Sunteți pe pagina 1din 14

Horm Mol Biol Clin Invest 2015; 24(1): 1124

Jolle Dupont*, Xavier Pollet-Villard, Maxime Reverchon, Namya Mellouk and Rachel Levy

Adipokines in human reproduction

Introduction
DOI 10.1515/hmbci-2015-0034
Received July 27, 2015; accepted October 5, 2015; previously published
online November 17, 2015
In a few decades, our view of adipose tissue has changed
Abstract: Adipose tissue communicates with other central from lipid storage to an endocrine tissue [1]. In fact,
and peripheral organs by the synthesis and release of sub- white adipose tissue may be the largest endocrine tissue
stances called adipokines. The most studied adipokine is in humans and is currently considered as an endocrine
leptin but others have been recently identified including organ in and of itself, secreting more than 600 potential
resistin, adiponectin, chemerin, omentin and visfatin. cell signaling mediators [2]. It communicates with other
These adipokines have a critical role in the development central and peripheral organs by synthesis and release of
of obesity-related complications and inflammatory condi- substances from adipocytes, adipocyte progenitors, mac-
tions. However, they are also involved in other functions rophages or stroma cells. These substances, called adi-
in the organism including reproductive functions. Indeed, pokines, include cytokines, hormones, growth factors,
many groups have demonstrated that adipokine receptors, chemokines, complement factors, and proteins involved
such as adiponectin and chemerin, but also adipokines in various functions including lipid metabolism, glucose
themselves (adiponectin, chemerin, resistin, visfatin and homeostasis, angiogenesis, haemostasis, inflammation
omentin) are expressed in human peripheral reproduc- and regulation of blood pressure. The plasma levels of
tive tissues and that these adipokines are likely to exert some adipokines are associated to specific metabolic
direct effects on these tissues. After a brief description of states [3]. For example, a dysregulation of plasma adi-
these new adipokines, an overview of their actions in dif- pokines is implicated in obesity, type 2 diabetes and
ferent human reproductive organs (hypothalamus, pitui- hypertension. Obesity is a risk factor for infertility and
tary, ovary, testis, uterus and placenta) will be presented. adverse reproductive outcomes in both men and women.
Finally, comments will be made on the eventual altera- In male patients, there is evidence that being obese or
tions of these adipokines in reproductive disorders, with overweight negatively affects reproductive potential,
special attention to polycystic ovary syndrome, a disease not only by reducing sperm quality, but also by altering
characterized by dysfunction of gonadal axis and systemic the physical and molecular structure of germ cells in the
nerve endocrine metabolic network with a prevalence of testes and consequently affecting the maturity and func-
up to 10% in women of reproductive age. tion of sperm cells [4]. In female patients, obesity can
affect the reproductive tissue at different levels: oocyte
Keywords: adipose tissue; endocrine signaling; fertility;
maturation, embryo development and implantation, pla-
human; male and female.
centa and uterine environment [5]. Furthermore, interest
in the reproductive endocrine aspects of adipose tissue
has increased partly due to interest in polycystic ovary
*Corresponding author: Dr. Jolle Dupont, INRA, UMR85, Unit syndrome (PCOS), one of the most investigated and prev-
Physiologie de la Reproduction et des Comportements, Nouzilly, alent endocrinopathies in women of reproductive age
France; CNRS, UMR7247, Nouzilly, France; Universit Franois [6]. PCOS is manifested by hyperandrogenism, polycys-
Rabelais, Tours, France; and LInstitut franais du cheval et de tic ovaries on ultrasound, oligomenorrhoea and anovu-
lquitation (IFCE), Nouzilly, France, Phone: +33 2 47 42 77 89,
lation. Excess adipose tissue is present in most of PCOS
Fax: +33 2 47 42 77 43, E-mail: jdupont@tours.inra.fr
Xavier Pollet-Villard: Service de Medecine de la Reproduction, patients [7].
Hopital Tenon (AP-HP), 4 Rue de la Chine, 75020, Paris It has been shown that adipokines are functionally
Maxime Reverchon and Namya Mellouk: INRA, UMR85, Unit involved at different levels of the reproductive axis includ-
Physiologie de la Reproduction et des Comportements, Nouzilly, ing the gonad and hypothalamic-pituitary axis. Many
France; CNRS, UMR7247, Nouzilly, France; Universit Franois
studies described the presence and the role of the adi-
Rabelais, Tours, France; and LInstitut franais du cheval et de
lquitation (IFCE), Nouzilly, France
pokines and their receptors in the male and female repro-
Rachel Levy: Service de Medecine de la Reproduction, Hopital ductive tract of different species including humans. These
Tenon (AP-HP), 4 Rue de la Chine, 75020, Paris adipokines regulate ovarian steroidogenesis, oocyte

Brought to you by | Universidade Federal do Esprito Santo UFES


Authenticated
Download Date | 7/12/17 9:08 PM
12Dupont etal.: Adipokines and human reproductive organs

maturation, and embryo development. They are also described as metabolic endotoxemia, may play a role in
present in the uterus and placenta. Since the discovery obesity-related metabolic disorder [27]. It has also been
of leptin in 1994, other new adipokines synthesized and hypothesized that activation of TLR-4 could positively reg-
released by the adipose tissue have been identified. Here, ulate the expression of resistin through NF-B, enabling
we review the recent progress regarding the role of some an autoregulation of resistin secretion through this recep-
of these new adipokines including resistin, adiponectin, tor [28]. Other receptors have been described for resistin,
chemerin, omentin, and visfatin in the human reproduc- such as a cytosolic protein called adenylyl cyclase associ-
tive organs. In human, these new adipokines have not ated protein 1 (CAP-1) in monocytes [29]. Also, an isoform
been extensively studied at the hypothalamus-pituitary of decorin, a proteoglycan of the extracellular matrix
axis and at the cellular level in the testis. Thus, for these acting as a pan-receptor tyrosine kinase inhibitor [30],
latter organs, we will report some data obtained in animal has been described as a receptor for resistin in adipocyte
models. progenitors [31]. It presents as a membrane protein most
likely generated by proteolytic cleavage of decorin and is
referred to as delta-decorin. Activation of delta-decorin

Resistin by resistin promotes adipocyte progenitor migration and


proliferation and impedes lipid droplet accumulation in
mice [31]. Lastly, resistin inhibits the activity of receptor
Resistin is a 12.5 kDa adipokine belonging to a family
tyrosine kinase-like orphan receptor 1 (ROR1) in murine
of cysteine-rich protein known as resistin-like mole-
pre-adipocytes [32], a receptor implicated in non canoni-
cules (RELM) or found in the inflammatory zones (FIZZ)
cal Wnt signaling involving Wnt5a as an activating ligand
proteins, characterized in 2001 by 3 different research
[33]. The binding of resistin to ROR1 promotes adipogen-
teams [810]. Resistin is secreted by adipocytes and
esis and inhibits glucose cellular uptake in murine pre-
functionally impairs glucose metabolism, inducing
adipocytes [32].
insulin resistance [9]; it has also been associated with
proinflammatory changes in the vascular endothelium
[11], pathways linked to various inflammatory diseases
[12] and angiogenesis [13]. In humans, resistin is mainly Ovary
produced by macrophages, rather than adipocytes,
and is implicated mostly in inflammatory pathways Recent studies have demonstrated the presence of resis-
[14]. Animal studies have demonstrated a contributory tin in human granulosa [34, 35], as well as in cumulus,
function in testosterone production and pubertal hypo- theca cells and oocytes [35]. Also, possible positive
thalamicpituitary signaling, attributing a role as an feedback through luteinizing hormone (LH) and pro-
endocrine mediator of the reproductive axis [1517] and gesterone and negative feedback through E2 (estradiol)
a potential one concerning obesity-associated insulin have been postulated [15]. In biological terms, resistin
resistance [9, 18]. In reproduction, there has been a con- reduces the secretion of both P450scc and P450 aro-
troversy on its possible linkage with anovulatory PCOS matase protein levels, and IGF-IR signaling in response to
[1921], or non PCOS women undergoing in vitro ferti- IGF-I in human granulosa cells ([35], Figure 1). In human
lization (IVF) [22], suggesting that resistin might have thecal cells in culture, resistin increases the expression
negative effects on pregnancy [23, 24]. In women under- of 17--hydroxylase in the presence of forskolin and/
going IVF, it was demonstrated that oocyte and embryo or forskolin and insulin, suggesting a role of resistin in
number correlated positively with resistin levels in fol- androgen production ([16], Figure 1). Receptors to resistin
licular fluid [25]. may be found in the ovary, although data are scarce and
Although no specific receptor for resistin has been no direct interactions have been reported to date. TLR-4
described, resistin is a ligand to several putative recep- is expressed in bovine granulosa cells and promotes
tors. Resistin is an agonist of toll like receptor-4 (TLR-4) inflammation and oocyte meiotic progression disruption
and competes for binding with its pathogen-associated [36]. Decorin is a constituent of the extracellular matrix
molecular pattern well-known ligand, the endotoxin of the ovary in primates and may regulate folliculogen-
lipopolysaccharide (LPS). It is also able to trigger the esis, ovulation, and survival of the corpus luteum [37],
inflammatory pathways associated with TLR-4 activation but no published studies mentioned delta-decorin in
[26]. Interestingly, a low level elevation of gut-derived the ovary. ROR1 is expressed in ovarian cancer and may
LPS has been associated with obesity. This phenomenon, represent a bad prognosis marker, but ROR1 staining has

Brought to you by | Universidade Federal do Esprito Santo UFES


Authenticated
Download Date | 7/12/17 9:08 PM
Dupont etal.: Adipokines and human reproductive organs13

Follicle

Ovary
Uterus

Uterus Granulosa Oocyte Theca


cells cells
Endometrial stromal E2, Pg
Adiponectin IVM (pig [74], T (pig [139])
cell proliferation [87] in response
Mouse [70]) Pg and and rostedione
to IGF-1 [69]
in response to insulin (bovine) [138]
E2, Pg
nd in response Competency
Visfatin nd
to IGF-1 [128] (aged mice) [135]
E2, Pg IVM (cow [95])
Chemerin nd in response
to IGF-1 [94] nd

Apoptosis [97]

Resistin nd E2, Pg nd
in response 17 hydroxylase
to IGF-1 [35] activity insulin induced [16]
E2, Pg
Omentin nd nd nd
in response
to IGF-1 [125]

Figure 1:In vitro effect of chemerin, adiponectin, resistin, visfatin and omentin on ovarian follicle cells.
When the data were not from human studies, the species was indicated in brackets. nd, Undetermined; , decreases; , increases;
IVM, in vitro maturation; T, testosterone; Pg, progesterone; E2, oestradiol.

been also reported in healthy ovarian tissue [38]. Interest- resistin exerts its proinflammatory effects and promotes
ingly, CAP-1 is overexpressed in human epithelial ovarian insulin resistance through the binding to TLR-4 in the rat
cancer, where it promotes cell proliferation and is associ- hypothalamus [41]. Also, decorin is secreted by follicu-
ated with poor outcomes. lostellar cells and pericytes in the rat anterior pituitary
glands [42]. However, the role of resistin in the hypo-
thalamus and the pituitary remains to be determined in
Pituitary-Hypothalamus humans.

To control the functions of reproduction, resistin may


also act on the hypothalamic-pituitary axis. Indeed, Testis
it is present in the hypothalamus and pituitary gland
in humans and rodents [39]. In the pituitary, resistin In humans, no studies about the localization of resis-
expression is low at birth in rats and increases until the tin in testis have been performed. In rat testis, resistin
age of 28 days. It increases during weaning in females is localized in the Leydig and Sertoli cells and in the
and during puberty in males. Moreover, its concentration seminiferous tubules. Its expression is regulated by
in the pituitary is two to three times higher in males than fasting, gonadotropins and leptin. Moreover, it varies
in females. On the other hand, resistin decreases expres- during postnatal development peaking in adulthood
sion of AdipoR1 and AdipoR2 receptors in rat pituitary [43]. Also, the expression of resistin in mouse testes
cells in culture [40]. Recently, it has been shown that may be directly or indirectly regulated by RHOX-5, an

Brought to you by | Universidade Federal do Esprito Santo UFES


Authenticated
Download Date | 7/12/17 9:08 PM
14Dupont etal.: Adipokines and human reproductive organs

androgen receptor and androgen-dependent transcrip- Placenta


tion factor mainly expressed in Sertoli cells and belong-
ing to a large X-linked homeobox gene cluster selectively Resistin was identified in the human placenta [50]. In
expressed in reproductive organs. Interestingly, Rhox- human, resistin is mainly expressed by trophoblast cells.
5 null mice exhibit impaired insulin signaling in testis Resistin is also present in the amniotic membrane. The
and an enhancement of germ cell apoptosis and show expression pattern of placental resistin is not yet known.
striking resemblances to a strain of infertile male mice However, a study shows that resistin is able to modulate
characterized by a defect in the Ins2 gene. The fact that glucose transport in human trophoblast cells [51]. Resis-
Rhox-5 null mice exhibit a significant up-regulation of tin, by interfering with placental development, may affect
resistin expression in testicular tissue and in Leydig cells pregnancy outcome and fetal growth. Strikingly, ROR-
during postnatal development compared to wild-type Wnt5a signaling has recently been implicated in decidu-
controls indicates that Rhox-5 inhibits resistin expres- alization and implantation as the conditional deletion of
sion indirectly in interstitial cells and perhaps directly ROR and/or Wnt5a in female mice led to poor placentation
in Sertoli cells [44]. Although delta-decorin has not yet and pregnancy outcomes [52].
been described in testis, decorin is secreted by human
peritubular cells surrounding seminiferous tubule and
high levels of decorin in the extracellular matrix of the Adiponectin
tubular wall is associated with testis inflammation,
fibrosis and male infertility in humans, rhesus monkeys Adiponectin is a protein of 244 amino acids mainly pro-
and mice [45]. duced by white adipose tissue but also found in other tissues
such as bone and muscle [53, 54]. Adiponectin is involved
in lipid and carbohydrate metabolism, and appears to
Semen play a key role in the pathophysiology of obesity, type 2
diabetes and coronary heart disease. Contrary to resistin,
Resistin has been found in human seminal plasma and adiponectin seems to exert anti-inflammatory and anti-
this seminal resistin correlated positively with IL-6 and diabetic effects [1]. In a healthy man, plasma adiponectin
elastase levels, two markers of semen inflammation is about 530 mg/L, which represents 0.01% of the total
[46]. In the same study, seminal resistin levels were not plasma protein. Low adiponectin is predictive of insulin
correlated with semen quality, BMI or plasma testos- resistance or type 2 diabetes. Serum adiponectin concen-
terone and FSH levels. However, Thomas et al. showed tration is also higher in men with normal weights than
that seminal resistin was positively correlated with BMI in overweight or obese men, and significantly positively
and waist circumference but not with semen parameters correlates with total serum testosterone [47]. Consistent
[47]. F
urthermore, seminal resistin was not significantly findings have been reported in bulls, where high fertility
altered in vasectomised patients [46], showing that the individuals presenting with higher serum adiponectin and
main resistin production takes place downstream of the testosterone levels than average and low fertility bulls [55].
vas deferens, possibly by the prostate or seminal vesicles. Plasma adiponectin exists in various molecular forms:
Seminal resistin may thus play a regulatory role in inflam- monomeric and multimeric form, in which adiponectin
mation of the male reproductive tract; this aspect has been monomers are organized in a more complex structure
emphasized in a more recent study [48]. Seminal resistin through disulfide bonds. The multimeric form can be clas-
levels showed negative correlations with sperm motility sified into high molecular weight (HMW), average molecu-
and positive correlations with the proportion of apop- lar weight (MMW) and low molecular weight (LMW). To act
totic and necrotic spermatozoa. Seminal resistin was also at the cellular level, adiponectin primarily binds to two
higher when patients presented with leukocytospermia, receptors, AdipoR1 and AdipoR2, mainly expressed in the
which is commonly considered as a male accessory gland muscle and liver, respectively. The AdipoR receptors have
inflammation marker. An eventual effect of seminal resis- seven transmembrane domains, but have a reversed topol-
tin at the spermatozoa level has not yet been described, ogy compared to the same type of G protein associated
although TLR-4, known as a binding site for resistin, has receptors; the amino-terminal part is intracellular and the
been found in human sperm. Interestingly, in mouse and carboxy-terminal part is extracellular [56]. Once activated,
human spermatozoa, the activation of this receptor by the AdipoR receptor binds an adapter protein, APPL,
endotoxins reduces sperm motility and promotes sperm which relays the activation of adenosine monophosphate-
apoptosis [49]. activated kinase (AMPK) [57]. AMPK is a major component

Brought to you by | Universidade Federal do Esprito Santo UFES


Authenticated
Download Date | 7/12/17 9:08 PM
Dupont etal.: Adipokines and human reproductive organs15

of the signaling pathway involved in the metabolic effects However, low adiponectin expression was detected in
of adiponectin. Another putative receptor for adipokine, human granulosa cells and cumulus cells [69, 70]. The
T-cadherin (CDH13), has been described in smooth muscle AdipoR1 and AdipoR2 receptors have been identified in
and endothelial cells [58]. This GPI-anchored receptor different cell types of the human follicle (oocyte cumulus
binds hexameric and HMW forms of adiponectin and has oophorus, granulosa cells and theca). In the human gran-
been implicated in liver fibrosis, cardiovascular protection ulosa cells, adiponectin is implicated in the regulation of
and anti-diabetic effects in rodents [5861]. Various data steroidogenesis by modulating the action of insulin and
have indicated that adiponectin is influential in male and insulin like growth factor 1 (IGF-1) ([69, 71], Figure1). Similar
female fertility and that adiponectin produced locally in results have been observed in rats where this adiponectin
reproductive organs or by endocrine pathways could be effect is explained by an increase in the signaling of the
an energy sensor capable of regulating female and male IGF-1 receptor [72]. In human granulosa, this improvement
gonadal function. of steroidogenesis by adiponectin is partially due to an
increase in the expression of the aromatase enzyme respon-
sible for the biosynthesis of estrogen. In human granulosa
Pituitary-Hypothalamus KGN cells, specific inactivation of AdipoR1 and AdipoR2
shows that AdipoR1 is involved in cell survival, whereas
In mice, adiponectin inhibits GnRH secretion in an AdipoR2 is mainly involved in steroidogenesis [71]. In 2009,
immortalized hypothalamic neurons cell-line through Gutman et al. [73] showed an increase in the concentra-
activation of AMPK [62], most likely by inhibiting Kiss-1 tion of adiponectin in follicular fluid in ovarian response
gene transcription [63]. AdipoR1 and R2 are also expressed to gonadotropins after administration of recombinant LH.
in a gonadotrope pituitary cell-line. In this cell line, adi- Adiponectin enhances oocyte maturation and early embryo
ponectin down-regulates basal and GnRH-stimulated LH development in mouse, pig and human IVF procedures
secretion through phosphorylation of AMPK [64]. More- ([70, 74], Figure 1). Several polymorphisms of the genes
over, in rat pituitary cell cultures, adiponectin inhibits encoding adiponectin and its two receptors have been iden-
GH and LH release [65]. Thus, in rodents, adiponectin, tified. Some variants and haplotypes identified are associ-
AdipoR1 and AdipoR2 are expressed in the pituitary and ated with sizes larger litters, a smaller number of stillborn
may be involved in gonadotrope regulation in a paracrine and mummified piglets and shorter weaning-estrus inter-
way [65]. In swine, AdipoR1 and R2 are expressed in the val [75]. Thus, in human ovary, adiponectin could modulate
pituitary and their expression patterns depend upon the not only follicle growth but also embryo development.
estrous cycle [66]. Adiponectin expression in the porcine In women with PCOS, the concentration of adiponec-
pituitary is also dependent of the estrous cycle phase and tin is impaired, after controlling for BMI-related effects, but
affects gonadotropin secretion in a manner dependent is associated with abdominal obesity and hyperandrogen-
on the phase of this cycle [67]. During in vitro studies, ism [76]. Plasma adiponectin is reduced in a mouse model
adiponectin increases FSH release by porcine anterior of PCOS [77]. Moreover, in this syndrome, metformin, an
pituitary cells [67]. The expression of adiponectin and anti-diabetic agent which improves insulin sensitivity,
adiponectin receptors has been studied in the human increases the ovulation rate in some studies [78], and
brain [68]. Adiponectin and adiponectin receptors are plasma adiponectin levels [79]. Sarray etal. observed some
expressed in gonadotrope, thyreotrope and somatotrope differences in adiponectin but not leptin or resistin serum
cells in the adenohypohyse, suggesting local regulation levels between women with PCOS and control women sug-
of these endocrine axes [39, 68]. Overall, these findings gesting that ratios of adiponectin/leptin and adiponectin/
suggest that adiponectin may act as a negative regulator resistin could constitute novel predictor factors to explain
of the gonadotrophin pathways at a central level through PCOS and its associated features [80]. Thus, adiponectin
a decrease in GnRH and LH secretion. However, in swine, could be a pathophysiological link between metabolic
adiponectin effects on gonadotropin secretion may disturbances (insulin resistance and obesity) and fertility
depends on the estrous cycle phase [67]. disorders found in patients suffering from PCOS.

Ovary Testis

In the human ovary, adiponectin has been identified in In chickens, an increase in adiponectin receptors in
the ovarian follicle, particularly in follicular fluid [69]. sexual maturation has been reported, and suggests a role

Brought to you by | Universidade Federal do Esprito Santo UFES


Authenticated
Download Date | 7/12/17 9:08 PM
16Dupont etal.: Adipokines and human reproductive organs

of adiponectin in steroidogenesis and spermatogenesis, adiponectin negatively regulating testosterone secretion


and in the function of Sertoli cells and sperm motility [81]. by Leydig cells. Furthermore, in murine Leydig cells, adi-
In rat testis, adiponectin is mainly present in the intersti- ponectin may also have a protective effect against pro-
tial Leydig cells, while its receptors (mainly AdipoR1) are inflammatory cytokines by suppressing NF-B activation
present in the seminiferous tubules, although AdipoR2 is through promotion of AMP-activated protein kinase phos-
also expressed in Leydig cells [82]. Adiponectin decreases phorylation [85]. There are currently no available data on
in vitro testosterone production in the presence or the role of adiponectin in the human testis (Figure 2), nor
absence of hCG (human chorionic gonadotropin) through on an eventual function of CDH13 in the male gonad.
down-regulation of the Steroidogenic Acute Regulatory
protein (StAR) expression, whereas it has no effect on the
expression of genes coding for anti-Mullerian hormone Semen
(AMH) and stem cell factor (SCF), specifically in Sertoli
cells [82, 83]. In mice, a deficiency in the AdipoR2 recep- Adiponectin and adiponectin receptors have been
tor leads to atrophy of the seminiferous tubules and described in bull sperm and they are immunolocalized
aspermia without modifying the testosterone concentra- in acrosomal, post-acrosomal, equatorial and tail regions
tion [84]. Adiponectin gene expression, like resistin, is of bull spermatozoa [55]. Adiponectin and its receptors
significantly up-regulated in Leydig cells obtained from may be implicated in capacitation and their presence on
Rhox5 null mice compared to wild-type animals, linking spermatozoa after the acrosomal reaction has led to the
adiponectin expression, insulin and androgen signaling hypothesis that they could participate in sperm-egg inter-
in mice testis [44]. actions [55]. In human, Thomas and colleagues showed
Altogether, data in rodents suggest that adiponec- that concentration of adiponectin is higher in seminal
tin and classic adiponectin receptors are involved in the plasma than in serum [47]. Accordingly, seminal plasma
regulation of spermatogenesis and steroidogenesis, with adiponectin levels are significantly higher in normal

Testis
Spermatozoa

Sertoli cells:
Adiponectin = no effect [82, rat]
Resistin, omentin, visfatin, chemerin = nd

Germ cells

Blood vessel

Leydig cells:
-In vitro testosterone production:

Resistin [43, rat], visfatin [137, rat]

Chemerin [102, 103, rat], adiponectin [82, 83, rat]


Omentin: nd

Figure 2:In vitro effect of chemerin, adiponectin, resistin, visfatin and omentin on testicular cells.
nd, Undetermined; , increases; , decreases.

Brought to you by | Universidade Federal do Esprito Santo UFES


Authenticated
Download Date | 7/12/17 9:08 PM
Dupont etal.: Adipokines and human reproductive organs17

weight men than in overweight or obese men [47] and several tissues, mainly the liver, kidney, and pancreas but
plasma adiponectin levels positively correlate with sperm also in brown and white adipose tissue. It is also present
count and normal morphology, suggesting a role in sper- in the pituitary, placenta, ovary and testis. These data
matogenesis. Like resistin, adiponectin is found in seminal suggest a potential role of chemerin in reproduction. To
plasma in vasectomized men, suggesting that adiponectin exert its cellular effects, chemerin can bind to three seven
may be produced downstream of the vas deferens. transmembrane G protein-coupled receptors: CMKLR1
(like chemokine receptor 1 also known as ChemR23 and
DEZ) GPR1 (G protein-coupled receptor 1) and CCRL2 (CC
Endometrium chemokine receptor-like 2, for review [92, 93]. In humans,
serum chemerin concentration is positively correlated
There is in vitro evidence that AdipoR1 and R2 are expressed with BMI and waist circumference and significantly
in the human myometrium and that their expression level higher in overweight or obese patients than in those with
changes throughout gestation [86]. The expression of both a normal weight [47]. There is also a statistically signifi-
genes is increased in the midluteal phase, the period of cant negative correlation between serum chemerin and
embryo implantation [86]. Adiponectin inhibits human serum testosterone[47].
endometrial stromal cell proliferation in a dose and time There is currently no available data on the role (if any)
dependant manner, and causes cell death. Therefore, it of chemerin in the reproduction functions at the brain
has suggested as an anti-endometriosis agent [87]. Some level in the pituitary and hypothalamus.
studies showed that changes of AdipoR expression pro-
files could be implicated in the development of uterine
receptivity, and might therefore be new potential targets Ovary
for prediction of implantation failure [88]. Thus, the
effects of adiponectin might be relevant to pathological Chemerin and three receptors are present in the ovarian
and physiological endometrium-related events such as follicle in women [94], cows [95] and rats [96, 97]. Spe-
implantation and endometriosis. cifically in the latter species, they are expressed in differ-
ent cells of the ovarian follicle (granulosa cells and theca,
cumulus cells and oocytes) [95]. Several in vitro studies in
Embryo rodents, humans and bovines show that chemerin plays
a role in the functions of the ovary including steroido-
In mice, adiponectin receptors are expressed in mature genesis, apoptosis, cell proliferation and oocyte matura-
oocytes and preimplantation embryos. Full-length adi- tion (Figure1). In rats, chemerin removes the stimulatory
ponectin may act as an embryotrophic factor increasing effect of FSH on the secretion of progesterone and estra-
blastocyst cell number, whereas other adiponectin iso- diol in cultured pre-antral follicles and granulosa cells
forms, such as mutated trimeric adiponectin, exert the [96]. It also inhibits the expression of the nuclear recep-
opposite effect [89]. However, after implantation, full- tor NR5A1 and NR5A2 in response to FSH. These receptors
length adiponectin seems to decrease fetal-growth by lim- bind to the promoter regions of the genes of steroidogenesis
iting fetal nutrient availability through down-regulation enzymes, P450scc and P450arom. These inhibitory effects
of placental amino-acid transporter activity [90]. of chemerin on the in vitro production of steroids by granu-
losa cells are found in other species such as cattle [95] and
humans ([94], Figure 1). In cow granulosa cells, chemerin
also inhibits the expression of the cholesterol transporter,
Chemerin StAR, P450 aromatase and cholesterol de novo synthe-
sis; these effects involve the CMKLR1 receptor [95]. In rats
Chemerin is a recently identified adipose hormone. It is treated with 5alpha-dihydrotestosterone (used to mimic
initially involved in regulation of the immune system, adi- polycystic ovarian syndrome), chemerin not only reduced
pogenesis and energy metabolism. Chemerin is a chem- the secretion of estradiol by the granulosa cells but also
oattractant composed of 163 amino acids, synthesized as induced apoptosis of these cells [97]. Chemerin blocks the
a pre-prochemerin then secreted as an inactive precursor expression of oocyte factor, Growth/differentiation factor
called prochemerin. Prochemerin is activated by cleav- 9 (GFD9), which promotes cell proliferation and differen-
age of its C-terminal leading to different isoforms lacking tiation of pre-antral follicles in antral follicles [97]. In addi-
more terminal amino acids [91]. Chemerin is expressed in tion to negative effects on steroidogenesis and the survival

Brought to you by | Universidade Federal do Esprito Santo UFES


Authenticated
Download Date | 7/12/17 9:08 PM
18Dupont etal.: Adipokines and human reproductive organs

and proliferation of granulosa cells, chemerin blocks Placenta


the in vitro maturation of bovine oocytes in the germinal
vesicle stage ([95], Figure 1). Chemerin thus appears to be In humans and rats, chemerin is localized to the tropho-
an important intra-ovarian regulator and could contribute blast cells of the placenta. At the last stage of pregnancy
to deregulation of follicular development. A significant in women, serum levels of several adipocytokines such
increase in the amount of chemerin in plasma and adipose as leptin [104, 105], visfatin [104], resistin [104, 105] and
tissue was observed in patients with PCOS [98100]. This TNF- [105] are higher, while circulating levels of adi-
increase in plasma chemerin was abolished after treat- ponectin [104, 105] are reduced. Similarly, contrary to the
ment with metformin, an anti-diabetic agent [99]. Finally, rat, the concentration of chemerin increases during preg-
expression (mRNA and protein) of chemerin and its recep- nancy in women [106, 107]. The high plasma concentration
tor (CMKLR1) is higher in the rat ovary stimulated with of chemerin in late pregnancy suggests that it plays a role
5-dehydrotestosterone used to induce PCOS [97]. It has in embryonic growth [106, 108]. Finally, several studies
been proposed that chemerin plays a role in the pathologi- have shown an increase in plasma chemerin in the case
cal process of PCOS in negatively controlling the synthesis of pre-eclampsia, which is a complication of pregnancy
of follicular steroids that are stimulated by FSH [101]. characterized by development of hypertension associ-
ated with proteinuria [109]. The more elevated the plasma
chemerin is, the more severe the preeclampsia is [109].
Testis This disease affects 2%5% of pregnancies in women and
is a major factor in fetal mortality.
In humans and rodent, chemerin and its receptors
(CMKLR1, GRP1 and CCRL2) are present in the testes [102,
103]. In rats, the expression of chemerin decreases during
postnatal development (590 days), whereas expression Omentin
of its three receptors increases [102]. In humans and rats,
chemerin and two of these receptors, CMKLR1 and GPR1, Omentin-1 (also named omentin, intelectin-1, endothelial
are specifically localized to Leydig cells ith low levels in the lectin HL-1 and intestinal lactoferrin receptor) is another
germ cells [102]. Moreover, in primary cultures of Leydig recently discovered adipokine predominantly produced
cells, chemerin inhibits in vitro testosterone secretion in by visceral adipose tissue in humans and rhesus monkeys
response to hCG ([102, 103], Figure 2). These results could [110112]. The mature omentin is a 120-kDa homotrimer in
be explained by a reduction in the expression of the dehy- which 40-kDa polypeptides are bridged by disulfide bonds.
drogenase 3-b-hydroxysteroid (3bHSD) enzyme and phos- A homolog of omentin-1, referred to as omentin-2, shares
phorylation of the MAPK signaling pathway ERK1/2. Thus, 83% amino acid identity with omentin-1 [110]. Omentin-1
in males, as in females, chemerin exerts an inhibitory is the major circulating form in human plasma [113]. Some
effect on steroidogenesis in vitro (Figures 1 and 2). evidence shows that omentin-1 expression is altered by
inflammatory states and obesity [113115]. Omentin-1
enhances insulin action [116]. It is inversely related to
Semen obesity [113] and is increased after weight loss [117], and
down-regulated by insulin and glucose [118]. Omentin-1
In human seminal plasma, chemerin is negatively corre- has also been linked to metabolic syndrome and PCOS
lated with sperm motility, suggesting an effect on spermat- [118]. Plasma omentin-1 levels are higher in women com-
ogenesis or sperm maturation in the epididymides, where pared with men [113] and are decreased in women with
spermatozoa acquire their progressive motility during PCOS compared with those without PCOS in BMI-matched
epididymal transport. This hypothesis is reinforced by the control subjects [118, 119]. Treatment with metformin for
fact that chemerin levels in vasectomized patients are sig- 3 or 6 months significantly increased serum omentin-1
nificantly reduced [47]. Thus, it has been suggested that levels, as well as the ratio of omentin-1 to insulin in PCOS
chemerin secretion from epididymal adipose tissue could patients [118, 120]. There is also a significant difference in
increase with obesity and reduce sperm motility through omentin-1 plasma levels between regular menstrual cycles
direct effects on spermatozoa or indirect effects during and irregular menstrual cycles in PCOS patients suggest-
epidydimal maturation and transport. Seminal plasma ing that omentin-1 could affect the secretion of hormones
chemerin also positively correlates with sperm concentra- that regulate ovarian and menstrual function or affect
tion, but not with sperm count [47]. receptors for these hormones [121]. Omentin-1 is expressed

Brought to you by | Universidade Federal do Esprito Santo UFES


Authenticated
Download Date | 7/12/17 9:08 PM
Dupont etal.: Adipokines and human reproductive organs19

in adipose tissue but also in reproductive tissues includ- showed that higher plasma visfatin/NAMPT may prevent
ing the placenta and ovary in humans, monkeys and a labor-associated decrease in SIRT1 leading to post-term
sheep [116, 122, 123]. In humans, lower omentin-1 expres- delivery in obesity [133].
sion in the placenta and the maternal plasma is associ- In mammals, there is some evidence to suggest that
ated with pre-existing maternal obesity. Thus, alteration NAMPT is directly involved in regulating human repro-
in omentin-1 in pregnancy may influence the develop- ductive functions. NAMPT is present in human ovarian
ment of metabolic disorders in offspring later in life [124]. follicles and increases insulin-like growth factor 1 (IGF1)-
Furthermore, in our lab, we have showed that omentin-1 induced steroidogenesis in primary human granulosa
is expressed in the human ovarian follicle [125]. In con- cells ([128], Figure 1). Furthermore, in women who are
trast to control patients, omentin-1 levels are higher in undergoing controlled ovarian stimulation, there is a cor-
follicular fluid than in plasma in PCOS patients [125]. In relation between the concentration of NAMPT in the fol-
human luteinized granulosa cells, omentin-1 expression is licular fluids and the number of oocytes retrieved [134]. In
increased at short term (12 or 24 h) by various hormones rats, the administration of NAMPT during superovulation
involved in follicle development, including FSH, insulin, has been shown to play an important role in the regula-
and IGF-1), and the insulin sensitizer, metformin. Further- tion of oocyte quality, and it can improve oocyte quality
more, omentin-1, through induction of visfatin expression, and fertility in aged female mice ([135], Figure 1). NAMPT
ameliorates IGF-1-induced steroidogenesis and IGF-1R is expressed in the testes of male rats, more specifically in
signaling (Figure 1). Thus, omentin-1 could be involved in Leydig cells, spermatocytes and sperm [136]. In cultured
folliculogenesis and may contribute to the pathophysiol- Leydig cells, NAMPT has been shown to increase testoster-
ogy of PCOS. one production ([137], Figure 2). In humans, NAMPT levels
There is currently no available data on the role (if any) are significantly higher in seminal plasma than in serum,
of omentin-1 in the testis and on higher levels of the repro- which suggests that testicular cells produce NAMPT [47].
ductive axis.

Conclusions
Visfatin
It is now clear that adipose tissue may communicate with
Visfatin is a novel adipokine mainly produced by perivas- the brain, gonads, and uterus to regulate reproductive
cular adipose tissue [126]. It is also known as a Nicoti- functions via these new adipokines. However, since these
namide phosphorybosyltransferase (NAMPT), or a pre-B molecules are also produced by the gonadotrope axis
cell colony-enhancing factor (PBEF), and is a 52kDa and genital tractus, we cannot exclude local autocrine
mammalian protein that is constitutively synthesized by or paracrine effects. Do these adipokines have beneficial
adipose tissue and many other tissues [127], including or negative effects on human fertility? Most studies have
reproductive tissue [128, 129]. NAMPT has been shown been performed in in vitro ovarian cells, so it is difficult
to have several intra- and extra-cellular functions, and to give an answer about the whole organism. However,
two isoforms, intracellular and extracellular, have been in the human ovary, and more precisely in granulosa
reported. Intracellular NAMPT plays a critical role in cells, adiponectin, visfatin and omentin increase in vitro
maintaining the activity of nicotinamide adenine dinu- steroid production induced by IGF-1, whereas resistin and
cleotide (NAD)-dependent enzymes [127]. It contributes chemerin have the opposite effect. In these ovarian cells,
to the biosynthesis of NAD by acting to convert nicoti- the effects of these adipokines appear to be mainly IGF-1-
namide into nicotinamide mononucleotide (NMN), and or insulin-dependent. Indeed, in human granulosa cells,
it represents the limiting factor for this enzymatic reac- omentin, visfatin and adiponectin increase phosphoryla-
tion [130]. The pattern of changes in circulating visfatin tion of the beta subunit of IGF-1R, whereas chemerin and
concentrations differs between normal and obese preg- resistin inhibit it (Figure 3). Abnormal levels of adipokines
nant women [131]. Its plasma level is increased during have been shown to be strongly associated with insulin
the development of obesity [132] and in pregnant women resistant and type 2 diabetes. Thus, further work will be
at term [133]. Studies found that visfatin at relatively low necessary to better understand the role of adipokines in
doses (110 nM), decreases both spontaneous and oxy- reproductive functions that may act as a link between
tocin induced contractions of pregnant human and rat obesity and PCOS. Fertility is strongly dependent on the
myometrial tissue in vitro [133]. In humans, a recent study pituitary-hypothalamus axis. Few data are available on

Brought to you by | Universidade Federal do Esprito Santo UFES


Authenticated
Download Date | 7/12/17 9:08 PM
20Dupont etal.: Adipokines and human reproductive organs

IGF-1
Adiponectin Chemerin
C-Term IGF-1R CMKLR1
AdipoR1 or GPR1
or AdipoR2
+ - G
P P i
N-Term +

+ Subunit Subunit ? Visfatin
-

Omentin ?
? Resistin
MAPK ERK1/2
- Inhibits
Human
+ Stimulates granulosa cells

Steroids secretion (Pg, E2)

Figure 3:In vitro effect of chemerin, adiponectin, resistin, visfatin and omentin on IGF-1R signaling in human granulosa cells.

the effects of these new adipokines on this axis, except 6. Spritzer PM, Lecke SB, Satler F, Morsch DM. Adipose tissue
for adiponectin, which can be a negative regulator of in dysfunction, adipokines, and low-grade chronic inflammation in
polycystic ovary syndrome. Reproduction 2015;149:R21927.
vitro gonadotrophin secretion in rodent pituitary cells.
7. Rotterdam ESHRE/ASRM-Sponsored PCOS Consensus Workshop
Adipokines also have crucial modulatory effects during Group. Revised 2003 consensus on diagnostic criteria and long-
pregnancy and are reported to be involved in the patho- term health risks related to polycystic ovary syndrome. Fertil
physiology of pregnancy-related complications. Changes Steril 2004;81:1925.
in serum levels of several adipokines throughout human 8. Holcomb IN, Kabakoff RC, Chan B, Baker TW, Gurney A,
HenzelW, Nelson C, Lowman HB, Wright BD, Skelton NJ,
gestation have been well documented. Further inves-
Frantz GD, Tumas DB, Peale FV Jr, Shelton DL, Hbert CC.
tigation should focus on the effects of endogenous adi- FIZZ1, a novel cysteine-rich secreted protein associated with
pokines on blastocyst development, as well as in patients pulmonary inflammation, defines a new gene family. EMBO J
with metabolic dysregulations such as obesity and PCOS. 2000;19:404655.
Taken together, all these adipokines produced locally 9. Steppan CM, Bailey ST, Bhat S, Brown EJ, Banerjee RR,
or by the endocrine pathway could be energy sensors WrightCM, Patel HR, Ahima RS, Lazar MA. The hormone resistin
links obesity to diabetes. Nature 2001;409:30712.
capable of regulating human fertility at different levels
10. Kim KH, Lee K, Moon YS, Sul HS. A cysteine-rich adipose tissue-
of the reproductive axis. However, their detailed role in specific secretory factor inhibits adipocyte differentiation. J Biol
reproduction awaits further clarification through future Chem 2001;276:112526.
studies. 11. Pang SS, Le YY. Role of resistin in inflammation and inflamma-
tion-related diseases. Cell Mol Immunol 2006;3:2934.
12. Filkova M, Senolt L, Vencovsky J. The role of resistin in inflam-
matory myopathies. Curr Rheumatol Rep 2013;15:336.
References 13. Su CM, Hsu CJ, Tsai CH, Huang CY, Wang SW, Tang CH. Resistin
promotes angiogenesis in endothelial progenitor cells through
1. Kershaw EE, Flier JS. Adipose tissue as an endocrine organ. J Clin inhibition of MicroRNA206: potential implications for rheuma-
Endocrinol Metab 2004;89:254856. toid arthritis. Stem Cells 2015;33:224355.
2. Lehr S, Hartwig S, Sell H. Adipokines: a treasure trove for the 14. Patel L, Buckels AC, Kinghorn IJ, Murdock PR, Holbrook JD,
discovery of biomarkers for metabolic disorders. Proteomics Clin Plumpton C, Macphee CH, Smith SA. Resistin is expressed
Appl 2012;6:91101. in human macrophages and directly regulated by PPAR
3. Molica F, Morel S, Kwak BR, Rohner-Jeanrenaud F, Steffens S. gamma activators. Biochem Biophys Res Commun
Adipokines at the crossroad between obesity and cardiovascular 2003;300:4726.
disease. Thromb Haemost 2015;113:55366. 15. Comninos AN, Jayasena CN, Dhillo WS. The relationship
4. Rosenblatt A, Faintuch J, Cecconello I. Abnormalities of reproduc- between gut and adipose hormones, and reproduction. Human
tive function in male obesity before and after bariatric surgery-A Reprod Update 2014;20:15374.
comprehensive review. Obes Surg 2015;25:128192. 16. Munir I, Yen HW, Baruth T, Tarkowski R, Azziz R, Magoffin DA,
5. Jungheim ES, Travieso JL, Hopeman MM. Weighing the impact of Jakimiuk AJ. Resistin stimulation of 17alpha-hydroxylase activity
obesity on female reproductive function and fertility. Nutr Rev in ovarian theca cells in vitro: relevance to polycystic ovary
2013;71(Suppl 1):S38. syndrome. J Clin Endocrinol Metab 2005;90:48527.

Brought to you by | Universidade Federal do Esprito Santo UFES


Authenticated
Download Date | 7/12/17 9:08 PM
Dupont etal.: Adipokines and human reproductive organs21

17. Maillard V, Froment P, Rame C, Uzbekova S, Elis S, DupontJ. 35. Reverchon M, Cornuau M, Rame C, Guerif F, Royere D, Dupont J.
Expression and effect of resistin on bovine and rat granu- Resistin decreases insulin-like growth factor I-induced steroid
losa cell steroidogenesis and proliferation. Reproduction production and insulin-like growth factor I receptor signaling in
2011;141:46779. human granulosa cells. Fertil Steril 2013;100:24755. e13.
18. Way JM, Gorgun CZ, Tong Q, Uysal KT, Brown KK, Harrington WW, 36. Bromfield JJ, Sheldon IM. Lipopolysaccharide initiates inflam-
Oliver WR Jr, Willson TM, Kliewer SA, Hotamisligil GS. Adipose mation in bovine granulosa cells via the TLR4 pathway and
tissue resistin expression is severely suppressed in obesity perturbs oocyte meiotic progression in vitro. Endocrinology
and stimulated by peroxisome proliferator-activated receptor 2011;152:502940.
gamma agonists. J Biol Chem 2001;276:256513. 37. Adam M, Saller S, Strobl S, Hennebold JD, Dissen GA, Ojeda SR,
19. Panidis D, Koliakos G, Kourtis A, Farmakiotis D, Mouslech T, Stouffer RL, Berg D, Berg U, Mayerhofer A. Decorin is a part of
Rousso D. Serum resistin levels in women with polycystic ovary the ovarian extracellular matrix in primates and may act as a
syndrome. Fertil Steril 2004;81:3616. signaling molecule. Hum Reprod 2012;27:324958.
20. Yilmaz M, Bukan N, Demirci H, Ozturk C, Kan E, Ayvaz G, ArslanM. 38. Zhang H, Qiu J, Ye C, Yang D, Gao L, Su Y, Tang X, Xu N, Zhang D,
Serum resistin and adiponectin levels in women with polycystic Xiong L, Mao Y, Li F, Zhu J. ROR1 expression correlated with poor
ovary syndrome. Gynecol Endocrinol 2009;25:24652. clinical outcome in human ovarian cancer. Sci Rep 2014;4:5811.
21. Seow KM, Juan CC, Wu LY, Hsu YP, Yang WM, Tsai YL, Hwang JL, 39. Wilkinson M, Brown R, Imran SA, Ur E. Adipokine gene
Ho LT. Serum and adipocyte resistin in polycystic ovary syn- expression in brain and pituitary gland. Neuroendocrinology
drome with insulin resistance. Hum Reprod 2004;19:4853. 2007;86:191209.
22. Chen YC, Tsai EM, Chen HS, Liu YH, Lee CH, Chou FH, Chen SY, 40. Rodrguez-Pacheco F, Vzquez-Martnez R, Martnez-Fuentes AJ,
Jong SB, Chan TF. Serum resistin level is a predictor of ovar- Pulido MR, Gahete MD, Vaudry H, Gracia-Navarro F, Diguez C,
ian response in in vitro fertilisation cycle. Acta Obstet Gynecol Castao JP, Malagn MM. Resistin regulates pituitary somato-
Scand 2007;86:9637. trope cell function through the activation of multiple signaling
23. Seow KM, Juan CC, Hsu YP, Ho LT, Wang YY, Hwang JL. Serum pathways. Endocrinology 2009;150:464352.
and follicular resistin levels in women with polycystic ovarian 41. Benomar Y, Gertler A, De Lacy P, Crepin D, Ould Hamouda H,
syndrome during IVF-stimulated cycles. Hum Reprod 2005;20: Riffault L,Taouis M. Central resistin overexposure induces
11721. insulin resistance through Toll-like receptor 4. Diabetes
24. Lu XE, Huang HF, Li MG, Zhu YM, Qiang YL, Dong MY. Resistin 2013;62:10214.
levels of serum and follicular fluid in non-obese patients with 42. Horiguchi K, Syaidah R, Fujiwara K, Tsukada T, Ramadhani D,
polycystic ovary syndrome during IVF cycles. J Zhejiang Univ Sci Jindatip D, Kikuchi M, Yashiro T. Expression of small leucine-rich
B 2005;6:897902. proteoglycans in rat anterior pituitary gland. Cell Tissue Res
25. Varnagy A, Bodis J, Kovacs GL, Sulyok E, Rauh M, Rascher W. 2013;351:20712.
Metabolic hormones in follicular fluid in women undergoing in 43. Nogueiras R, Barreiro ML, Caminos JE, Gaytan F, SuominenJS,
vitro fertilization. J Reprod Med 2013;58:30511. Navarro VM, Casanueva FF, Aguilar E, Toppari J, Diguez C,
26. Tarkowski A, Bjersing J, Shestakov A, Bokarewa MI. Resistin Tena-Sempere M. Novel expression of resistin in rat testis:
competes with lipopolysaccharide for binding to toll-like functional role and regulation by nutritional status and hormo-
receptor 4. J Cell Mol Med 2010;14:141931. nal factors. JCell Sci 2004;117(Pt 15):324757.
27. Boutagy NE, McMillan RP, Frisard MI, Hulver MW. Metabolic 44. MacLean JA, Hu Z, Welborn JP, Song HW, Rao MK, Wayne CM,
endotoxemia with obesity: Is it real and is it relevant? Biochimie Wilkinson MF. The RHOX homeodomain proteins regulate the
2015. http://dx.doi.org/10.1016/j.biochi.2015.06.020. expression of insulin and other metabolic regulators in the
28. Al Hannan F, Culligan KG. Human resistin and the RELM of testis. J Biol Chem 2013;288:3480925.
inflammation in diabesity. Diabetol Metab Syndr 2015;7:54. 45. Mayerhofer A. Human testicular peritubular cells: more than
29. Lee S, Lee HC, Kwon YW, Lee SE, Cho Y, Kim J, Lee S, Kim JY, meets the eye. Reproduction 2013;145:R10716.
LeeJ, Yang HM, Mook-Jung I, Nam KY, Chung J, Lazar MA, 46. Kratzsch J, Paasch U, Grunewald S, Mueller MA, Thiery J,
KimHS. Adenylyl cyclase-associated protein 1 is a receptor for Glander HJ. Resistin correlates with elastase and interleukin-6
human resistin and mediates inflammatory actions of human in human seminal plasma. Reprod Biomed Online 2008;16:2838.
monocytes. Cell Metab 2014;19:48497. 47. Thomas S, Kratzsch D, Schaab M, Scholz M, Grunewald S,
30. Neill T, Schaefer L, Iozzo RV. Decoding the matrix: instructive ThieryJ, Paasch U, Kratzsch J. Seminal plasma adipokine levels
roles of proteoglycan receptors. Biochemistry 2015;54:458398. are correlated with functional characteristics of spermatozoa.
31. Daquinag AC, Zhang Y, Amaya-Manzanares F, Simmons PJ, Fertil Steril 2013;99:125663. e3.
Kolonin MG. An isoform of decorin is a resistin receptor on the 48. Moretti E, Collodel G, Mazzi L, Campagna M, Iacoponi F, F iguraN.
surface of adipose progenitor cells. Cell Stem Cell 2011;9:7486. Resistin, interleukin-6, tumor necrosis factor-alpha, and human
32. Sanchez-Solana B, Laborda J, Baladron V. Mouse resistin modu- semen parameters in the presence of leukocytospermia, smok-
lates adipogenesis and glucose uptake in 3T3-L1 preadipocytes ing habit, and varicocele. Fertil Steril 2014;102:35460.
through the ROR1 receptor. Mol Endocrinol 2012;26:11027. 49. Fujita Y, Mihara T, Okazaki T, Shitanaka M, Kushino R, Ikeda C,
33. Ho HY, Susman MW, Bikoff JB, Ryu YK, Jonas AM, Hu L, KuruvillaR, Negishi H, Liu Z, Richards JS, Shimada M. Toll-like receptors
Greenberg ME. Wnt5a-Ror-Dishevelled signaling constitutes a (TLR) 2 and 4 on human sperm recognize bacterial endotoxins
core developmental pathway that controls tissue morphogenesis. and mediate apoptosis. Hum Reprod 2011;26:2799806.
Proc Natl Acad Sci USA 2012;109:404451. 50. Yura S, Sagawa N, Itoh H, Kakui K, Nuamah MA, Korita D,
34. Niles LP, Lobb DK, Kang NH, Armstrong KJ. Resistin expression Takemura M, Fujii S. Resistin is expressed in the human
in human granulosa cells. Endocrine 2012;42:7425. placenta. J Clin Endocrinol Metab 2003;88:13947.

Brought to you by | Universidade Federal do Esprito Santo UFES


Authenticated
Download Date | 7/12/17 9:08 PM
22Dupont etal.: Adipokines and human reproductive organs

51. Di Simone N, Di Nicuolo F, Marzioni D, Castellucci M, 67. Kiezun M, Smolinska N, Maleszka A, Dobrzyn K, Szeszko K,
SanguinettiM, DLppolito S, Caruso A. Resistin modulates Kaminski T. Adiponectin expression in the porcine pituitary dur-
glucose uptake and glucose transporter-1 (GLUT-1) expression in ing the estrous cycle and its effect on LH and FSH secretion. Am
trophoblast cells. J Cell Mol Med 2009;13:38897. J Physiol Endocrinol Metab 2014;307:E103846.
52. Cha J, Bartos A, Park C, Sun X, Li Y, Cha SW, Ajima R, Ho HY, 68. Psilopanagioti A, Papadaki H, Kranioti EF, Alexandrides TK,
Yamaguchi TP, Dey SK. Appropriate crypt formation in the uterus Varakis JN. Expression of adiponectin and adiponectin recep-
for embryo homing and implantation requires Wnt5a-ROR sign- tors in human pituitary gland and brain. Neuroendocrinology
aling. Cell Rep 2014;8:38292. 2009;89:3847.
53. Lord E, Ledoux S, Murphy BD, Beaudry D, Palin MF. E xpression 69. Chabrolle C, Tosca L, Rame C, Lecomte P, Royere D, DupontJ.
of adiponectin and its receptors in swine. J Anim Sci Adiponectin increases insulin-like growth factor I-induced
2005;83:56578. progesterone and estradiol secretion in human granulosa cells.
54. Berner HS, Lyngstadaas SP, Spahr A, Monjo M, Thommesen L, Fertil Steril 2009;92:198896.
Drevon CA, Syversen U, Reseland JE. Adiponectin and its recep- 70. Richards JS, Liu Z, Kawai T, Tabata K, Watanabe H, Suresh D,
tors are expressed in bone-forming cells. Bone 2004;35:8429. Kuo FT, Pisarska MD, Shimada M. Adiponectin and its receptors
55. Kasimanickam VR, Kasimanickam RK, Kastelic JP, Stevenson JS. modulate granulosa cell and cumulus cell functions, fertility,
Associations of adiponectin and fertility estimates in Holstein and early embryo development in the mouse and human. Fertil
bulls. Theriogenology 2013;79:76677. e1-3. Steril 2012;98:4719. e1.
56. Yamauchi T, Kamon J, Ito Y, Tsuchida A, Yokomizo T, Kita S, 71. Pierre P, Froment P, Negre D, Rame C, Barateau V, Chabrolle C,
Sugiyama T, Miyagishi M, Hara K, Tsunoda M, Murakami K, OhtekiT, Lecomte P, Dupont J. Role of adiponectin receptors, AdipoR1 and
Uchida S, Takekawa S, Waki H, Tsuno NH, Shibata Y, Terauchi Y, AdipoR2, in the steroidogenesis of the human granulosa tumor
Froguel P, Tobe K, Koyasu S, Taira K, KitamuraT, Shimizu T, Nagai R, cell line, KGN. Hum Reprod 2009;24:2890901.
Kadowaki T. Cloning of adiponectin receptors that mediate antidia- 72. Chabrolle C, Tosca L, Dupont J. Regulation of adiponectin and its
betic metabolic effects. Nature 2003;423:7629. receptors in rat ovary by human chorionic gonadotrophin treat-
57. Tosca L, Chabrolle C, Dupont J. AMPK: a link between metabo- ment and potential involvement of adiponectin in granulosa cell
lism and reproduction? Med Sci 2008;24:297300. steroidogenesis. Reproduction 2007;133:71931.
58. Hug C, Wang J, Ahmad NS, Bogan JS, Tsao TS, Lodish HF. 73. Gutman G, Barak V, Maslovitz S, Amit A, Lessing JB, Geva E.
T-cadherin is a receptor for hexameric and high-molecular- Recombinant luteinizing hormone induces increased produc-
weight forms of Acrp30/adiponectin. Proc Natl Acad Sci USA tion of ovarian follicular adiponectin in vivo: implications for
2004;101:1030813. enhanced insulin sensitivity. Fertil Steril 2009;91:183741.
59. Asada K, Yoshiji H, Noguchi R, Ikenaka Y, Kitade M, Kaji K, 74. Chappaz E, Albornoz MS, Campos D, Che L, Palin MF, MurphyBD,
YoshiiJ, Yanase K, Namisaki T, Yamazaki M, Tsujimoto T, Bordignon V. Adiponectin enhances in vitro development of
Akahane T, Uemura M, Fukui H. Crosstalk between high- swine embryos. Domest Anim Endocrinol 2008;35:198207.
molecular-weight adiponectin and T-cadherin during liver 75. Houde AA, Murphy BD, Mathieu O, Bordignon V, Palin MF.
fibrosis development in rats. Int J Mol Med 2007;20:7259. Characterization of swine adiponectin and adiponectin receptor
60. Denzel MS, Scimia MC, Zumstein PM, Walsh K, Ruiz-LozanoP, polymorphisms and their association with reproductive traits.
Ranscht B. T-cadherin is critical for adiponectin-mediated Anim Genet 2008;39:24957.
cardioprotection in mice. J Clin Invest 2010;120:434252. 76. Escobar-Morreale HF, Villuendas G, Botella-Carretero JI, Alvarez-
61. Takeuchi T, Adachi Y, Ohtsuki Y, Furihata M. Adiponectin receptors, Blasco F, Sanchon R, Luque-Ramirez M, San Milln JL. Adiponec-
with special focus on the role of the third receptor, T-cadherin, in tin and resistin in PCOS: a clinical, biochemical and molecular
vascular disease. Med Mol Morphol 2007;40:11520. genetic study. Hum Reprod 2006;21:225765.
62. Cheng XB, Wen JP, Yang J, Yang Y, Ning G, Li XY. GnRH secretion 77. van Houten EL, Kramer P, McLuskey A, Karels B, Themmen AP,
is inhibited by adiponectin through activation of AMP-activated Visser JA. Reproductive and metabolic phenotype of a mouse
protein kinase and extracellular signal-regulated kinase. model of PCOS. Endocrinology 2012;153:28619.
Endocrine 2011;39:612. 78. Tang T, Lord JM, Norman RJ, Yasmin E, Balen AH. Insulin-
63. Wen JP, Liu C, Bi WK, Hu YT, Chen Q, Huang H, Liang JX, Li LT, sensitising drugs (metformin, rosiglitazone, pioglitazone,
Lin LX, Chen G. Adiponectin inhibits KISS1 gene transcription D-chiro-inositol) for women with polycystic ovary syndrome,
through AMPK and specificity protein-1 in the hypothalamic oligo amenorrhoea and subfertility. Cochrane Database Sys Rev
GT1-7 neurons. J Endocrinol 2012;214:17789. 2012;5:CD003053.
64. Lu M, Tang Q, Olefsky JM, Mellon PL, Webster NJ. Adiponectin 79. Jakubowska J, Bohdanowicz-Pawlak A, Milewicz A, Szymczak J,
activates adenosine monophosphate-activated protein kinase Bednarek-Tupikowska G, Demissie M. Plasma cytokines
and decreases luteinizing hormone secretion in LbetaT2 gon- in obese women with polycystic ovary syndrome, before
adotropes. Mol Endocrinol 2008;22:76071. and after metformin treatment. Gynecol Endocrinol
65. Rodriguez-Pacheco F, Martinez-Fuentes AJ, Tovar S, Pinilla L, 2008;24:37884.
Tena-Sempere M, Dieguez C, Castao JP, Malagon MM. Regula- 80. Sarray S, Madan S, Saleh LR, Mahmoud N, Almawi WY. Valid-
tion of pituitary cell function by adiponectin. Endocrinology ity of adiponectin-to-leptin and adiponectin-to-resistin ratios
2007;148:40110. as predictors of polycystic ovary syndrome. Fertil Steril
66. Kiezun M, Maleszka A, Smolinska N, Nitkiewicz A, Kaminski T. 2015;104:4606.
Expression of adiponectin receptors 1 (AdipoR1) and 2 (AdipoR2) 81. Ocon-Grove OM, Krzysik-Walker SM, Maddineni SR, H endricksGL,
in the porcine pituitary during the oestrous cycle. Reprod Biol Ramachandran R. Adiponectin and its receptors are expressed in
Endocrinol 2013;11:18. the chicken testis: influence of sexual maturation on t esticular

Brought to you by | Universidade Federal do Esprito Santo UFES


Authenticated
Download Date | 7/12/17 9:08 PM
Dupont etal.: Adipokines and human reproductive organs23

ADIPOR1 and ADIPOR2 mRNA abundance. Reproduction 96. Wang Q, Kim JY, Xue K, Liu JY, Leader A, Tsang BK. Chemerin, a
2008;136:62738. novel regulator of follicular steroidogenesis and its potential
82. Caminos JE, Nogueiras R, Gaytan F, Pineda R, GonzalezCR, involvement in polycystic ovarian syndrome. Endocrinology
Barreiro ML, Castao JP, Malagn MM, Pinilla L, Toppari J, 2012;153:560011.
Diguez C, Tena-Sempere M. Novel expression and direct 97. Kim JY, Xue K, Cao M, Wang Q, Liu JY, Leader A, Han JY,
effects of adiponectin in the rat testis. Endocrinology TsangBK. Chemerin suppresses ovarian follicular develop-
2008;149:3390402. ment and its potential involvement in follicular arrest in rats
83. Pfaehler A, Nanjappa MK, Coleman ES, Mansour M, Wanders D, treated chronically with dihydrotestosterone. Endocrinology
Plaisance EP, Judd RL, Akingbemi BT. Regulation of adiponectin 2013;154:291223.
secretion by soy isoflavones has implication for endocrine func- 98. March WA, Moore VM, Willson KJ, Phillips DI, Norman RJ,
tion of the testis. Toxicol Lett 2012;209:7885. Davies MJ. The prevalence of polycystic ovary syndrome in a
84. Bjursell M, Ahnmark A, Bohlooly YM, William-Olsson L, RhedinM, community sample assessed under contrasting diagnostic
Peng XR, Ploj K, Gerdin AK, Arnerup G, Elmgren A, Berg AL, criteria. Hum Reprod 2010;25:54451.
Oscarsson J, Lindn D. Opposing effects of adiponectin receptors 99. Tan BK, Chen J, Farhatullah S, Adya R, Kaur J, Heutling D,
1 and 2 on energy metabolism. Diabetes 2007;56:58393. Lewandowski KC, OHare JP, Lehnert H, Randeva HS. Insu-
85. Wu L, Xu B, Fan W, Zhu X, Wang G, Zhang A. Adiponectin lin and metformin regulate circulating and adipose tissue
protects Leydig cells against proinflammatory cytokines by chemerin. Diabetes 2009;58:19717.
suppressing the nuclear factor-kappaB signaling pathway. 100. Guzel EC, Celik C, Abali R, Kucukyalcin V, Celik E, Guzel M,
FEBS J 2013;280:39207. Yilmaz M. Omentin and chemerin and their association with
86. Takemura Y, Osuga Y, Yamauchi T, Kobayashi M, Harada M, obesity in women with polycystic ovary syndrome. Gynecol
Hirata T, Morimoto C, Hirota Y, Yoshino O, Koga K, Yano T, Endocrinol 2014;30:41922.
Kadowaki T, Taketani Y. Expression of adiponectin receptors and 101. Wang Q, Leader A, Tsang BK. Inhibitory roles of prohibitin and
its possible implication in the human endometrium. Endocrinol- chemerin in FSH-induced rat granulosa cell steroidogenesis.
ogy 2006;147:320310. Endocrinology 2013;154:95667.
87. Bohlouli S, Khazaei M, Teshfam M, Hassanpour H. Adiponectin 102. Li L, Ma P, Huang C, Liu Y, Zhang Y, Gao C, Xiao T, Ren PG,
effect on the viability of human endometrial stromal cells and ZabelBA, Zhang JV. Expression of chemerin and its receptors in
mRNA expression of adiponectin receptors. Int J Fertil Steril rat testes and its action on testosterone secretion. J Endocrinol
2013;7:438. 2014;220:15563.
88. Dos Santos E, Serazin V, Morvan C, Torre A, Wainer R, de 103. Li L, Huang C, Zhang X, Wang J, Ma P, Liu Y, Xiao T, Zabel BA,
Mazancourt P, Dieudonn MN. Adiponectin and leptin systems Zhang JV. Chemerin-derived peptide C-20 suppressed gonadal
in human endometrium during window of implantation. Fertility steroidogenesis. Am J Reprod Immunol 2014;71:26577.
and sterility. 2012;97(3):771-8. e1. 104. DIppolito S, Tersigni C, Scambia G, Di Simone N. Adipokines,
89. Cikos S, Burkus J, Bukovska A, Fabian D, Rehak P, Koppel J. an adipose tissue and placental product with biological func-
Expression of adiponectin receptors and effects of adiponec- tions during pregnancy. Biofactors 2012;38:1423.
tin isoforms in mouse preimplantation embryos. Hum Reprod 105. Noureldeen AF, Qusti SY, Al-Seeni MN, Bagais MH. Maternal
2010;25:224755. leptin, adiponectin, resistin, visfatin and tumor necrosis
90. Rosario FJ, Schumacher MA, Jiang J, Kanai Y, Powell TL, factor-alpha in normal and gestational diabetes. Indian J Clin
JanssonT. Chronic maternal infusion of full-length adiponectin Biochem 2014;29:46270.
in pregnant mice down-regulates placental amino acid trans- 106. Kasher-Meron M, Mazaki-Tovi S, Barhod E, Hemi R, Haas J,
porter activity and expression and decreases fetal growth. GatI, Zilberberg E, Yinon Y, Karasik A, Kanety H. Chemerin
J Physiol 2012;590(Pt 6):1495509. concentrations in maternal and fetal compartments: implica-
91. Wittamer V, Franssen JD, Vulcano M, Mirjolet JF, Le Poul E, tions for metabolic adaptations to normal human pregnancy.
Migeotte I, Brzillon S, Tyldesley R, Blanpain C, Detheux M, JPerinat Med 2014;42:3718.
Mantovani A, Sozzani S, Vassart G, Parmentier M, Communi D. 107. Garces MF, Sanchez E, Acosta BJ, Angel E, Ruiz AI, Rubio-
Specific recruitment of antigen-presenting cells by chemerin, a Romero JA, Angel-Mller E, Suarez MA, Bohrquez LF, Bravo SB,
novel processed ligand from human inflammatory fluids. J Exp Nogueiras R, Diguez C, Caminos JE. Expression and regulation
Med 2003;198:97785. of chemerin during rat pregnancy. Placenta 2012;33:3738.
92. Mattern A, Zellmann T, Beck-Sickinger AG. Processing, signal- 108. Garces MF, Sanchez E, Ruz-Parra AI, Rubio-Romero JA,
ing, and physiological function of chemerin. IUBMB Life Angel-Mller E, Suarez MA, Bohrquez LF, Bravo SB,
2014;66:1926. NogueirasR, Diguez C, Caminos JE. Serum chemerin levels
93. Fatima SS, Rehman R, Baig M, Khan TA. New roles of the multidi- during normal human pregnancy. Peptides 2013;42:13843.
mensional adipokine: chemerin. Peptides 2014;62:1520. 109. Duan DM, Niu JM, Lei Q, Lin XH, Chen X. Serum levels of
94. Reverchon M, Cornuau M, Rame C, Guerif F, Royere D, the adipokine chemerin in preeclampsia. J Perinat Med
Dupont J. Chemerin inhibits IGF-1-induced progesterone and 2012;40:1217.
estradiol secretion in human granulosa cells. Hum Reprod 110. Lee JK, Schnee J, Pang M, Wolfert M, Baum LG, Moremen KW,
2012;27:1790800. Pierce M. Human homologs of the Xenopus oocyte cortical
95. Reverchon M, Bertoldo MJ, Rame C, Froment P, Dupont J. granule lectin XL35. Glycobiology 2001;11:6573.
CHEMERIN (RARRES2) decreases in vitro granulosa cell ster- 111. Suzuki YA, Shin K, Lonnerdal B. Molecular cloning and
oidogenesis and blocks oocyte meiotic progression in bovine functional expression of a human intestinal lactoferrin
species. Biol Reprod 2014;90:102. receptor. Biochemistry 2001;40:157719.

Brought to you by | Universidade Federal do Esprito Santo UFES


Authenticated
Download Date | 7/12/17 9:08 PM
24Dupont etal.: Adipokines and human reproductive organs

112. Tsuji S, Uehori J, Matsumoto M, Suzuki Y, Matsuhisa A, lutein cells: role in IGF-1-induced steroidogenesis through
Toyoshima K, Seya T. Human intelectin is a novel soluble lectin NAMPT. Biol Reprod 2014;91:50.
that recognizes galactofuranose in carbohydrate chains of 126. Fukuhara A, Matsuda M, Nishizawa M, Segawa K, Tanaka M,
bacterial cell wall. J Biol Chem 2001;276:2345663. Kishimoto K, Matsuki Y, Murakami M, Ichisaka T, MurakamiH,
113. de Souza Batista CM, Yang RZ, Lee MJ, Glynn NM, Yu DZ, Watanabe E, Takagi T, Akiyoshi M, Ohtsubo T, Kihara S,
PrayJ, Ndubuizu K, Patil S, Schwartz A, Kligman M, Fried SK, Yamashita S, Makishima M, Funahashi T, Yamanaka S,
GongDW, Shuldiner AR, Pollin TI, McLenithan JC. Omentin Hiramatsu R, Matsuzawa Y, Shimomura I. Visfatin: a protein
plasma levels and gene expression are decreased in obesity. secreted by visceral fat that mimics the effects of insulin.
Diabetes 2007;56:165561. Science 2005;307:42630.
114. Cai RC, Wei L, Di JZ, Yu HY, Bao YQ, Jia WP. Expression of 127. Revollo JR, Grimm AA, Imai S. The NAD biosynthesis path-
omentin in adipose tissues in obese and type 2 diabetic way mediated by nicotinamide phosphoribosyltransferase
patients. Zhonghua Yi Xue Za Zhi. 2009;89:3814. regulates Sir2 activity in mammalian cells. J Biol Chem
115. Auguet T, Quintero Y, Riesco D, Morancho B, Terra X, 2004;279:5075463.
CrescentiA, Broch M, Aguilar C, Olona M, Porras JA, 128. Reverchon M, Cornuau M, Cloix L, Rame C, Guerif F, RoyereD,
Hernandez M, Sabench F, del Castillo D, Richart C. New Dupont J. Visfatin is expressed in human granulosa cells: regu-
adipokines vaspin and omentin. Circulating levels and gene lation by metformin through AMPK/SIRT1 pathways and its role
expression in adipose tissue from morbidly obese women. in steroidogenesis. Mol Hum Reprod 2013;19:31326.
BMC Med Genet 2011;12:60. 129. Reverchon M, Rame C, Bertoldo M, Dupont J. Adipokines
116. Yang RZ, Lee MJ, Hu H, Pray J, Wu HB, Hansen BC, Shuldiner AR, and the female reproductive tract. Int J Endocrinol
Fried SK, McLenithan JC, Gong DW. Identification of omentin 2014;2014:232454.
as a novel depot-specific adipokine in human adipose tis- 130. Rongvaux A, Shea RJ, Mulks MH, Gigot D, Urbain J, Leo O,
sue: possible role in modulating insulin action. Am J Physiol Andris F. Pre-B-cell colony-enhancing factor, whose expression
Endocrinol Metab 2006;290:E125361. is up-regulated in activated lymphocytes, is a nicotinamide
117. Moreno-Navarrete JM, Catalan V, Ortega F, Gomez-Ambrosi J, phosphoribosyltransferase, a cytosolic enzyme involved in
Ricart W, Fruhbeck G, Fernandez-Real JM. Circulating omen- NAD biosynthesis. Eur J Immunol 2002;32:322534.
tin concentration increases after weight loss. Nutr Metab 131. Ozias MK, Li S, Hull HR, Brooks WM, Carlson SE. Relationship
2010;7:27. of circulating adipokines to body composition in pregnant
118. Tan BK, Adya R, Farhatullah S, Lewandowski KC, OHare P, women. Adipocyte 2015;4:449.
Lehnert H, Randeva HS. Omentin-1, a novel adipokine, is 132. Tsai PJ, Davis J, Thompson K, Bryant-Greenwood G. Visfatin/
decreased in overweight insulin-resistant women with polycys- nampt and SIRT1: roles in postterm delivery in pregnancies
tic ovary syndrome: ex vivo and in vivo regulation of omentin-1 associated with obesity. Reprod Sci 2015;22:102836.
by insulin and glucose. Diabetes 2008;57:8018. 133. Mumtaz S, AlSaif S, Wray S, Noble K. Inhibitory effect of
119. Choi JH, Rhee EJ, Kim KH, Woo HY, Lee WY, Sung KC. Plasma visfatin and leptin on human and rat myometrial contractility.
omentin-1 levels are reduced in non-obese women with Life Sci 2015;125:5762.
normal glucose tolerance and polycystic ovary syndrome. Eur J 134. Shen CJ, Tsai EM, Lee JN, Chen YL, Lee CH, Chan TF. The concen-
Endocrinol 2011;165:78996. trations of visfatin in the follicular fluids of women undergoing
120. Shaker M, Mashhadani ZI, Mehdi AA. Effect of treatment with controlled ovarian stimulation are correlated to the number of
metformin on omentin-1, ghrelin and other biochemical, clini- oocytes retrieved. Fertil Steril 2010;93:184450.
cal features in PCOS patients. Oman Med J 2010;25:28993. 135. Choi KH, Joo BS, Sun ST, Park MJ, Son JB, Joo JK, Lee KS.
121. Mahde A, Shaker M, Al-Mashhadani Z. Study of omentin1 and Administration of visfatin during superovulation improves
other adipokines and hormones in PCOS patients. Oman Med J developmental competency of oocytes and fertility potential in
2009;24:10818. aged female mice. Fertil Steril 2012;97:123441. e13.
122. Schaffler A, Neumeier M, Herfarth H, Furst A, ScholmerichJ, 136. Gurusubramanian G, Roy VK. Expression of visfatin in alloxan-
Buchler C. Genomic structure of human omentin, a new induced diabetic rat testis. Acta Histochem 2014;116:14628.
adipocytokine expressed in omental adipose tissue. Biochim 137. Hameed W, Yousaf I, Latif R, Aslam M. Effect of visfatin on
Biophys Acta 2005;1732:96102. testicular steroidogenesis in purified Leydig cells. J Ayub Med
123. French AT, Knight PA, Smith WD, Pate JA, Miller HR, Coll Abbottabad 2012;24:624.
PembertonAD. Expression of three intelectins in sheep and 138. Lagaly DV, Aad PY, Grado-Ahuir JA, Hulsey LB, Spicer LJ. Role
response to a Th2 environment. Vet Res 2009;40:53. of adiponectin in regulating ovarian theca and granulosa cell
124. Barker G, Lim R, Georgiou HM, Lappas M. Omentin-1 is function. Mol Cell Endocrinol 2008;284:3845.
decreased in maternal plasma, placenta and adipose tissue of 139. Maleszka A, Smolinska N, Nitkiewicz A, Kiezun M,
women with pre-existing obesity. PloS one 2012;7:e42943. Chojnowska K, Dobrzyn K, Szwaczek H, Kaminski T.
125. Cloix L, Reverchon M, Cornuau M, Froment P, Rame C, Costa C, Adiponectin expression in the porcine ovary during the
Froment G, Lecomte P, Chen W, Royre D, Guerif F, Dupont J. oestrous cycle and its effect on ovarian steroidogenesis.
Expression and regulation of INTELECTIN1 in human granulosa- Int J Endocrinol 2014;2014:957076.

Brought to you by | Universidade Federal do Esprito Santo UFES


Authenticated
Download Date | 7/12/17 9:08 PM

S-ar putea să vă placă și