho p38
p38
0
0.5
1
1.5
2
Ratio pAMPK/AMPK
a
bbbb
Phospho AMPKα
AMPKα
62
62
kDa
Phospho ERK1/2
ERK2
44
42
42
kDa
0
0.2
0.4
0.6
0.8
1.0
1.2
1.4
1.6
1.8
Ratio pERK1/2/ERK2
Time of stimulation (min)
resistin 10 ng/ml
0
a
1
a
5
a
30
a
120
Time of stimulation (min)
resistin 10 ng/ml
0 1 5 30 120
Time of stimulation (min)
resistin 10 n
g
/ml
0 1 5 30 120
Time of stimulation (min)
resistin 10 n
g
/ml
0 1 5 30 120
a
ACow B
CD
Figure 7 Effect of rh resistin on phosphorylation of AKT (A), ERK1/2 (B)
and p38 (C) MAPK, and AMPKa(D) in bovine granulosa cells. After
overnight serum starvation, granulosa cells from small follicles were
incubated in serum-free medium with rh resistin (10 ng/ml) from 1 to
120 min. Protein extracts were separated by electrophoresis on 12%
(w:v) SDS-polyacrylamide gel. After transfer to nitrocellulose mem-
branes, theproteins were probed with anti-phospho-AKT1/2/3 (A)or anti-
phospho-ERK1/2 (B) or anti-phospho-p38 (C) or anti-phospho-AMPKa
(D). The blots were stripped and reprobed with antibodies against AKT,
ERK2, p38, or AMPKarespectively. The immunoblots shown are
representative of three independentexperiments. Bands on theblots were
quantified, and the phosphorylated/total protein ratio was calculated.
The results are reported as meansGS.E.M. Different letters indicate
significant difference (P!0.05).
AB
CD
Phospho AKT
Rat
AKT
60
60
kDa Phospho ERK1/2
ERK2
44
42
42
kDa
38
38
kDa
Phospho p38
p38
Phospho AMPKα
AMPKα
62
62
kDa
Time of stimulation (min)
resistin 10 ng/ml
0
0.5
1
1.5
2
2.5
3
Ratio pAKT/AKT
0
a
1
b
5
a
120
a
0
0.5
1
1.5
2
2.5
3
Ratio pERK1/2/ERK2
Time of stimulation (min)
resistin 10 ng/ml
0
a
1
b
5
a
120
a
Time of stimulation (min)
resistin 10 n
g
/ml
0
0.5
1
1.5
2
2.5
0
a
1
b
5
a
120
a,b
Ratio pAMPK/AMPK
Time of stimulation (min)
resistin 10 n
g
/ml
Ratio pp38/p38
0
0.1
0.2
0.3
0.4
0.5
0.6
0.7
0.8
0
a
1
b
5
a
120
a
Figure 8 Effect of rr resistin on phosphorylation of AKT (A), ERK1/2 (B)
and p38 (C) MAPK, and AMPKa(D) in rat granulosa cells. After overnight
serum starvation, granulosa cells were stimulated with rh resistin
(10 ng/ml) in serum-free medium from 1 to 120 min. Protein extracts
were separated by electrophoresis on 12% (w:v) SDS-polyacrylamide
gel. After transfer to nitrocellulose membranes, the proteins were probed
with anti-phospho-AKT1/2/3 (A) or anti-phospho-ERK1/2 (B) or
anti-phospho-p38 (C) or anti-phospho-AMPKa(D). The blots were
stripped and reprobed with antibodies against AKT, ERK2, p38, or
AMPKarespectively. Bands on the blots were quantified, and the
phosphorylated/total protein ratio was calculated. Values represent
meansGS.E.M. from at least three independent experiments. Bars with
different superscripts are significantly different (P!0.05).
Role of resistin in granulosa cells 475
www.reproduction-online.org Reproduction (2011) 141 467–479
amplification program (30 s at 95 8C, followed by 30 s at 60 8C
and 30 s at 72 8C) repeated 40 times. At the end of the PCR,
dissociation was performed by slowly heating the samples from
60 to 95 8C and continuous recording of the decrease in SYBR
Green fluorescence resulting from the dissociation of double-
stranded DNA. The threshold cycle (C
t
), defined as the cycle at
which an increase in fluorescence above a defined baseline
can be first detected, was determined for each sample. Bovine
and rat resistin mRNA levels were estimated on the basis of PCR
efficiency and C
t
deviation of an unknown sample versus a
control according to the equation proposed by Pfaffl (2001):
EDCTtarget ðcontrolKsampleÞ
target . Cyclophilin A was chosen as the
reference gene. The results were expressed as the bovine or
the rat resistin mRNA/cyclophilin A mRNA ratio. Each PCR run
included a no template control and replicates of control and
unknown samples. Runs were performed in duplicates.
Protein isolation and immunoblotting
Protein extraction and separation and immunoblotting were
performed as reported previously (Maillard et al.2010).
Analyses were conducted on total protein extracts from
whole ovary, dissected healthy follicles (small follicles
!6mm %large follicles), corpus luteum, fresh isolated and
cultured GCs, cumulus cells, oocytes and mammary gland,
uterus (Coviduct), and adipose tissue (as positive controls).
Protein lysates were subjected to electrophoresis on 12 or
15% (w:v) SDS-polyacrylamide gel and electrotransferred. The
membranes were then incubated overnight at 4 8C with
appropriate primary antibodies at a 1/1000 final dilution. The
blots were washed with Tris-buffered saline containing 0.1%
Tween 20 several times and were further incubated for 2 h at
room temperature with a HRP-conjugated anti-rabbit or anti-
mouse or anti-goat IgG (dilution 1/5000). The signal of specific
bands, detected by ECL (Western Lightning Plus-ECL, Perkin
Elmer, Life and Analytical Sciences, Courtaboeuf, France), was
quantified with the software Scion Image for Windows (Scion
Corporation, Frederick, MA, USA).
Immunohistochemistry
After fixation and dehydration, rat and cow ovaries were
embedded in paraffin and serially sectioned (7 mm thickness).
Immunohistochemistry was performed as described previously
(Maillard et al. 2010). Sections were incubated overnight at
48C with rabbit antibody raised against either human resistin
(dilution 1/200) for bovine ovaries or rat resistin (dilution
1/200) for rat ovaries or with rabbit IgG (dilution 1/200, as
negative control) in PBS with 5% lamb serum. After several
washes in PBS, sections were incubated for 30 min at room
temperature with a ready-to-use labeled polymer HRP anti-
rabbit antibody (Kit DakoCytomation EnVision Plus System-
HRP; Dako, Trappes, France). Immunoreactivity was revealed
by incubation in DAB at room temperature (Kit DakoCytoma-
tion EnVision Plus System-HRP). The slides were counter-
stained with hematoxylin and observed using an Axioplan
Zeiss transmission microscope.
Progesterone and estradiol measurements
The steroid concentration was determined in serum-free
medium from bovine and rat GCs after 48 h of culture in the
presence or absence of several doses of resistin (10, 100, 333,
or 667 ng/ml)GIGF1 (76 ng/ml). Initially, GCs were grown in
48-well dishes (1.25!10
5
viable cells/250 ml medium/well) in
modified McCoy’s 5A medium with androstenedione
(0.1 mmol/l), amphotericin B (2.7 mmol/l), and 10% FBS for
24 h. After an overnight serum starvation, GCs were incubated
with the different treatments for 48 h. The concentration of
progesterone in the culture medium from bovine and rat GCs
was measured by an EIA protocol as described previously
(Canepa et al. 2008). For a range of progesterone concen-
trations between 0.4 and 10 ng/ml, the intra-assay coefficients
of variation (CV) were in the majority inferior to 10%. The
concentration of estradiol was performed by using the Estradiol
EIA kit from Cayman Chemical (Interchim, Montluc¸on, France)
according to the manufacturer’s procedure. The intra-assay CV
ranged from 22% to about 12% for estradiol concentrations
between 16.4 and 256 pg/ml. The results are expressed as the
amount of steroids (pg/mL) secreted per 48 h per basal amount.
They are representative of three to four independent cultures
with each condition in quadruplicate.
Measurement of GC proliferation and cyclin D2
expression in GCs
Cell proliferation was assessed by the measurement of
[
3
H]thymidine incorporation after 24 h of culture. GCs (2!10
5
viable cells/400 ml medium/well) were cultured into 24-well
dishes in modified McCoy’s 5A medium supplemented with
amphotericin B (2.7 mmol/l) and 10% FBS for 48 h. Cells were
then serum starved overnight followed by the addition of
Table 1 Oligonucleotide primer sequences for RT-PCR amplification.
Primer name Primer sequence Accession number Product size (bp)
Bovine resistin
Sense 50-TGT GCC CCA TAG ATA AAG CC-30NM_183362 300
Antisense 50-CAG GCC TGC AGC AGT CTT AG-30
Rat resistin
Sense 50-CCT CCT TTT CCT TTT CTT CC-30NM_144741 321
Antisense 50-AAC CAA CCC GCA GGG TAC AG-30
Cyclophilin A
Sense 50-GCA TACA GGT CCT GGC ATC T-30NM_178320 217
Antisense 50-TGT CCA CAG TCA GCA ATG GT-30
476 V Maillard and others
Reproduction (2011) 141 467–479 www.reproduction-online.org
0.25 mCi [
3
H]thymidine (Perkin Elmer; Life and Analytical
Sciences) in the presence or absence of resistin (10, 100, 333,
or 667 ng/ml)GIGF1 (76 ng/ml). Cultures were maintained at
37 8C under 5% CO
2
in air. After 24 h of culture, excess of
thymidine was removed by washing twice with PBS, fixed
with cold 50% trichloroacetic acid for 15 min, and lysed by
0.5 mol/l NaOH. The radioactivity was determined in Ultima
Gold MV scintillation fluid (Perkin Elmer) by counting in a
b-photomultiplier. The values, expressed as counts per min
(c.p.m.), are representative of three independent cultures with
each condition in triplicate.
For the determination of the protein levels of cyclin D2,
bovine GCs were starved overnight. Cells were then pre-
incubated in new serum-free medium without or with rh
resistin (10 ng/ml) for 24 h, and then IGF1 (76 ng/ml) was
added or not for 3 h in the serum-free medium. Cells were then
lysed, and immunoblots were performed.
Statistical analysis
All experimental results are presented as meansGS.E.M.
Statistical analyses were carried out using an one-way (for
data on signaling pathways and cyclin D2 expression) or a two-
way (for results on steroidogenesis and cell proliferation)
factorial ANOVA test followed by Fisher’s PLSD test, when
the ANOVA revealed significant effects. In the various graphs,
bars with different superscripts were considered statistically
significant at P!0.05.
Declaration of interest
The authors declare that there is no conflict of interest that
could be perceived as prejudicing the impartiality of the
research reported.
Funding
This study was supported by the GIS-AGENAE Programme,
ANR, and APIS-GENE. V Maillard is a recipient of a grant
from INRA.
Acknowledgements
We thank Thierry Delpuech and Pascal Papillier for the
collection of bovine ovaries; Claude Cahier and his staff for
the rat care; and Gilles Gomot, Jean-Philippe Dubois, and
Albert Arnoud for the collection of bovine tissue samples.
References
Asimakopoulos B, Milousis A, Gioka T, Kabouromiti G, Gianisslis G,
Troussa A, Simopoulou M, Katergari S, Tripsianis G & Nikolettos N 2009
Serum pattern of circulating adipokines throughout the physiological
menstrual cycle. Endocrine Journal 56 425–433. (doi:10.1507/endocrj.
K08E-222)
Banerjee RR & Lazar MA 2001 Dimerization of resistin and resistin-like
molecules is determined by a single cysteine. Journal of Biological
Chemistry 276 25970–25973. (doi:10.1074/jbc.M103109200)
Banerjee RR, Rangwala SM, Shapiro JS, Rich AS, Rhoades B, Qi Y, Wang J,
Rajala MW, Pocai A, Scherer PE et al. 2004 Regulation of fasted blood
glucose by resistin. Science 303 1195–1198. (doi:10.1126/science.
1092341)
Barnes KM & Miner JL 2009 Role of resistin in insulin sensitivity in rodents
and humans. Current Protein & Peptide Science 10 96–107. (doi:10.
2174/138920309787315239)
Brown R, Wiesner G, Ur E & Wilkinson M 2005 Pituitary resistin gene
expression is upregulated in vitro and in vivo by dexamethasone but is
unaffected by rosiglitazone. Neuroendocrinology 81 41–48. (doi:10.
1159/000084873)
Calabro P, Samudio I, Willerson JT & Yeh ET 2004 Resistin promotes smooth
muscle cellproliferationthrough activation of extracellular signal-regulated
kinase 1/2 and phosphatidylinositol 3-kinase pathways. Circulation 110
3335–3340. (doi:10.1161/01.CIR.0000147825.97879.E7)
Canepa S, Laine A, Bluteau A, Fagu C, Flon C & Monniaux D 2008
Validation d’une me
´thode immunoenzymatique pour le dosage de la
progeste
´rone dans le plasma des ovins et des bovins. Les Cahiers
Techniques de l’Inra 64 19–30.
Carmina E, Orio F, Palomba S, Cascella T, Longo RA, Colao AM,
Lombardi G & Lobo RA 2005 Evidence for altered adipocyte function
in polycystic ovary syndrome. European Journal of Endocrinology 152
389–394. (doi:10.1530/eje.1.01868)
Chabrolle C, Tosca L, Crochet S, Tesseraud S & Dupont J 2007aExpression
of adiponectin and its receptors (AdipoR1 and AdipoR2) in chicken
ovary: potential role in ovarian steroidogenesis. Domestic Animal
Endocrinology 33 480–487. (doi:10.1016/j.domaniend.2006.08.002)
Chabrolle C, Tosca L & Dupont J 2007bRegulation of adiponectin and its
receptors in rat ovary by human chorionic gonadotrophin treatment and
potential involvement of adiponectin in granulosa cell steroidogenesis.
Reproduction 133 719–731. (doi:10.1530/REP-06-0244)
Chabrolle C, Jeanpierre E, Tosca L, Rame C & Dupont J 2008 Effects of high
levels of glucose on the steroidogenesis and the expression of
adiponectin receptors in rat ovarian cells. Reproductive Biology and
Endocrinology 611. (doi:10.1186/1477-7827-6-11)
Chabrolle C, Tosca L, Rame C, Lecomte P, Royere D & Dupont J 2009
Adiponectin increases insulin-like growth factor I-induced progesterone
and estradiol secretion in human granulosa cells. Fertility and Sterility 92
1988–1996. (doi:10.1016/j.fertnstert.2008.09.008)
Chen C, Jiang J, Lu JM, Chai H, Wang X, Lin PH & Yao Q 2010 Resistin
decreases expression of endothelial nitric oxide synthase through
oxidative stress in human coronary artery endothelial cells. American
Journal of Physiology. Heart and Circulatory Physiology 299
H193–H201. (doi:10.1152/ajpheart.00431.2009)
Di Simone N, Di Nicuolo F, Sanguinetti M, Castellani R, D’Asta M,
Caforio L & Caruso A 2006 Resistin regulates human choriocarcinoma
cell invasive behaviour and endothelial cell angiogenic processes.
Journal of Endocrinology 189 691–699. (doi:10.1677/joe.1.06610)
Di Simone N, Di Nicuolo F, Marzioni D, Castellucci M, Sanguinetti M,
D’Lppolito S & Caruso A 2009 Resistin modulates glucose uptake and
glucose transporter-1 (GLUT-1) expression in trophoblast cells. Journal of
Cellular and Molecular Medicine 13 388–397. (doi:10.1111/j.1582-
4934.2008.00337.x)
Donnay I, Faerge I, Grondahl C, Verhaeghe B, Sayoud H, Ponderato N,
Galli C & Lazzari G 2004 Effect of prematuration, meiosis activating
sterol and enriched maturation medium on the nuclear maturation and
competence to development of calf oocytes. Theriogenology 62
1093–1107. (doi:10.1016/j.theriogenology.2003.12.019)
Duggal PS, Ryan NK, Van der Hoek KH, Ritter LJ, Armstrong DT,
Magoffin DA & Norman RJ 2002 Effects of leptin administration and feed
restriction on thecal leucocytes in the preovulatory rat ovary and the
effects of leptin on meiotic maturation, granulosa cell proliferation,
steroid hormone and PGE
2
release in cultured rat ovarian follicles.
Reproduction 123 891–898. (doi:10.1530/rep.0.1230891)
Gambineri A, Pelusi C, Vicennati V, Pagotto U & Pasquali R 2002 Obesity
and the polycystic ovary syndrome. International Journal of Obesity and
Related Metabolic Disorders 26 883–896. (doi:10.1038/sj.ijo.0801994)
Hansen D, Dendale P, Beelen M, Jonkers RA, Mullens A, Corluy L,
Meeusen R & van Loon LJ 2010 Plasma adipokine and inflammatory
marker concentrations are altered in obese, as opposed to non-obese,
type 2 diabetes patients. European Journal of Applied Physiology 109
397–404. (doi:10.1007/s00421-010-1362-5)
Role of resistin in granulosa cells 477
www.reproduction-online.org Reproduction (2011) 141 467–479
Holcomb IN, Kabakoff RC, Chan B, Baker TW, Gurney A, Henzel W,
Nelson C, Lowman HB, Wright BD, Skelton NJ et al. 2000 FIZZ1, a
novel cysteine-rich secreted protein associated with pulmonary inflam-
mation, defines a new gene family. EMBO Journal 19 4046–4055.
(doi:10.1093/emboj/19.15.4046)
Jones AM, Rodgers J, Antibus D, Knoop A, Bruot B & Marcinkiewicz J 2009
Relative ovarian resistin expression in normal cycling rats and rats with
cystic ovaries. Biology of Reproduction 81 (Supplement 1) 532.
Kayampilly PP & Menon KM 2004 Inhibition of extracellular signal-
regulated protein kinase-2 phosphorylation by dihydrotestosterone
reduces follicle-stimulating hormone-mediated cyclin D2 messenger
ribonucleic acid expression in rat granulosa cells. Endocrinology 145
1786–1793. (doi:10.1210/en.2003-1029)
Kayampilly PP & Menon KM 2009 Follicle-stimulating hormone inhibits
adenosine 50-monophosphate-activated protein kinase activation and
promotes cell proliferation of primary granulosa cells in culture through
an Akt-dependent pathway. Endocrinology 150 929–935. (doi:10.1210/
en.2008-1032)
Kim KH, Zhao L, Moon Y, Kang C & Sul HS 2004 Dominant inhibitory
adipocyte-specific secretory factor (ADSF)/resistin enhances adipogen-
esis and improves insulin sensitivity. PNAS 101 6780–6785. (doi:10.
1073/pnas.0305905101)
Komatsu T, Itoh F, Mikawa S & Hodate K 2003 Gene expression of resistin
in adipose tissue and mammary gland of lactating and non-lactating
cows. Journal of Endocrinology 178 R1–R5. (doi:10.1677/joe.0.
178R001)
Lazar MA 2007 Resistin- and obesity-associated metabolic diseases.
Hormone and Metabolic Research 39 710–716. (doi:10.1055/s-2007-
985897)
Li Y, Wang Y, Li Q, Chen Y, Sun SZ, Zhang WD & Jia Q 2007 Effect of resistin
on vascular endothelium secretion dysfunction in rats. Endothelium 14
207–214. (doi:10.1080/10623320701617225)
Lin Q, Poon SL, Chen J, Cheng L, HoYuen B & Leung PC 2009 Leptin
interferes with 30,50-cyclic adenosine monophosphate (cAMP) signaling
to inhibit steroidogenesis in human granulosa cells. Reproductive
Biology and Endocrinology 7115. (doi:10.1186/1477-7827-7-115)
Maillard V, Uzbekova S, Guignot F, Perreau C, Rame C, Coyral-Castel S &
Dupont J 2010 Effect of adiponectin on bovine granulosa cell steroido-
genesis, oocyte maturation and embryo development. Reproductive
Biology and Endocrinology 823. (doi:10.1186/1477-7827-8-23)
Moon B, Kwan JJ, Duddy N, Sweeney G & Begum N 2003 Resistin
inhibits glucose uptake in L6 cells independently of changes in insulin
signaling and GLUT4 translocation. American Journal of Physiology.
Endocrinology and Metabolism 285 E106–E115. (doi:10.1152/ajpendo.
00457.2002)
Moore RK, Otsuka F & Shimasaki S 2001 Role of ERK1/2 in the differential
synthesis of progesterone and estradiol by granulosa cells. Biochemical
and Biophysical Research Communications 289 796–800. (doi:10.1006/
bbrc.2001.6052)
Morash BA, Willkinson D, Ur E & Wilkinson M 2002 Resistin expression
and regulation in mouse pituitary. FEBS Letters 526 26–30. (doi:10.1016/
S0014-5793(02)03108-3)
Morash BA, Ur E, Wiesner G, Roy J & Wilkinson M 2004 Pituitary resistin
gene expression: effects of age, gender and obesity. Neuroendocrinology
79 149–156. (doi:10.1159/000077273)
Mu H, Ohashi R, Yan S, Chai H, Yang H, Lin P, Yao Q & Chen C 2006
Adipokine resistin promotes in vitro angiogenesis of human endothelial
cells. Cardiovascular Research 70 146–157. (doi:10.1016/j.cardiores.
2006.01.015)
Munir I, Yen HW, Baruth T, Tarkowski R, Azziz R, Magoffin DA &
Jakimiuk AJ 2005 Resistin stimulation of 17alpha-hydroxylase activity in
ovarian theca cells in vitro: relevance to polycystic ovary syndrome.
Journal of Clinical Endocrinology and Metabolism 90 4852–4857.
(doi:10.1210/jc.2004-2152)
Nagaev I & Smith U 2001 Insulin resistance and type 2 diabetes are not
related to resistin expression in human fat cells or skeletal muscle.
Biochemical and Biophysical Research Communications 285 561–564.
(doi:10.1006/bbrc.2001.5173)
Nagaev I, Bokarewa M, Tarkowski A & Smith U 2006 Human resistin is a
systemic immune-derived proinflammatory cytokine targeting both
leukocytes and adipocytes. PLoS ONE 1e31. (doi:10.1371/journal.
pone.0000031)
Nogueiras R, Barreiro ML, Caminos JE, Gaytan F, Suominen JS,
Navarro VM, Casanueva FF, Aguilar E, Toppari J, Dieguez C et al.
2004 Novel expression of resistin in rat testis: functional role and
regulation by nutritional status and hormonal factors. Journal of Cell
Science 117 3247–3257. (doi:10.1242/jcs.01196)
Ort T, Arjona AA, MacDougall JR, Nelson PJ, Rothenberg ME, Wu F,
Eisen A & Halvorsen YD 2005 Recombinant human FIZZ3/resistin
stimulates lipolysis in cultured human adipocytes, mouse adipose
explants, and normal mice. Endocrinology 146 2200–2209. (doi:10.
1210/en.2004-1421)
Palanivel R, Maida A, Liu Y & Sweeney G 2006 Regulation of insulin
signalling, glucose uptake and metabolism in rat skeletal muscle cells
upon prolonged exposure to resistin. Diabetologia 49 183–190. (doi:10.
1007/s00125-005-0060-z)
Pan B, Zhao MH, Chen Z, Lu L, Wang Y, Shi DW & Han PZ 2007 Inhibitory
effects of resistin-13-peptide on the proliferation, adhesion, and invasion
of MDA-MB-231 in human breast carcinoma cells. Endocrine-Related
Cancer 14 1063–1071. (doi:10.1677/erc.1.01304)
Panidis D, Koliakos G, Kourtis A, Farmakiotis D, Mouslech T & Rousso D
2004 Serum resistin levels in women with polycystic ovary syndrome.
Fertility and Sterility 81 361–366. (doi:10.1016/j.fertnstert.2003.06.021)
Park S, Hong SM, Sung SR & Jung HK 2008 Long-term effects of central
leptin and resistin on body weight, insulin resistance, and beta-cell
function and mass by the modulation of hypothalamic leptin and insulin
signaling. Endocrinology 149 445–454. (doi:10.1210/en.2007-0754)
Patel L, Buckels AC, Kinghorn IJ, Murdock PR, Holbrook JD, Plumpton C,
Macphee CH & Smith SA 2003 Resistin is expressed in human
macrophages and directly regulated by PPAR gamma activators.
Biochemical and Biophysical Research Communications 300 472–476.
(doi:10.1016/S0006-291X(02)02841-3)
Peter AT & Dhanasekaran N 2003 Apoptosis of granulosa cells: a review on
the role of MAPK-signalling modules. Reproduction in Domestic
Animals 38 209–213. (doi:10.1046/j.1439-0531.2003.00438.x)
Pfaffl MW 2001 A new mathematical model for relative quantification
in real-time RT-PCR. Nucleic Acids Research 29 e45. (doi:10.1093/nar/
29.9.e45)
Rajala MW, Lin Y, Ranalletta M, Yang XM, Qian H, Gingerich R, Barzilai N
& Scherer PE 2002 Cell type-specific expression and coregulation of
murine resistin and resistin-like molecule-alpha in adipose tissue.
Molecular Endocrinology 16 1920–1930. (doi:10.1210/me.2002-0048)
RangwalaSM, Rich AS, Rhoades B,Shapiro JS, Obici S, Rossetti L & Lazar MA
2004 Abnormal glucose homeostasis due to chronic hyperresistinemia.
Diabetes 53 1937–1941. (doi:10.2337/diabetes.53.8.1937)
Robertson SA, Rae CJ & Graham A 2009 Induction of angiogenesis by
murine resistin: putative role of PI3-kinase and NO-dependent pathways.
Regulatory Peptides 152 41–47. (doi:10.1016/j.regpep.2008.07.008)
Rodriguez-Pacheco F, Vazquez-Martinez R, Martinez-Fuentes AJ,
Pulido MR, Gahete MD, Vaudry H, Gracia-Navarro F, Dieguez C,
Castano JP & Malagon MM 2009 Resistin regulates pituitary somatotrope
cell function through the activation of multiple signaling pathways.
Endocrinology 150 4643–4652. (doi:10.1210/en.2009-0116)
Ryan KE, Glister C, Lonergan P, Martin F, Knight PG & Evans AC 2008
Functional significance of the signal transduction pathways Akt and Erk
in ovarian follicles: in vitro and in vivo studies in cattle and sheep.
Journal of Ovarian Research 12. (doi:10.1186/1757-2215-1-2)
Satoh H, Nguyen MT, Miles PD, Imamura T, Usui I & Olefsky JM 2004
Adenovirus-mediated chronic "hyper-resistinemia" leads to in vivo
insulin resistance in normal rats. Journal of Clinical Investigation 114
224–231. (doi:10.1172/JCI200420785)
Savage DB, Sewter CP, Klenk ES, Segal DG, Vidal-Puig A, Considine RV &
O’Rahilly S 2001 Resistin/Fizz3 expression in relation to obesity and
peroxisome proliferator-activated receptor-gamma action in humans.
Diabetes 50 2199–2202. (doi:10.2337/diabetes.50.10.2199)
Shankar K, Kang P, Harrell A, Zhong Y, Marecki JC, Ronis MJ & Badger TM
2010 Maternal overweight programs insulin and adiponectin signaling
in the offspring. Endocrinology 151 2577–2589. (doi:10.1210/en.
2010-0017)
Sherr CJ 1993 Mammalian G1 cyclins. Cell 73 1059–1065. (doi:10.1016/
0092-8674(93)90636-5)
Sherr CJ & Roberts JM 1999 CDK inhibitors: positive and negative
regulators of G1-phase progression. Genes and Development 13
1501–1512. (doi:10.1101/gad.13.12.1501)
478 V Maillard and others
Reproduction (2011) 141 467–479 www.reproduction-online.org
Sicinski P, Donaher JL, Geng Y, Parker SB, Gardner H, Park MY, Robker RL,
Richards JS, McGinnis LK, Biggers JD et al. 1996 Cyclin D2 is an
FSH-responsive gene involved in gonadal cell proliferation and
oncogenesis. Nature 384 470–474. (doi:10.1038/384470a0)
Silva JR, Figueiredo JR & van den Hurk R 2009 Involvement of growth
hormone (GH) and insulin-like growth factor (IGF) system in ovarian
folliculogenesis. Theriogenology 71 1193–1208. (doi:10.1016/j.therio-
genology.2008.12.015)
Sirotkin AV & Grossmann R 2007 Leptin directly controls proliferation,
apoptosis and secretory activity of cultured chicken ovarian cells.
Comparative Biochemistry and Physiology. Part A, Molecular &
Integrative Physiology 148 422–429. (doi:10.1016/j.cbpa.2007.06.001)
Sirotkin AV & Meszarosova M 2010 Comparison of effects of leptin and
ghrelin on porcine ovarian granulosa cells. Domestic Animal
Endocrinology 39 1–9. (doi:10.1016/j.domaniend.2009.06.001)
Sirotkin AV, Mlyncek M, Makarevich AV, Florkovicova I & Hetenyi L 2008
Leptin affects proliferation-, apoptosis- and protein kinase A-related
peptides in human ovarian granulosa cells. Physiological Research 57
437–442.
Spicer LJ, Chamberlain CS & Francisco CC 2000 Ovarian action of leptin:
effects on insulin-like growth factor-I-stimulated function of granulosa
and thecal cells. Endocrine 12 53–59. (doi:10.1385/ENDO:12:1:53)
Steppan CM, Bailey ST, Bhat S, Brown EJ, Banerjee RR, Wright CM,
Patel HR, Ahima RS & Lazar MA 2001aThe hormone resistin links
obesity to diabetes. Nature 409 307–312. (doi:10.1038/35053000)
Steppan CM, Brown EJ, Wright CM, Bhat S, Banerjee RR, Dai CY,
Enders GH, Silberg DG, Wen X, Wu GD et al. 2001bA family of tissue-
specific resistin-like molecules. PNAS 98 502–506. (doi:10.1073/pnas.
98.2.502)
Tosca L, Froment P, Solnais P, Ferre P, Foufelle F & Dupont J 2005
Adenosine 50-monophosphate-activated protein kinase regulates pro-
gesterone secretion in rat granulosa cells. Endocrinology 146
4500–4513. (doi:10.1210/en.2005-0301)
Tosca L, Chabrolle C, Uzbekova S & Dupont J 2007 Effects of metformin
on bovine granulosa cells steroidogenesis: possible involvement of
adenosine 50monophosphate-activated protein kinase (AMPK). Biology
of Reproduction 76 368–378. (doi:10.1095/biolreprod.106.055749)
Tovar S, Nogueiras R, Tung LY, Castaneda TR, Vazquez MJ, Morris A,
Williams LM, Dickson SL & Dieguez C 2005 Central administration of
resistin promotes short-term satiety in rats. European Journal of
Endocrinology 153 R1–R5. (doi:10.1530/eje.1.01999)
Vazquez MJ, Gonzalez CR, Varela L, Lage R, Tovar S, Sangiao-Alvarellos S,
Williams LM, Vidal-Puig A, Nogueiras R, Lopez M et al. 2008 Central
resistin regulates hypothalamic and peripheral lipid metabolism in a
nutritional-dependent fashion. Endocrinology 149 4534–4543. (doi:10.
1210/en.2007-1708)
Westfall SD, Hendry IR, Obholz KL, Rueda BR & Davis JS 2000 Putative
role of the phosphatidylinositol 3-kinase-Akt signaling pathway in the
survival of granulosa cells. Endocrine 12 315–321. (doi:10.1385/
ENDO:12:3:315)
Wilkinson M, Wilkinson D, Wiesner G, Morash B & Ur E 2005
Hypothalamic resistin immunoreactivity is reduced by obesity in
the mouse: co-localization with alpha-melanostimulating hormone.
Neuroendocrinology 81 19–30. (doi:10.1159/000084871)
Yilmaz M, Bukan N, Demirci H, Ozturk C, Kan E, Ayvaz G & Arslan M
2009 Serum resistin and adiponectin levels in women with polycystic
ovary syndrome. Gynecological Endocrinology 25 246–252. (doi:10.
1080/09513590802653833)
Yu FQ, Han CS, Yang W, Jin X, Hu ZY & Liu YX 2005 Activation of the p38
MAPK pathway by follicle-stimulating hormone regulates steroidogen-
esis in granulosa cells differentially. Journal of Endocrinology 186 85–96.
(doi:10.1677/joe.1.05955)
Zachow RJ & Magoffin DA 1997 Direct intraovarian effects of leptin:
impairment of the synergistic action of insulin-like growth factor-I on
follicle-stimulating hormone-dependent estradiol-17 beta production by
rat ovarian granulosa cells. Endocrinology 138 847–850. (doi:10.1210/
en.138.2.847)
Received 7 October 2010
First decision 8 November 2010
Revised manuscript received 26 December 2010
Accepted 14 January 2011
Role of resistin in granulosa cells 479
www.reproduction-online.org Reproduction (2011) 141 467–479
... What is interesting is that a close relationship between resistin and ovarian cell function has been described previously: for example, resistin influences ovarian follicular cell steroidogenesis (19). Importantly, resistin promote the proliferation of human ovarian cancer cells (20), whereas in cows it increases basal proliferation and decreases insulin-like growth factor 1 (IGF1)-induced proliferation of granulosa cells (21). An antiapoptotic effect of resistin was observed in porcine ovarian follicular cells by activation of MAP3/1, AKT and STAT3 kinase pathways (22). ...
... Furthermore, in our previous study, we showed that resistin increases the oestradiol level (an important mitogenic factor) in luteal cells, thus confirming the present data (24). In our study, we noted the stimulating effect of resistin on cell viability, which is consistent with the literature data showing the mitogenic role of resistin in bovine, rat and porcine ovarian cells (21,20). We confirm the mitogenic properties of resistin by showing a stimulatory effect on the PCNA level, which is a marker of proliferation described also in porcine luteal cells; its accumulation was observed in the S proliferator phase of the cell cycle (34) and during CL formation compared to an atretic CL (35). ...
... We confirm the mitogenic properties of resistin by showing a stimulatory effect on the PCNA level, which is a marker of proliferation described also in porcine luteal cells; its accumulation was observed in the S proliferator phase of the cell cycle (34) and during CL formation compared to an atretic CL (35). On the other hand, as showed resistin had no effect on rat (21), human (36) and porcine (22) follicular cells proliferation confirmed by lack of changes in cyclin A and B protein level (22). Nevertheless, resistin role in cells proliferation was never studied in luteal cells of any species and observed differences between ovarian follicular and luteal cells may be linked with different hormonal status of the studied structure and different expression of resistin system itself. ...
Article
The formation and luteolysis of the corpus luteum (CL) is strictly controlled by many factors. Imbalance between proliferation and apoptosis processes leads to deficiency of the luteal phase and infertility. Our previous study showed resistin expression in porcine luteal cells and an inhibitory effect on progesterone synthesis. Thus, the aim of the present study was to examine the in vitro effect of resistin on the proliferation/viability, apoptosis and autophagy of porcine luteal cells as well as the involvement of mitogen-activated kinase (MAP3/1), protein kinase B (AKT) and signal transducer and activator of transcription 3 (STAT3) in these processes. Porcine luteal cells were incubated with resistin (0.1-10 ng/mL) for 24-72 h and viability was assessed using the alamarBlue or 3-[4,5-dimethylthiazole-2-yl]-2,5-diphenyltetrazolium bromide (MTT) assay. Then, the time-dependent effect of resistin on mRNA and protein expression of proliferating cell nuclear antigen (PCNA), caspase 3, BCL2-like protein 4 (BAX), B-cell lymphoma 2 (BCL2), beclin1, microtubule-associated protein 1A/1B-light chain 3 (LC3) and lysosomal-associated membrane protein 1 (LAMP1) was measured by real-time polymerase chain reaction (PCR) and immunoblotting, respectively. We found that resistin enhanced luteal cell viability with no effect on caspase 3 mRNA and protein, increased the BAX/BCL2 mRNA and protein ratio and significantly stimulated the initiation of autophagy, which promotes the maintenance of CL function rather than its regression. Additionally, using pharmacological inhibitors of MAP3/1 (PD98059), AKT (LY294002) and STAT3 (AG490), we observed that the effect of resistin was reversed to the control level in viability and, by influence, MAP3/1 and STAT3 in autophagy. Taken together, our results suggest that resistin, in addition to its well-known effect on granulosa cell function has direct influence on CL luteolysis and the formation and maintenance of luteal cell function.
... Intraperitoneal administration of leptin resulted in increased plasma testosterone levels at lower dose while had no effect when the dosage was increased (Hoffmann et al., 2016). In addition, in vitro studies on granulosa cells steroidogenesis demonstrated that lower concentrations of adiponectin (Messini et al., 2021) and resistin (Maillard et al., 2011) were able to stimulate and inhibit estradiol production, respectively, whereas the adipokines had no such effect at higher concentrations of treatment. The available reports point towards species-specific as well as treatmentdependent variation in the effect of nesfatin-1 on steroidogenesis, hence, further studies are required to develop a comprehensive understanding of its role in steroid biosynthesis. ...
Article
Nesfatin-1 is a pleiotropic hormone implicated in various physiological functions including reproduction. Studies though limited, have established an important role of the peptide in regulation of testicular functions in mammals and fishes. However, role of nesfatin-1 in regulation of spermatogenesis and testicular steroidogenesis remains completely unexplored in reptiles. Therefore, present study aimed to develop an insight into reproductive phase-dependent testicular expression, function and regulation of nucb2/nesfatin-1 in a reptile, Hemidactylus flaviviridis. Expression of nucb2/nesfatin-1 in testis of wall lizard varied significantly depending upon reproductive phase, being highest in the active phase while lowest during regressed phase. Further, in vitro treatment of wall lizard testis with nesfatin-1 showed a concentration- and time-dependent stimulatory effect of the peptide on expression of cell proliferation and differentiation markers like scf, c-kit and pcna suggesting a spermatogenic role of nesfatin-1 in wall lizard. Also, nesfatin-1 stimulated the anti-apoptotic marker, bcl-2 while inhibited the apoptotic marker, caspase-3, suggesting its role as an inhibitor of apoptosis of testicular cells. Further, treatment with nesfatin-1 resulted in significantly higher expression of star along with a concomitant increase in testosterone production by the lizard testis. The present study also demonstrates hormonal regulation of testicular nucb2/nesfatin-1 wherein follicle-stimulating hormone (FSH) inhibited while sex steroids like dihydrotestosterone (DHT) and 17β-estradiol-3-benzoate (E2) stimulated the mRNA expression of nesfatin-1. Observations from the current study for the first time provide comprehensive evidence of spermatogenic and steroidogenic role of nesfatin-1 as well as its hormonal regulation in the testis of a reptile, H. flaviviridis.
... A study conducted on bovine and rat ovaries has demonstrated the expression of resistin. This study revealed that resistin has an inhibitory effect on granulosa cell steroidogenesis in bovine ovaries, but has a stimulatory effect on P4 secretion in rats [60]. In a recent study, it was observed that resistin decreased progesterone (P4) levels, increased estradiol (E2) levels, and regulated various pathways, such as those of steroidogenic acute regulatory protein (STAR), cholesterol side-chain cleavage enzyme, 3β-hydroxysteroid dehydrogenase, estrogen synthetase, through the activation of protein kinase A and mitogen-activated protein kinase 1 [61]. ...
Article
Full-text available
Simple Summary Adipokines are cytokines secreted by adipose tissue that play a crucial role in several biological processes, including energy balance, food intake, immune status, and reproduction. Research has shown that there is a close correlation between energy balance, metabolism, and reproductive function in mammals. Among these factors, ovarian physiology has been found to have its own local regulation. In this study, we aimed to gain insight into the local production of novel adipokines and their receptors in the corpus luteum of cycling and pregnant cows. We examined corpora lutea at various stages in cycling and pregnant cows to investigate the involvement of locally produced adipokines and their receptors in ovarian physiology. Our findings reveal significant differences in the expression levels of adipokines throughout the estrous cycle and pregnancy, indicating their potential role in corpus luteum lysis and/or resumption of cyclicity. Abstract This study aimed to determine the gene expression of different local novel adipokines, such as vaspin, adiponectin, visfatin, and resistin, and their known receptors, namely, heat shock 70 protein 5, adiponectin receptor 1, and adiponectin receptor 2, in the bovine corpus luteum (CL) during different phases of the estrous cycle (on days 1–2, 3–4, 5–7, 8–12, 13–18, >18) and pregnancy (at months 1–2, 3–4, 5–7, >7). The mRNA expression was measured by reverse transcription polymerase chain reaction (RT-qPCR). The mRNA expression levels were normalized to the geometric mean of all three constantly expressed reference genes (cyclophilin A, ubiquitin, ubiquitin C). Our findings suggest that adipokines are expressed and present in all investigated groups, and are specifically up- or downregulated during the estrus cycle and during pregnancy. Vaspin and adiponectin levels were upregulated in the middle and late cycle stages. Resistin was abundant during the CL regression stage and in the first months of pregnancy. The specific expression of adipokine receptors indicates their involvement in the local mechanisms that regulate CL function. Further investigations are required to elucidate the regulative mechanisms underlying the different local effects of adipokines on the ovarian physiology of cows.
... Resistin has been found to affect reproductive activity in males and females, in addition to its direct association with various metabolic disorders like diabetes and obesity (Tovar et al., 2005). Due to the presence of resistin in the ovarian granulosa cells of cows, it can potentially modify steroidogenesis and cellular proliferation making it a possible metabolic signal for regulating reproductive activity (Maillard et al., 2011). Owing to its involvement in oocytes' steroidogenesis, proliferation, maturation, and apoptosis, resistin (RETN) gene has been demonstrated to play a significant role in animal production (Rak-Mardyła et al., 2013). ...
Article
Full-text available
Resistin is one of the most important adipocytokines in mammalian cells due to its involvement in insulin resistance, obesity, and autoimmune diseases. Resistin is encoded by RETN gene that is primarily expressed in adipose tissues. Mutations in this gene have been associated with several productive traits in animals. This study was conducted to assess the possible biomarker capacity of RETN by evaluating its association with growth traits in two economically important sheep in the Middle East. Genotyping was conducted using PCR-single strand conformation polymorphism (SSCP), and the polymorphism of RETN was associated with several growth traits for three months intervals starting from birth until one year of age. In a total of 190 Karakul sheep and 245 Awassi sheep, only one SNP (233A > C) was detected in the RETN gene. The identified novel SNP showed significant associations with all growth traits at the ages of six, nine, and twelve months. At the age of six months onward, lambs with AC and CC genotypes showed respectively lower body weight and length, chest and abdominal circumferences, and wither and rump heights than those with AA genotype. Due to the remarkable association between RETN;233A > C and lower growth traits, this genotype is suggested as a promising marker to assess growth traits in Karakul and Awassi sheep. This is the first study that demonstrated the importance of RETN as a possible tool for evaluating growth traits in two breeds of sheep with a possibility to be applied to other breeds via large-scale association analysis.
... Ovaries and adipose tissue have a close relationship via adipokine actions Nikanfar et al., 2021;Reverchon et al., 2014). Several adipokines have been reported to regulate ovarian steroidogenesis, including leptin (Dbrannian et al., 1999;Spicer and Francisco, 1997), adiponectin (Chabrolle et al., 2009;Lagaly et al., 2008), resistin (Maillard et al., 2011;Spicer et al., 2011), chemerin Wang et al., 2012), visfatin (Reverchon et al., 2013, irisin (Poretsky et al., 2017;Ulker et al., 2020), and apelin (Rak et al., 2017;Roche et al., 2016). ...
Article
Asprosin is an adipokine synthesized by the white adipose tissue that regulates glucose homeostasis and that has been reported to affect bovine theca cell function and follicular growth, but its role on granulosa cell functions remains to be unveiled. Hence, the objective of this study was to investigate asprosin impacts on granulosa cell steroidogenesis. Bovine granulosa cells from small ovarian follicles were cultured in vitro to investigate the effects of asprosin on cell proliferation, production of steroids, mRNA abundance of genes that encode steroidogenic enzymes and cell cycle regulators, and protein relative abundance of steroidogenic signaling pathways. Asprosin was shown to affect granulosa cell functions in a dose-dependent manner. In the presence of FSH, asprosin enhanced estradiol production and stimulated an increase in mRNA expression of FSHR and CYP19A1 in a dose-dependent manner. In the presence of IGF1, asprosin decreased estradiol production, increased progesterone production, altered PKA relative protein expression, and tended to alter the ratio of p-ERK1/2/total ERK1/2 protein expression in a dose-dependent manner. Furthermore, asprosin increased p-53 gene expression in basal culture conditions and with or without FSH and IGF1. Taken together, findings of this study show that asprosin is a regulator of granulosa cell functions and the effects of asprosin depend on dose and cell culture conditions.
... Although the impact of latter three adipokines on dairy cows' metabolism is well documented (De Koster et al., 2017), the metabolic role of resistin in dairy cows is not yet fully explored. Previous in vitro reasearch demonstrated that resistin activates AMP-activated protein kinase (AMPK) signaling pathway in bovine granuslosa cells (Maillard et al., 2011), whereas it induces lipolysis in bovine adipocytes (Reverchon et al., 2014). ...
Article
A number of alterations have been identified in lipid metabolism within adipose tissue and liver in obesity. Less is known about the capacity of skeletal muscle for the metabolism of fatty acids in obesity-related insulin resistance, though it is evident that dry cow muscles may contain increased triglyceride content. The current study was therefore undertaken to evaluate the skeletal muscle expression of proteins of the fatty acid metabolism in dry cows with different body condition scores (BCS). Sixteen Holstein-Friesian close-up cows were divided into 2 equal groups based on their BCS as optimal (3.25 ≤ BCS ≤ 3.5) and high (4.0 ≤ BCS ≤ 4.25). Blood samples collection and skeletal muscle biopsies were carried out at day 10 before calving. Blood serum was assayed for concentration of resistin using a bovine specific ELISA. Protein expression of insulin receptor beta subunit (IRβ), glucose transporter 4 (GLUT4), fatty acid translocase (FAT/CD36), fatty acid transporter 1 (FATP1), carnitine palmitoyltransferase 1 (CPT1), AMP-acitvated protein kinase (AMPK) and lipin 1 were analyzed in semitendinosus muscle by immunoblot. Resistin differed non-significantly between high-BCS and optimal-BCS cows. Insulin-resistant lipid metabolism in obese cows was paralleled with increased skeletal muscle expression of lipin 1 and GLUT4, and decreased expression of IRβ and FATP1. These data suggest that in obesity-related insulin resistance, metabolic capacity in dry cow skeletal muscles appears to be organized towards the synthesis of signaling intermediates rather than fatty acids oxidation and that altered fatty acid uptake does not contribute to this disposition.
Article
Background: Obesity is a complex multifactorial disease in which the accumulation of excess body fat has adverse health effects, as it can increase the risk of several problems, including infertility, in both men and women. Obesity and infertility have risen together in recent years. Against this background, the present review aims to highlight the impact of obesity on infertility and the underlying pathophysiology of obesity-related infertility (ORI) in men and women, and to provide readers with knowledge of current trends in the effective development of phytotherapeutics for its treatment. Methods: We thoroughly searched in PubMed, MEDLINE, Scopus, EMBASE, and Google Scholar to find all relevant papers on ORI and the therapeutic effects of phytotherapeutics on ORI in men and women. Results: The extensive search of the available literature revealed that obesity affects reproductive function through several complex mechanisms such as hyperlipidaemia, hyperinsulinaemia, hyperandrogenism, increased body mass index, disruption of the hormonal milieu, systemic inflammation, oxidative stress, alterations in epigenetics and dysbiosis. On the other hand, several studies reported that phytotherapeutics has a broad therapeutic spectrum of action by improving sex hormone homeostasis, ovarian dysfunction, menstrual cycle and inhibiting ovarian hyperplasia, as well as down-regulating ovarian apoptosis, inflammation and oxidative stress, and controlling metabolic dysfunction in obese women. Male infertility is also addressed by phytotherapeutics by suppressing lipogenesis, increasing testosterone, 3β-HSD and 17β-HSD levels, improving sperm parameters and attenuating testicular dyslipidaemia, oxidative stress, inflammation and germ cell apoptosis. Conclusions: In the present review, we discussed the effects of obesity on reproductive dysfunction in men and women and the underlying pathophysiology of ORI. In addition, the therapeutic effect of phytotherapeutics against ORI was highlighted.
Article
Postnatal ovarian and uterine development is crucial to accomplished female fertility. Thus, the investigations of factors that present in pre‐pubertal stages are important as it might be responsible for the regulation of ovarian and uterine function. Apelin, an adipokine and its receptor (APJ) are present in female reproductive organs. However, no study has reported its postnatal expression in uterus and ovary. Thus, we investigated the postnatal developmental changes in expression and localization of apelin and APJ in the ovary and uterus of mice. Postnatal ovary and uterus were collected from postnatal day (PND) 1, 7, 14, 21, 42, 65 and performed western blot analysis and immunohistochemistry. Uterine APJ is elevated in PND14 and PND65, whereas, ovarian APJ elevated in PND7, PND14, and PND65. Apelin expression in both ovary and uterus showed intense staining at PND65 and PND14. Our results showed that apelin and APJ abundance was lower at PND21 in uterus and ovary. In conclusion, apelin and APJ are developmentally regulated in the ovary and uterus, and its localization in the different compartments of ovary and uterus suggest its distribution specific physiological role in the uterus and ovary.
Article
Psoriasis is a chronic genetically determined disease of a multifactorial nature associated with immune-mediated inflammation and characterized by a recurrent course with frequent associative damage to other organs and systems. According to global recommendations, nowadays, despite the era of biologic therapy, phototherapy (PUVA and UVB) continues to occupy its important niche in the treatment of moderate-to-severe psoriasis, due to the pathogenetically justified therapeutic effect, safety and low cost of procedures. The review provides detailed data on the mechanism of action, the effectiveness of phototherapy; as well as potential biomarkers of psoriasis (calprotectin, lipocalin 2, resistin), which can enhance an accuracy assessment of the effectiveness of the treatment and, if necessary, its timely correction, which will allow faster achievement of the desired effect resulting in improvement of the quality of patients lives.
Article
Full-text available
We investigated the chronic in vivo effect of resistin on insulin sensitivity and glucose metabolism by overexpressing resistin protein in male Wistar rats using intravenous administration of an adenovirus encoding mouse resistin. After 7 days of elevated resistin levels at a supraphysiological concentration, the animals displayed glucose intolerance and hyperinsulinemia during glucose tolerance tests, and insulin tolerance tests demonstrated an impaired glucose-lowering effect of insulin. The glucose clamp studies were performed at submaximal (4 mU/kg/min) and maximal (25 mU/kg/min) insulin infusion rates and demonstrated the presence of insulin resistance induced by elevated resistin levels. Indeed, the insulin-stimulated glucose infusion rate was decreased by 12-31%; suppression of hepatic glucose output was attenuated by 28-55%; and insulin suppression of circulating FFA levels was inhibited by 7%. Insulin receptor substrate-1 and -2 phosphorylation and Akt activation were impaired in muscle and adipose tissue. Interestingly, activation of AMP-activated protein kinase in skeletal muscle, liver, and adipose tissue was also significantly downregulated. Together, these results indicate that chronic "hyper-resistinemia" leads to whole-body insulin resistance involving impaired insulin signaling in skeletal muscle, liver, and adipose tissue, resulting in glucose intolerance, hyperinsulinemia, and hypertriglyceridemia. Thus elevated resistin levels in normal rats fed a regular chow diet produce many of the features of human syndrome X.
Article
Full-text available
Adiponectin is an adipokine, mainly produced by adipose tissue. It regulates several reproductive processes. The protein expression of the adiponectin system (adiponectin, its receptors, AdipoR1 and AdipoR2 and the APPL1 adaptor) in bovine ovary and its role on ovarian cells and embryo, remain however to be determined. Here, we identified the adiponectin system in bovine ovarian cells and embryo using RT-PCR, immunoblotting and immunohistochemistry. Furthermore, we investigated in vitro the effects of recombinant human adiponectin (10 micro g/mL) on proliferation of granulosa cells (GC) measured by [3H] thymidine incorporation, progesterone and estradiol secretions measured by radioimmunoassay in the culture medium of GC, nuclear oocyte maturation and early embryo development. We show that the mRNAs and proteins for the adiponectin system are present in bovine ovary (small and large follicles and corpus luteum) and embryo. Adiponectin, AdipoR1 and AdipoR2 were more precisely localized in oocyte, GC and theca cells. Adiponectin increased IGF-1 10(-8) M-induced GC proliferation (P < 0.01) but not basal or insulin 10(-8) M-induced proliferation. Additionally, adiponectin decreased insulin 10(-8) M-induced, but not basal or IGF-1 10(-8) M-induced secretions of progesterone (P < 0.01) and estradiol (P < 0.05) by GC. This decrease in insulin-induced steroidogenesis was associated with a decrease in ERK1/2 MAPK phosphorylation in GC pre-treated with adiponectin. Finally, addition of adiponectin during in vitro maturation affected neither the percentage of oocyte in metaphase-II nor 48-h cleavage and blastocyst day 8 rates. In bovine species, adiponectin decreased insulin-induced steroidogenesis and increased IGF-1-induced proliferation of cultured GC through a potential involvement of ERK1/2 MAPK pathway, whereas it did not modify oocyte maturation and embryo development in vitro.
Article
Resistin (RETN) is a cytokine that has been identified in adipose tissue and macrophages and implicated in insulin resistance. Recently, RETN mRNA was identified in Leydig cells of male rats. Some studies have shown elevated concentrations of serum RETN in women with polycystic ovaries. These findings led us to hypothesize that RETN may be expressed in the rat ovary. The objectives were to determine if: 1) RETN mRNA is expressed in the rat ovary, 2) RETN mRNA fluctuates throughout the estrous cycle, and 3) RETN mRNA expression changes in rats with induced ovarian cysts. Experiment 1 - RETN Expression During the Estrous Cycle Long-Evans rats were monitored by daily vaginal cytology to determine cycle stage. After five weeks, rats were assigned to one of three groups: diestrus I & II (D, n=5), proestrus (P, n=5), and estrus (E, n=6). Animals were anesthetized, decapitated, and ovaries dissected and frozen at -70°C for later analyses. RNA was extracted from ovaries and reverse transcribed using RNeasy Midi and QuantiTect Reverse Transcription kits (Qiagen). Ovarian RETN mRNA was quantified by real time PCR using Taqman probes for rat RETN and rat B-actin mRNA (internal control). The threshold cycle number (Ct, when sample signal exceeds background) was used to quantify mRNA. Data were normalized to B-actin by calculating the delta Ct (RETN Ct - actin Ct). Lower values indicated higher RETN mRNA expression. The average delta Cts were: D=10.7, P=11.8, and E=11.1. We determined fold changes between groups using an algorithm that accounts for the doubling of cDNA with each PCR cycle (P vs E=1.6, P vs D=2.1, E vs D=1.4). While RETN mRNA was detected in all rat ovaries throughout the cycle, we were unable to detect significant differences between stages. Experiment 2 - RETN Expression in Cystic Ovaries: To induce ovarian cyst formation, the experimental group (n=4) received 0.04% propylthiouracil in drinking water for 30 days to cause hypothyroidism. On days 11-30, they also received daily s.c. injections of 10 IU hCG in 0.1 mL of saline. Control animals (n=4) received normal drinking water for 30 days and daily injections of 0.1 mL of saline on days 11-30. All ovaries from rats in the hypothyroid/hCG group had large cystic follicles; whereas none of the euthyroid ovaries exhibited evidence of cysts. This was supported by higher ovarian weight in hypothyroid/hCG-treated rats (mean=505 ± 52mg) compared to controls (mean=105 ± 17mg). Results of RT-PCR showed an average delta Ct of 7.2 in rats with cystic ovaries compared with 9.1 in control animals, indicating that the hypothyroid/hCG-treated group had 3.8-fold higher RETN mRNA expression. Although we were able to detect RETN mRNA in rat ovaries, we could find no significant differences in RETN expression throughout the estrous cycle. RETN expression, as quantified by RT-PCR, was significantly higher in the induced-cyst rats than in the normally cycling controls. Our results indicate that RETN is normally expressed at a basal level throughout the estrous cycle and is elevated in animals with induced ovarian cysts. The potential involvement of RETN in normal ovarian physiology, as well as the process of cyst induction, clearly merit further investigation. Future studies should include localization of RETN in the ovary, quantification of RETN protein and studies of its regulation by the endocrine syste. (poster)
Article
During the last decade, involvement of growth hormone (GH), insulin-like growth factors (IGFs) and IGF binding proteins (IGFBPs) in ovarian folliculogenesis has been extensively studied. This review provides an update on the GH, IGF system and their role in ovarian follicular development. In vitro studies and knockout experiments demonstrated an important role of GH in preantral follicle growth and differentiation through their binding with GH receptors, which are located both in the oocyte and follicular somatic tissues. Furthermore, GH stimulates the development of small antral follicles to gonadotrophin-dependent stages, as well as maturation of oocytes. With regard to the IGF system, IGF-I has no effects on primordial follicle development, but both IGF-I and IGF-II stimulate growth of secondary follicles. Depending on the species studies and method used, these proteins have been detected in oocytes and/or somatic cells. In antral follicles, these IGFs stimulate granulosa cell proliferation and steroidogenesis in most mammals. The bioavailability of IGFs is regulated by a family of intrafollicular expressed IGF binding proteins (IGFBPs). Facilitation of IGF can be increased through the activity of specific IGFBP proteases, which degrade the IGF/IGFBP complex, resulting in the production of IGFBP fragments and release of attached IGF.
Article
Use of the real-time polymerase chain reaction (PCR) to amplify cDNA products reverse transcribed from mRNA is on the way to becoming a routine tool in molecular biology to study low abundance gene expression. Real-time PCR is easy to perform, provides the necessary accuracy and produces reliable as well as rapid quantification results. But accurate quantification of nucleic acids requires a reproducible methodology and an adequate mathematical model for data analysis. This study enters into the particular topics of the relative quantification in real-time RT-PCR of a target gene transcript in comparison to a reference gene transcript. Therefore, a new mathematical model is presented. The relative expression ratio is calculated only from the real-time PCR efficiencies and the crossing point deviation of an unknown sample versus a control. This model needs no calibration curve. Control levels were included in the model to standardise each reaction run with respect to RNA integrity, sample loading and inter-PCR variations. High accuracy and reproducibility (<2.5% variation) were reached in LightCycler PCR using the established mathematical model.
Article
The Ob gene product, leptin, is secreted by adipocytes and is required for fertility in the mouse. Leptin-deficient mice are obese and infertile, symptoms reminiscent of polycystic ovary syndrome (PCOS). Prior studies have shown that serum leptin levels are elevated in a significant sub-population of anovulatory women with PCOS, suggesting that elevated leptin levels may adversely affect ovarian function. Since leptin receptor mRNA has been detected in the ovary, this study was designed to test the hypothesis that leptin may impair granulosa cell (GC) estradiol-17β (E2) production by a direct mechanism. GC were isolated from the ovaries of 26-day-old Sprague-Dawley rats, and were cultured (60,000 GC/well) in 96-well plates in the presence and absence of ovine FSH (0.001–100 ng/ml) and androstenedione (0.1 μM), with and without recombinant murine leptin (0.1–100 ng/ml) for 48 h. Leptin alone had no effect on E2 production. FSH caused a dose-related increase in E2 production by GC (ED50 = 1.9 ± 0.4 ng/ml). Addition of leptin did not alter FSH-stimulated E2 levels. Concomitant treatment with FSH and IGF-I (30 ng/ml) augmented maximal FSH-dependent E2 production five-fold. Leptin caused a dose-dependent (IC50 = 2.7 ± 0.6 ng/ml) inhibition (30–50%) of the IGF-I increase in FSH-stimulated E2 production. The inhibitory effect of leptin was specific for E2 production since there was no effect on basal, FSH-, or FSH + IGF-I-dependent progesterone levels. The results of this study demonstrate that leptin can directly impair the IGF-I-mediated augmentation of FSH-stimulated E2 synthesis by GC. These data raise the possibility that high leptin levels may contribute to infertility in some women with PCOS by counteracting the sensitizing effects of IGF-I in dominant follicles.
Article
The aim of our studies was to compare the roles of leptin and ghrelin in the direct control of proliferation, apoptosis, and secretory activity by porcine ovarian cells. In our in vitro experiments, we analyzed the effects of leptin and ghrelin treatments (at 0, 1, 10, or 100ng/mL medium) on the accumulation of proliferation-related peptides (PCNA, cyclin B1, MAP kinase [MAPK]) and apoptosis-associated peptides (Bax, caspase 3, p53), and on progesterone secretion by cultured porcine granulosa cells, using immunocytochemistry, SDS PAGE-Western immunoblotting, and radioimmunoassay (RIA). Leptin stimulated proliferation (PCNA, cyclin B1, MAPK), apoptosis (Bax, p53), and progesterone secretion. Ghrelin promoted proliferation (PCNA, cyclin B1, MAPK) and progesterone secretion but suppressed apoptosis (Bax, caspase 3, p53). These observations suggest that both leptin and ghrelin directly control proliferation, apoptosis, and secretory activity by porcine ovarian cells. At the level of the ovary, in contrast to the hypothalamo-hypophysial system, leptin and ghrelin may have similar action in promoting granulosa cell proliferation and progesterone secretion, but they may be antagonistic to one another (leptin, stimulator; ghrelin, inhibitor) in controlling apoptosis.
Article
Resistin is a newly discovered adipocyte-derived cytokine that may play an important role in insulin resistance, diabetes, adipogenesis, inflammation, and cardiovascular disease. However, it is largely unknown whether resistin impairs endothelial functions by affecting the endothelial nitric oxide synthase (eNOS) system. In this study, we determined the effect of human recombinant resistin protein on eNOS expression and regulation in human coronary artery endothelial cells (HCAECs). When cells were treated with clinically relevant concentrations of resistin (40 or 80 ng/ml) for 24 h, the levels of eNOS mRNA, protein, and activity and eNOS mRNA stability were significantly reduced. Cellular nitric oxide levels were also decreased. In addition, the cellular levels of reactive oxygen species (ROS), including superoxide anion, were significantly increased in resistin-treated HCAECs. Mitochondrial membrane potential and the activities of catalase and superoxide dismutase were reduced. Three antioxidants, seleno-L-methionine, ginsenoside Rb1, and MnTBAP (superoxide dismutase mimetic), effectively blocked resistin-induced eNOS downregulation. Meanwhile, resistin activated the mitogen-activated protein kinases p38 and c-Jun NH(2)-terminal kinase (JNK), and the specific p38 inhibitor SB-239063 effectively blocked resistin-induced ROS production and eNOS downregulation. Furthermore, immunoreactivity of resistin was increased in atherosclerotic regions of human aorta and carotid arteries. Thus resistin directly induces eNOS downregulation through overproduction of ROS and activation of p38 and JNK in HCAECs. Resistin-induced mitochondrial dysfunction and imbalance in cellular redox enzymes may be the underlying mechanisms of oxidative stress.
Article
Gestational exposure to maternal overweight (OW) influences the risk of obesity in adult life. Male offspring from OW dams gain greater body weight and fat mass and develop insulin resistance when fed high-fat diets (45% fat). In this report, we identify molecular targets of maternal OW-induced programming at postnatal d 21 before challenge with the high-fat diet. We conducted global transcriptome profiling, gene/protein expression analyses, and characterization of downstream signaling of insulin and adiponectin pathways in conjunction with endocrine and biochemical characterization. Offspring born to OW dams displayed increased serum insulin, leptin, and resistin levels (P < 0.05) at postnatal d 21 preceding changes in body composition. A lipogenic transcriptome signature in the liver, before development of obesity, was evident in OW-dam offspring. A coordinated locus of 20 sterol regulatory element-binding protein-1-regulated target genes was induced by maternal OW. Increased nuclear levels of sterol regulatory element-binding protein-1 and recruitment to the fatty acid synthase promoter were confirmed via ELISA and chromatin immunoprecipitation analyses, respectively. Higher fatty acid synthase and acetyl coenzyme A carboxylase protein and pAKT (Thr(308)) and phospho-insulin receptor-beta were confirmed via immunoblotting. Maternal OW also attenuated AMP kinase/peroxisome proliferator-activated receptor-alpha signaling in the offspring liver, including transcriptional down-regulation of several peroxisome proliferator-activated receptor-alpha-regulated genes. Hepatic mRNA and circulating fibroblast growth factor-21 levels were significantly lower in OW-dam offspring. Furthermore, serum levels of high-molecular-weight adiponectin (P < 0.05) were decreased in OW-dam offspring. Phosphorylation of hepatic AMP-kinase (Thr(172)) was significantly decreased in OW-dam offspring, along with lower AdipoR1 mRNA. Our results strongly suggest that gestational exposure to maternal obesity programs multiple aspects of energy-balance regulation in the offspring.