You are on page 1of 31

SPECIAL CONTRIBUTIONS

Adipose tissue and reproduction in women


Henry Bohler, Jr., M.D.,
a
Sriprakash Mokshagundam, M.D.,
b
and Stephen J. Winters, M.D.
b
a
Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Gynecology, and Womens Health; and
b
Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, University of Louisville School of
Medicine, Louisville, Kentucky
Adipose tissue has been viewed as the primary source of stored energy, but with the discovery of novel adipose
tissue gene products, i.e., adipokines, another equally important role has emerged. Adipose tissue is a key endo-
crine organ involved in multiple processes, including glucose homeostasis, steroid production, immunoregulation,
hematopoesis, and reproduction. The distribution of adipose tissue may also have a signicant impact on reproduc-
tive function. (Fertil Steril

2010;94:795825. 2010 by American Society for Reproductive Medicine.)


Adipose tissue is the largest endocrine organ in the human
body, with effects on glucose homeostasis, steroid produc-
tion, the immune system, hematopoesis, and reproductive
function (16). The last several years have seen an expanded
interest in understanding the role of adipose tissue in repro-
duction, partly due to the discovery of novel adipose tissue
products, adipokines (7, 8). Adipokines may include any sub-
stance released by adipose tissue, whether from adipocytes
(fat cells), inltrating macrophages, stromovascular cells,
or other parts of the adipose tissue matrix (connective tissue
and blood vessels) (1, 9, 10). These substances include cyto-
kines, hormones, growth factors, chemokines, complement
factors, and proteins involved in vascular hemostasis, the reg-
ulation of blood pressure, lipid metabolism, glucose homeo-
stasis, and angiogenesis (11, 12).
Interest in the reproductive endocrine aspects of adipose tis-
sue has increased partly owing to the polycystic ovary syn-
drome (PCOS), one of the most investigated and prevalent
endocrinopathies in women of reproductive age (1316).
Adipose tissue excess (obesity) is present in at least 50% of
PCOS patients, and is responsible for unmasking the syndrome
in those who might otherwise go undetected (11, 17, 18). With
the present obesity epidemic, the number of patients diagnosed
with PCOS is likely to increase (19).
Obesity per se may not be as important as the distribution
of adipose tissue. Upper body fat has long been associated
with metabolic and endocrine-related ovulatory dysfunction,
but for the most part lower body fat has not (20, 21). Besides
an increase in abdominal subcutaneous adipose tissue (SAT),
upper body obesity (UBO) usually includes an accumulation
of intra-abdominal or visceral adipose tissue (VAT), which
may be the most signicant factor in disrupting normal me-
tabolism (22, 23). Both an excess and scarcity of body fat
have a signicant negative impact on several aspects of repro-
duction, including fertility and optimal pregnancy mainte-
nance (2426).
For the reproductive scientist and clinician, adipose tissue
represents an intriguing endocrine organ that is evidently
vital for reproductive function in women. The present review
focuses on adipose biology, its secretory products, and
their impact in physiologic and dysfunctional reproductive
conditions.
ADIPOSE TISSUE
Adipose tissue has several components, including adipo-
cytes, preadipocytes, endothelial cells, pericytes, monocytes,
macrophages, and broblastic connective tissue (27, 28). The
lipid droplet in adipocytes may increase in size (hypertrophy)
to reach a threshold that triggers the recruitment and differen-
tiation of preadipocytes (hyperplasia) (29). The potential for
adipocyte replication extends into adulthood, and once new
adipocytes form they remain permanently (27, 28).
Food consumption, physical activity, genetics, psycho-
logic factors, gender, and ethnicity are among the important
factors that contribute to the quantity and distribution of
adipose tissue (30). The quantity of adipocytes reects the
balance between adipose deposition (lipogenesis) and break-
down (lipolysis). Lipids account for 85% of the weight of ad-
ipose tissue, and 95%of the adipocyte content is triglycerides
(TG). The remaining 5% is water and proteins, including
adipokinesendocrine and paracrine factors, including
members of the cytokine family (27).
Received January 28, 2009; revised March 20, 2009; accepted March 24,
2009; published online July 8, 2009.
H.B. has nothing to disclose. S.M. has nothing to disclose. S.W. has noth-
ing to disclose.
Reprint requests: Henry Bohler, Jr., M.D., Division of Reproductive
Endocrinology and Infertility, Department of Obstetrics, Gynecology &
Womens Healthcare, University of Louisville, 401 E. Chestnut Street,
4th Floor, Suite 410, Louisville, KY 40202 (FAX: 502-271-5984; E-mail:
h.bohler@louisville.edu).
0015-0282/$36.00 Fertility and Sterility

Vol. 94, No. 3, August 2010


795
doi:10.1016/j.fertnstert.2009.03.079 Copyright 2010 American Society for Reproductive Medicine, Published by Elsevier Inc.
Histology and Differentiation
There are two types of mature adipocytes in mammals: brown
adipocytes and white adipocytes. Brown adipocytes are more
abundant in infancy but are scarcely represented in adults
(12). Besides their color and histologic differences (brown
adipocytes are multilocular), brown adipocytes are involved
in basal heat production, whereas white adipocytes serve as
a long-term fuel reservoir and are usually the type of fat dis-
cussed or implied in investigations involving obesity
(3133). Preadipocytes are also a component of adipose tis-
sue and respond to various stimuli, particularly insulin and
fatty acids, to become mature adipocytes (34). They initiate
differentiation through the peroxisome proliferator-activated
receptor g (PPAR-g) (12).
Innervation
Adipose tissue is regulated by the sympathetic nervous sys-
tem (SNS) (35, 36). b-Adrenergic receptors (adrenoceptors)
stimulate lipolysis to generate free fatty acids (FFAs) and
glycerol, whereas activation of a2A-adrenoceptors inhibits
lipolysis. Activation varies by fat depot and is also dependent
on age, gender, species, and amount of obesity (35, 36).
Activation of b-receptors through the G
s
protein increases
adenyl cyclase activity leading to an increase in cyclic aden-
osine monophosphate (cAMP) with phosphorylation of
hormone-sensitive lipase (HSL) and perilipin (35, 36).
HSL releases FFAs and glycerol from TGs. On the other
hand, activation of G
i
by a2-adrenoceptors decreases
cAMP to inhibit lipolysis (35).
Melanocortin receptors (MCR) have been colocalized with
SNS neurons and modulate SNS signaling to adipose cells
(35, 37). MCR activation reduces food intake and body fat
and increases energy expenditure and lipolysis (35, 38, 39).
In rodents, SNS stimulation also inhibits an increase in fat
cell number, and SNS denervation leads to obesity (4042).
Further evidence demonstrating an SNS inuence on adipose
tissue includes the presence of polymorphisms with b-recep-
tors in some morbidly obese humans and possibly the weight
gain that occurs with b-receptor antagonist therapy (4345).
Whether the parasympathetic nervous system is involved in
lipolysis is less clear (4648). There are comprehensive
reviews by Bartness and Song and others (35, 36, 44).
Metabolism
Adipose tissue metabolism results in either accumulation or
release of lipids from adipocytes as FFAs (29). Lipogenesis
and lipolysis are affected by the quantity of adipose tissue
(e.g., obese compared with nonobese), gender, neuronal in-
put, the fat depot examined (e.g., intra-abdominal compared
with extra-abdominal fat), hormones, including estrogens,
androgens, insulin, growth hormone, and cortisol, and phys-
ical activity (12, 34, 49, 50). Lipolysis is preferentially acti-
vated over lipogenesis when energy is needed, and this is
especially true during exercise and fasting (28, 51). Lipolytic
activity is reduced in subcutaneous fat compared with
visceral fat, which is also more resistant to the antilipolytic
effects of insulin (22, 52, 53).
Lipoprotein lipase (LPL) is synthesized in adipocytes as
well as muscle and many other tissues and is one of the key
enzymes involved in lipid storage (34, 36). After a meal, di-
etary TGs are transported in lymph to the circulation by chy-
lomicrons, where they acquire a series of proteins that allow
for interaction with LPL (54). LPL is stimulated by insulin
and is secreted into the circulation and resides on the capil-
lary endothelium, where it hydrolyzes TGs, forming FFAs
and glycerol (55, 56). FFAs are used for oxidative metabo-
lism or enter adipocytes by a protein-facilitated process to
be stored as TGs (34, 36, 55). During fasting, the net ow
of FFAs is outward from adipocytes into the circulation,
where they are bound by albumin to be oxidized as fuel by
liver, muscle, and other cells while preserving glucose for uti-
lization by CNS neurons and erythrocytes (12, 57). Insulin
promotes the uptake of FFAs by adipocytes by increasing
LPL synthesis and release but is less effective in visceral fat
compared with SAT (36, 52). The antilipolytic effect of insu-
lin is mediated by a reduction of cAMP through phosphoino-
sitide 3-kinase with suppression of protein kinase A (Fig. 1).
Hormone sensitive lipase, the key enzyme regulating lipol-
ysis, is found in many tissues, including adipocytes.(36, 51,
52). Phosphorylated HSL hydrolyzes TGs into FFAs and
glycerol, and insulin decreases this process (36, 55). Other li-
pases, such as adipose triglyceride lipase, may also regulate
adipocyte metabolism (56, 58).
Testosterone and dihydrotestosterone (DHT) promote cen-
tral obesity because they decrease HSL expression while up-
regulating LPL action (59). Conversely, estrogen decreases
central fat accumulation while increasing fat in the femo-
ral-gluteal region (60, 61). Estrogen suppresses adipose
growth in part by diminishing LPLgene transcription through
an estrogen responselike element in the promoter region of
the LPL gene or by modulating posttranscriptional activity
(60, 61). Cortisol (in the presence of insulin) and androgens
generally stimulate LPL activity and increase central fat
quantities (34, 52, 62). Catecholamines, growth hormone,
thyroid hormone, natriuretic peptide, and tumor necrosis fac-
tor alpha (TNF-a) are among other hormones affecting lipid
storage.
Tumor necrosis factor alpha promotes lipolysis by down-
regulating membrane proteins and perilipin, a protein that
forms the protective coat of lipid droplets, which are then
susceptible to degradation by HSL (36, 63). Owing to ele-
vated TNF-a in adipocytes, plasma FFAs are increased in
obesity. There are a number of excellent reviews of this eld
(12, 34, 36, 49, 52, 64) (Table 1).
Energy Homeostasis
Besides providing thermal insulation and protection from
mechanical trauma, adipose tissue is a key source of energy
in times of nutritional insufciency (65, 66). This is
796
Bohler et al. Adipose tissue and reproduction in women Vol. 94, No. 3, August 2010
important between meals and is critical during starvation.
The greater the quantity of adipose tissue, the greater the
chance of survival when food supply is inadequate (28, 65).
The probability of successful reproduction decreases when
food is scarce; therefore, adequate energy stores in times of
limited food intake appear to be teleologically linked to the
processes of reproduction, including the onset and mainte-
nance of ovulatory cycles, pregnancy, and lactation (67, 68).
Energy homeostasis is a complex process (65, 66, 6971).
A steady state of energy ux is reached when energy intake
(food consumption) equals energy expended and there is nei-
ther a gain nor loss of weight (70, 72). Energy expended is
accounted for by basal metabolism (the energy it takes to
conduct normal physiologic processes), adaptive thermogen-
esis (the energy dissipated as heat in response to environmen-
tal changes), and physical activity or voluntary movement
(72). A unilateral increase of energy intake or expenditure
will alter white adipose tissue mass quantity (11). Obesity
results when food intake exceeds energy expended. Mutated
genes may be responsible for an excess or deciency in
adipose tissue mass, but a genetic component is thought to
play a minor role in overall energy homeostasis (72).
Adipocytes, which account for approximately 50% of adi-
pose tissue, store energy as TGs, which are energy efcient
because their water content is small compared with other sour-
ces of energy, such as protein and carbohydrates (36, 65, 66).
During lipolysis, TGs release 9 kcal/g, whereas carbohydrates
and protein release 4 kcal/g (36, 51, 65).
The primary signal indicating the quantity of energy stores is
believed to be leptin, but other hormones are known to inu-
ence energy homeostasis by encouraging feeding or signaling
satiety (7, 70, 73). The gastrointestinal hormones ghrelin and
peptide YY
336
(PYY
336
) increase and decrease food intake,
respectively (71). Other hypothalamic orexigenic neurohor-
mones include neuropeptide Y (NPY), melanin-concentrating
FIGURE 1
Hormone regulation of TGmetabolism. Fat mass is regulated by the processes of lipogenesis and lipolysis which
are controlled primarily by lipoprotein lipase (LPL) and hormone sensitive lipase (HSL), respectively. LPL is
released to the capillary endothelium where it hydrolyzes TGs in very low density lipoproteins and chylomicrons.
FFA are re-esteried with glycerol-3-phosphate to form TG in the adipocyte. HSL, so named because of its
responsiveness to catecholamines and insulin, hydrolyzes adipose tissue TGs into FFA and glycerol. Androgens
increase LPL and decrease HSL activity. Estrogens decrease LPL and increase HSL activity. *Actions are fat
depot-specic. (See text) ANP atrial natriuretic peptide; TGtriglyceride; LPL lipoprotein lipase; FFA free
fatty acids; HSL hormone sensitive lipase. Modied and used with permission fromFrayn KN, Karpe F, Fielding
BA, Macdonald IA, Coppack SW. Integrative physiology of human adipose tissue. Int J Obes Relat Metab Disord
2003;27:87588.
Bohler. Adipose tissue and reproduction in women. Fertil Steril 2010.
Fertility and Sterility

797
hormone, and agouti-related peptide (AgRP), all of which are
down-regulated by leptin (11, 71, 72, 74). Leptin receptors
are colocalized with NPY-containing neurons in the arcuate
nucleus. A decrease in hypothalamic leptin is associated with
an increase in NPY/AgRP, which, along with other neurohor-
mones, initiates food intake (11, 71, 7477). Leptin also regu-
lates endocannabinoids, which activate receptors involved in
maintaining food intake (78, 79). The activities of pro-opiome-
lanocortin/melanocortin, cocaine- and amphetamine-related
transcript neurons, andcorticotropin-releasinghormone are an-
orexigenic and are stimulated by leptin (11, 79). In the brain,
insulin also suppresses NPY (71, 72).
Glucose Homeostasis
Skeletal muscle is the primary target for insulin-stimulated
glucose disposal, and signals from body fat are thought to
disrupt insulin signaling in skeletal muscle, predisposing to
insulin resistance (12, 80). If the Glut 4 adipose-specic
transporter is reduced in mice, insulin resistance develops
not only in adipocytes but also in muscle and liver, and in
obesity and type 2 diabetes Glut 4 expression is decreased
in adipose tissue but not in skeletal muscle (80, 81). Therapy
directed at increasing insulin sensitivity selectively in adi-
pose tissue signicantly improves glucose tolerance (12).
Clearly, obesity is linked to insulin resistance and diabetes,
again due to the importance of fat cells in glucose homeosta-
sis. Adipocyte products may promote glucose uptake,
e.g., leptin, adiponectin, omentin, visfatin, or impede glucose
uptake, e.g., TNF-a, interleukin (IL) 6, resistin, and retinol-
binding protein 4 (12) (Fig. 2).
Chronic Inammation
A likely bridge between obesity and insulin resistance is the
inammatory response (82). Obesity is a chronic inamma-
tory state marked by an invasion of mononuclear phagocytes
or macrophages into adipose tissue and by an increase in
proinammatory cytokines not seen in lean individuals
(83). There is a 40% increase in macrophage inltration,
which is positively correlated (along with body mass index
[BMI]) with adipocyte size (83). Thirty percent of protein
products expressed in white adipose tissue is related to in-
ammatory and macrophage-specic genes (84). Precursors
for macrophages, most likely derived from the bone marrow,
are attracted to increasing numbers of adipocytes as a result
of an overexpression of the chemotaxin gene for monocyte
chemotactic protein 1 (82, 83, 85).
Proinammatory cytokines also are up-regulated in obesity.
They are released from adipose tissue and macrophages and
include IL-6, TNF-a, and C-reactive protein (CRP) (82, 83).
As mentioned previously, IL-6 is secreted by macrophages
as well as adipocytes and may be causally related to insulin
resistance (1, 86).
Tumor necrosis factor a originates primarily from
macrophages and in turn stimulates macrophage activity
TABLE 1
Hormone effects in adipose tissue.
Hormone Action Clinical example(s)
Insulin (36, 42) Potent antilipolytic Diabetes?
Cortisol (in the presence of
insulin) (42, 52)
Increased in visceral
fat and central obesity
Cushing syndrome,
polycystic ovary syndrome
Catecholamines (36) Lipolytic, but action
depends on the balance
of b- and a-adrenergic
receptors
Activated in fasting
and exercise
Androgens (20, 327, 332, 333,
336)
Promotes central obesity Polycystic ovary
syndrome, postmenopause,
men, female-to-male
transsexuals
Estrogens (50, 51, 334, 335, 454) Decreases central
fat; increases femoral-gluteal
fat
Reproductive-age women,
postmenopause estrogen
administration
Growth hormone (34, 36, 37) Decreases central
fat; opposes action
of cortisol insulin
Acromegaly
TNF-a (36, 53, 233) Lipolytic; promotes
insulin resistance
Increased with obesity
Note: TNF tumor necrosis factor.
Bohler. Adipose tissue and reproduction in women. Fertil Steril 2010.
798
Bohler et al. Adipose tissue and reproduction in women Vol. 94, No. 3, August 2010
(82, 83, 87). TNF-aexpression also is increased in obesity and
inhibits insulin-stimulated glucose uptake (1, 36, 37, 86).
However, the clinical signicance of augmented adipose tis-
sue levels of TNF-a in humans is not clear (88).
Another marker of chronic inammation is highly sensi-
tive CRP (hs-CRP), which is produced in hepatocytes and ex-
hibits inammatory effects (89). It is activated by cytokines
and is positively associated with obesity, fasting insulin,
and insulin resistance (90). Serum hs-CRP levels are an inde-
pendent marker for cardiovascular disease (89). Sex hor-
mones may affect hs-CRP levels. Transdermal estrogen
may lower hs-CRP levels perhaps through IL-6 and TNF-
a (89). Moreover, hs-CRP levels vary during the menstrual
cycle in normal-weight and overweight women, with increas-
ing levels in the early follicular phase and decreasing levels
during the late follicular phase when E
2
levels are increased.
Excess visceral adiposity is a predictor for increased in-
ammatory markers (91). In a population-based study,
women with a waist-to-hip ratio (WHR) >0.8 were more
likely to have increases in inammatory markers than those
diagnosed with obesity based on a BMI >29.9 kg/m
2
(43%
compared with 15%). Compared with those with normal
body fat distribution, women with central fat exhibited
53% higher CRP levels, 30% higher TNF-a levels, 17%
higher white blood cell counts, and 42% higher IL-6 levels
(all P<.05) (91).
ADIPOSE TISSUE PRODUCTS (ADIPOKINES)
Awide variety of proteins and other products are produced by
adipose tissue. Some of these are produced by adipocytes,
whereas others are produced by adipose stromal cells, which
are important in adipocyte development, or from macro-
phages which migrated to adipose tissue in obesity (92)
(Table 2). Collectively, these products are referred to as adi-
pokines or adipocytokines, even though not all are classic cy-
tokines or from adipocytes (1, 9, 34, 9294). Adipokine
quantities may vary by metabolic state (e.g., morbid obesity),
fat compartment (visceral fat compared with subcutaneous
fat), and cell development (preadipocytes compared with ma-
ture adipocytes) (9, 95). Some adipokines are released in sub-
stantial amounts into the circulation (e.g., leptin), whereas
others, such as TNF-a, remain primarily localized to adipose
tissue and function as paracrine or autocrine regulators (52).
Several adipokines, such as leptin, adiponectin, IL-6, plas-
minogen activator inhibitor (PAI) 1, and TNF-a, are known
to inuence the functions of reproductive tissues and will
be discussed here in detail, whereas others, such as omentin,
vaspin, and acylation-stimulating protein have been reviewed
in detail elsewhere (9, 52, 92, 94).
Leptin
Leptin is a 167amino acid polypeptide that is synthesized
predominantly by fat cells but is also expressed in stomach,
placenta, and mammary gland (96100). A mutation of the
gene (ob/ob) that produces morbidly obese mice led to its
discovery, (7). The ob/ob gene was cloned in 1994 and the
16-kDa protein product was named from the Greek leptos,
meaning thin, because leptin was initially thought to act pri-
marily as an antiobesity protein (7, 101). Morbid obesity may
occur in human subjects from congenital leptin deciency or
from a mutation in the leptin receptor, but these are rare
conditions (102, 103).
Leptin circulates in free and bound forms, and total plasma
leptin represents the sum of both (104). Like other plasma
binding proteins, it protects its ligand from rapid clearance,
and may have other functions (105). Leptin is not appreciably
stored, but instead is constitutively secreted by adipocytes;
the larger the adipocyte, the more leptin it secretes (106).
Circulating levels are increased in proportion to body fat,
which coincides with its most important function: to relay
energy status (73, 76, 101, 107).
Similar in structure to cytokines, leptin and its cognate
receptor belong to the cytokine-receptor family activating
primarily the Janus kinase 2/signal transducer and activator
of transcription 3 pathways (73, 108). The soluble leptin re-
ceptor, the major leptin-binding protein in plasma, is created
by the shedding of the ectodomain of the membrane receptor
(105, 109, 110). Leptin receptors are expressed in the hypo-
thalamic arcuate nucleus, anterior pituitary, T lymphocytes,
FIGURE 2
Adipokines that promote or inhibit glucose uptake.
Adipocytes secrete proteins with variable effects on
glucose homeostasis. Adipocyte-derived proteins
with antihyperglycemic action include leptin,
adiponectin, omentin and visfatin. Other factors tend
to raise blood glucose, including resistin, TNF-a and
RBP4. TNF-a and human resistin are probably
secreted by cells other than adipocytes within the fat
pad. TNF-a tumor necrosis factor; IL, interleukin;
RBP retinol binding protein. Modied and used
with permission from Rosen ED, Spiegelman BM.
Adipocytes as regulators of energy balance and
glucose homeostasis. Nature 2006;444:84753.
Bohler. Adipose tissue and reproduction in women. Fertil Steril 2010.
Fertility and Sterility

799
vascular endothelial cells, pancreas, ovary, and endometrium
(110116).
Exhibiting a pulsatile and diurnal pattern of release, leptin
levels peak at 0100 hours and reach a nadir at 0900 hours
(117, 118). The number of pulses is related to BMI and to
pulse amplitude, but the mechanism controlling pulsatile lep-
tin release is still unknown, although it is independent of go-
nadal steroids and glucose (119).
Although serum leptin levels are positively correlated with
fat mass, they fall acutely with fasting, even before weight or
percentage body fat decrease (1, 120). Despite leptins re-
pressive effect on food intake, obese patients have high serum
leptin levels and may be leptin resistant, similar to the in-
sulin resistance that is often seen in obesity (108, 121). Other
factors that inuence serum leptin levels include gender, sex
steroids, and fat distribution (122124) (Table 3). There is
a differential expression of leptin in subcutaneous and vis-
ceral adipose tissue (125). In vitro studies have demonstrated
that leptin secretion from SAT is signicantly higher than
from intra-abdominal omental fat, in both obese and nonob-
ese individuals (116). Leptin transcripts follow the same pat-
tern, probably owing to a larger cell size in subcutaneous fat
(52). Because subcutaneous fat comprises 80% of total body
fat, SAT is considered to be the most important source of
leptin (125).
Estrogens stimulate the production of leptin from adipose
cells from women, but not from men, and increase leptin
mRNA in ovariectomized rats (126). Ovariectomy in rats re-
duced leptin mRNA in white adipose tissue, and the effect
was reversed if rats were given estrogen supplementation
(124). Women have higher leptin concentrations in serum
and in cerebral spinal uid than men, partly because andro-
gens suppress leptin expression (127, 128). It is also likely
that estrogens increase the sensitivity to leptin in the brain
(61, 62) (Fig. 3).
Leptin plays a role in glucose homeostasis. Leptin receptors
have been localized in rodent and human pancreatic b-cells,
and leptin administration inhibits preproinsulin gene tran-
scription and reverses hyperinsulinemia in leptin-decient
ob/ob mice before effects on body weight (129, 130). Cultured
human pancreatic islet cells also demonstrated a suppression
of preproinsulin gene expression and secretion when treated
with leptin (129).
Insulin, on the other hand, increases rodent and human ob
mRNA in vivo and in cultured adipocytes (131). Acute
changes only were seen in rodents, whereas prolonged insulin
administration in human subjects was associated with a rise in
leptin levels invivo and an increase in ob mRNAand leptin se-
cretion from cultured adipocytes (132, 133). Thus, there is an
adipoinsular axis involved in serum glucose control (134).
Serum leptin levels in girls begin to rise between ages 7
and 8 years and reach peak levels by age 1315 years (135,
136). Although the timing of these changes suggests a role
for leptin in pubertal development, it remains controversial
whether increasing leptin initiates puberty in primates
(137140). Instead, leptin may play a permissive role in
this process (135, 138, 140, 141).
After menarche with the onset of menstrual cycles, serum
leptin levels parallel LH and E
2
(117, 142). Leptin levels are
at their nadir during the early follicular phase and increase
across the menstrual cycle, peaking during the luteal phase.
At this time, leptin receptors in the endometrium are also at
their peak (142145).
Leptin is found in follicular uid, and receptors for leptin
have been localized in the preovulatory follicle and mature
oocyte, as well as in granulosa, theca, and interstitial cells
(114). There is in vitro evidence that leptin may act as a gate-
way for steroidogenesis. Huang et al. demonstrated that leptin
inhibits FSH/insulin-like growth factor (IGF) 1 stimulation of
TABLE 2
Adipokines and their sites of origin in adipose tissue.
Site of origin in adipose tissue
Adipokines Adipocytes Macrophages Stromal cells
Leptin (7)
Adiponectin (8)
Visfatin (158)
TNF-a (36)
IL-6 (9) ?
PAI-1 (456) (9)
Resistin (216, 217, 457)
Retinol-binding protein (455)
Omentin (458)
Note: IL interleukin; PAI plasminogen activator inhibitor; TNF tumor necrosis factor.
Bohler. Adipose tissue and reproduction in women. Fertil Steril 2010.
800
Bohler et al. Adipose tissue and reproduction in women Vol. 94, No. 3, August 2010
E
2
in rat granulosa cells, and leptin inhibits insulin-induced P,
E
2
, and A secretion (146149).
Leptin may play a role in embryonic development, because
leptin receptors have been localized in preimplantation em-
bryos (143). Maternal and serum fetal levels are elevated in
term pregnancies (150).
Adiponectin
Discovered shortly after leptin in1995, adiponectin is a 30-kDa
protein (with 244 amino acids) that is the most abundant se-
creted protein expressed exclusively by adipocytes (151). Its
globular form is similar in structure to complement proteins
(8, 151153). It circulates in the blood in three main forms:
a low-molecular-weight (trimeric) form, a medium-molecu-
lar-weight (hexameric) form, and a high-molecular-weight
(dodeca- to octadecameric) form. The different forms have
varying effects on target tissues, with the high-molecular-
weight form affecting the liver and endothelial cells (154).
Factors that affect circulating adiponectin levels are not
fully understood, but adiponectin is secreted in pulses and
follows a circadian pattern in the circulation, declining at
night and reaching its nadir in the early morning hours
(155, 156). Circulating levels of adiponectin (825 mg/mL)
are higher in women than in men (157). It is known that an-
drogens decrease adiponectin secretion, but the mechanisms
of regulation by sex hormones remain undened (158, 159).
Other factors that might increase adiponectin levels include
dietary soy proteins and sh oils, whereas adiponectin levels
are decreased by oxidative stress and high-carbohydrate
meals (160, 161).
At least two distinct receptors have been identied for
adiponectin: AdipoR1 and AdipoR2 (162). These have the
typical seven-transmembrane structure but differ from
G-proteincoupled receptors in that the C-terminal portion
is extracellular whereas the N-terminal is cytoplasmic (163,
164). The main intracellular signaling pathway is through
AMP-activated kinase, but several other pathways are also
involved. Adiponectin receptors have been demonstrated
in reproductive tissues, including ovary, placenta, and endo-
metrium (165). In addition, the pituitary expresses both
adiponectin and its receptors, and adiponectin stimulates
LH secretion in vitro (166).
The chief action of adiponectin is to increase insulin-
stimulated glucose uptake in muscle and liver by decreasing
serumFFAs and triglycerides and by suppressing hepatic glu-
cose production (36, 164). Effects on fatty acid oxidation and
reduced TG accumulation in muscle could enhance muscle
insulin sensitivity (167).
Although it is an adipocyte-derived hormone, adiponectin
levels in serum actually decrease with obesity and increase
with weight reduction (83, 155, 163, 167). Adiponectin levels
are affected by a variety of pathologic states in addition to
obesity, including diabetes mellitus, cardiovascular disease,
metabolic syndrome, insulin resistance, and hypertension
(156, 168). Renal failure, congestive heart failure, and thiazo-
lidinedione drugs increase adiponectin levels (169).
Ledoux et al. (170) have recently demonstrated that in por-
cine granulosa cells, adiponectin at physiologically relevant
levels provokes expression of genes associated with periovu-
latory remodeling of the ovarian follicle. These genes include
cyclooxygenase-2, prostaglandin E synthase, and vascular
endothelial growth factor. Adiponectin interacts with both
LH and insulin to induce expression of cyclooxygenase-2
transcripts in granulosa cells (165).
The potential interaction between adiponectin, insulin, and
FSHin the regulation of periovulatory ovarian follicular gene
expression was recently summarized by Campos (94). She
postulated that the adapter protein APPL1 may play a key
role in the cross-talk between adiponectin, FSH, and insulin
receptor signaling in the ovary. Although overexpression of
adiponectin in mice results in infertility as well as improved
insulin sensitivity, adiponectin knockout has not been shown
to have a signicant impact on insulin sensitivity or fertility
(171).
Pregnancy is an insulin-resistant state, and gestational
diabetes complicates 3%12% of all pregnancies (172).
TABLE 3
Factors affecting leptin levels.
Factor Leptin level
Adipocyte mass
(87, 459)
Increased
Adipocyte size (7, 86) Increased
Adipocyte distribution
(visceral vs.
subcutaneous fat)
(105)
More from
subcutaneous fat
(total volume and
secretion are greater)
Secretion pattern
(97, 98)
Diurnal variation:
low at 0900 hours,
peak at 0100 hours
Glucocorticoids
(86, 460)
Increased
Insulin (111, 460) Controversial, but
possibly increased
Cytokines (TNF-a, IL-1)
(460)
Increased
Gender (103, 460) Higher levels in women
Sex steroids (104, 106) Increased with
estrogen; decreased
by androgens
Acute caloric
restriction (100)
Decreased
Acute caloric
consumption (112)
Increased
Note: Abbreviations as in Table 2.
Bohler. Adipose tissue and reproduction in women. Fertil Steril 2010.
Fertility and Sterility

801
Therefore, several studies have measured adiponectin levels
in pregnancy and gestational diabetes (173). Nien et al. (86)
reported no signicant differences in the median adiponectin
concentration between pregnant and nonpregnant women.
Plasma adiponectin concentrations were negatively corre-
lated with gestational age only among normal-weight preg-
nant women, whereas overweight pregnant women had
signicantly lower plasma adiponectin concentrations than
normal-weight women (86). Consistent with the increased
insulin resistance and weight gain that occur in pregnancy,
adiponectin concentrations correlated negatively with gesta-
tional age (174).
Vascular dysfunction is a major feature of preeclampsia.
High levels of circulating adiponectin have been reported
in preeclampsia and may be a compensatory mechanism to
protect the vasculature (86, 174).
Obesity has been associated with an increased risk of endo-
metrial cancer (175). Circulating adiponectin levels are in-
versely associated with the risk of endometrial cancer in
pre- and postmenopausal women (175). This relationship
remains signicant after adjustment for BMI and other
obesity-related risk factors, such as C-reactive protein,
IGF-1, IGF-binding proteins 1 and 2, SHBG, T, and estrone
(175).
Visfatin
Another adipokine with ties to reproduction is visfatin. This
52-kDA protein is expressed in a variety of cell types and
tissues, including adipose tissue and adipocytes, lympho-
cytes, bone marrow, liver, muscle, trophoblast, and fetal
membranes (88).
Because of the metabolic importance of visceral fat, Fuku-
hara et al. (88) sought to identify a secretory product which
might be unique to intra-abdominal adipose tissue. Using
a differential display method, they compared visceral fat
products with those of subcutaneous tissue and found a pro-
tein whose mRNA was much more abundant in visceral fat
(therefore the name, visfatin). Their work also revealed that
visfatin levels in mice and humans were in direct proportion
to the amount of visceral fat measured by computerized to-
mography (CT), a correlation not seen with subcutaneous
fat, and that plasma visfatin levels were elevated with obesity.
FIGURE 3
Leptin effects on the reproductive axis. When fat stores are adequate, leptin stimulates GnRHrelease. This action
is most likely indirect through other neurohormones, including kisspeptin, neuropeptide-y, and pro-
opiomelanocortin. Leptin and its receptor are also found in the pituitary. Whether estrogen has a physiologic role
in releasing leptin from fat stores is unclear. Receptors for leptin have been localized in the ovary and
endometriumimplying additional direct actions on these tissues. GnRHgonadotropin releasing hormone; LH
luteinizing hormone; FSH follicle stimulation hormone; KISS kisspeptin; POMC pro-opiomelanocortin;
NPY neuropeptide-Y.
Bohler. Adipose tissue and reproduction in women. Fertil Steril 2010.
802
Bohler et al. Adipose tissue and reproduction in women Vol. 94, No. 3, August 2010
The visfatin protein actually had been previously character-
ized as a growth factor for early-stage Bcells, and was known
as preB-cell colonyenhancing factor (89).
Visfatin was reported to have insulin-like properties (88).
In mice, recombinant visfatin lowered plasma glucose levels,
and mice with one mutant copy of the visfatin gene had
higher plasma glucose levels compared with wild-type
mice. Furthermore, visfatin was shown to bind to the insulin
receptor but at a different site, producing downstream effects
unlike those of insulin (88). The relationship between visfatin
and obesity and insulin action is presently less clear, however,
because there has been a retraction of a portion of the work
originally published in Science (176). Subsequent studies
have not conrmed the high level of expression in visceral
fat or the correlation between circulating levels and measures
of adiposity (177, 178). Some of the conicting ndings may
be due to the presence of many forms of visfatin in plasma
that are variably detected by immunoassays, and the connec-
tion to obesity may instead relate to inammation and other
cytokines (178, 179).
As mentioned previously, pregnancy is characterized by
a progressive increase in insulin resistance, and women
who fail to compensate with a sufcient increase in insulin
secretion develop gestational diabetes. Visfatin levels were
reported to progressively increase throughout gestation in
normal pregnancy, and rst-trimester levels predicted the in-
sulin sensitivity in the second trimester (180). Interestingly,
plasma visfatin levels in third-trimester healthy women
were not signicantly different from values in healthy non-
pregnant controls. Moreover, visfatin levels have been re-
ported to be both increased and decreased in gestational
diabetes (181, 182).
Insulin resistance increases the risk for preeclampsia, and
serum visfatin levels were reported to be reduced in women
with preeclampsia and further reduced with severe disease
(183). On the other hand, visfatin levels were increased in
women in the third trimester in pregnancies characterized by
intrauterine growthretardation(184). These associations await
conrmation, and their importance remains to be determined.
Tumor Necrosis Factor Alpha
Tumor necrosis factor a is constitutively expressed by many
cell types, including macrophages, lymphocytes, and adipo-
cytes (36). It is a 26-kDA transmembrane protein that was
originally found to cause necrosis of tumors (185). The bioac-
tive form is a 17-kDa cleavage product which interacts with
either type I or II TNF-a receptors in adipose tissue
(1, 186). Macrophages, whose numbers increase with an
increase in fat cell mass, are the predominant source of
TNF-a (187189). The production of TNF-a also parallels
fat cell size (1). The quantity of adipose tissue mass may be
partly determined by TNF-a because of its ability to impair
human preadipocyte differentiation, increase adipocyte apo-
ptosis, and increase lipolysis (27, 190, 191). In massively
obese women (BMI >45 kg/m
2
), TNF-a was more abundant
in SAT than in VAT, but the difference became insignicant
with a BMI <32 kg/m
2
(9).
Sex steroids may inuence TNF-a production. Circulating
levels were unchanged during the menstrual cycle, despite
signicant changes in hs-CRP insulin resistance, determined
by homeostatic model assessment (HOMA), and SHBG
levels (192). On the other hand, TNF-a release was signi-
cantly increased in cultured bone marrow cells in early post-
menopausal compared with younger women and may have
been decreased by estrogen treatment (193).
One of the mechanisms of insulin resistance in obesity may
be inhibition of insulin signaling by TNF-a (194, 195).
TNF-a impedes insulin signaling by inhibiting tyrosine
kinase activity and favoring serine phosphorylation of insulin
receptor substrate 1 (196). Furthermore, TNF-a decreases ad-
ipocyte secretion of adiponectin in vitro and suppresses Glut-
4 glucose transporters and insulin receptors (190, 197, 198).
Tumor necrosis factor a injected into the brain in rats
reduces GnRH and LH secretion, and thus it may be partly
responsible for the gonadotropin deciency that accompanies
acute illness (199). TNF-a also inhibits gonadotropin activa-
tion of steroidogenesis in the ovary as well as the testis and
adrenal cortex through the inhibition of cAMP production
and downstream signaling (1).
Interleukin-6
Interleukin-6 is a cytokine with multiple forms ranging in
size from 22 to 27 kDa (1, 200). Like other adipokines, IL-
6 binds to a transmembrane receptor (IL-6R) which is homol-
ogous to the leptin receptor and is expressed by adipose tissue
although at a higher level in hepatocytes). IL-6 associates
with two molecules of gp130 protein to initiate intracellular
signaling (1, 9, 201, 202).
One-third of circulating IL-6 is accounted for by adipose
tissue, and serum levels parallel adipocyte size and fat mass
(1). Omental fat produces threefold more IL-6 than does
SAT (9, 52, 194). IL-6 levels are positively correlated with
TGs, and IL-6induced release of TGs contributes to an in-
crease in serum FFAs (196). Although IL-6 levels increase
with obesity, mice decient in IL-6 develop obesity, and
intracerebral administration of IL-6 decreased body fat and
increased energy expenditure, suggesting actions at central
and peripheral sites (1). Levels of IL-6 are also increased by
TNF-a (200).
Interleukin-6 has multiple functions, but it is best known
for its association with inammation and insulin resistance
(200). It reduces downstream insulin receptor signaling, adi-
pogenesis, and adiponectin release (1, 52, 200, 203). In rat
pancreatic cells, IL-6 down-regulates glucose-stimulated in-
sulin release, but IL-6 administered to normal human subjects
increased plasma glucose without altering insulin concentra-
tions (196, 204). Finally, it is not clear whether IL-6 induces
or is the consequence of impaired glucose tolerance and insu-
lin resistance (196).
Fertility and Sterility

803
Willis et al. (205) investigated the production of proinam-
matory cytokines by monocytes in culture (including IL-6)
during ovulatory menstrual cycles. There was no signicant
change in IL-6 secretion during ovulation, although other cy-
tokines increased, but there was an increase in IL-6 produc-
tion in the luteal phase. This is in agreement with Van der
Hoek et al. (206) and others (83, 207209), who found no
role for IL-6 in ovulatory regulation based on experiments us-
ing perfused rat ovaries. OBrien et al. (207), however, noted
an increase in soluble IL-6 receptor in the luteal phase.
Minkunis ndings (210) depart fromthose above. In a per-
fusion system, IL-6 suppressed LH-induced ovulation as well
as FSH and LH-induced E
2
production from granulosa cells.
Deura et al. (211) also demonstrated a reduction in estrogen
production from human granulosa cells by IL-6.
The effect of IL-6 on hypothalamic and pituitary cells has
also been examined (212). Cultured rat pituitary cells and hy-
pothalamic explants known to contain GnRH neurons were
exposed to IL-6. IL-6 had no effect on GnRH release from
proestrus female hypothalamic explants, nor did it have an ef-
fect on GnRH-stimulated release of LH in dispersed pituitary
cells from proestrus females (212).
Endometriosis induces an inammatory response in the
peritoneal cavity, where an increase in peritoneal uid and
IL-6 levels has been documented in subfertile women
(213). The source of IL-6 in endometriosis is thought to be
macrophages and endometriotic stromal cells (214, 215).
IL-6 may also contribute to infertility by impairing sperm
transport. Yoshida et al. (214) demonstrated a signicant de-
crease in sperm motility and rapidly moving sperm in a dose-
dependent manner when the combination of IL-6 and soluble
IL-6R were added to sperm cultures.
Plasminogen Activator Inhibitor 1
Plasminogen activator inhibitor 1, a 50-kDa glycoprotein, is
the primary inhibitor of brinolysis and is expressed in liver,
endothelial cells, platelets, smooth muscle cells, and adipose
tissue (adipocytes and inltrating macrophages) (52, 216).
By inactivating urokinase-type and tissue-type plasminogen
activator, PAI-1 prevents the conversion of plasminogen to
plasmin, which reduces thrombin formation (52, 217). Ele-
vated levels of PAI-1 are associated with venous thrombosis,
cardiovascular disease, recurrent pregnancy loss, and preg-
nancy complications beyond the rst trimester (1, 217220).
Elevated PAI-1 levels are also associated with obesity, in-
sulin resistance, type 2 diabetes, dyslipidemia, hypertension,
and the metabolic syndrome (1, 10, 218, 221). A common
denominator in these conditions may be hyperinsulinemia,
because insulin as well as glucose, angiotensin II, and fatty
acids have been demonstrated to increase PAI-1 expression
(222). Weight loss, metformin, and thiazolidinediones, each
of which reduces insulin levels, decrease PAI-1 (1, 10,
222). Interestingly, PAI-1 may contribute to the development
of obesity inasmuch as PAI-1 knockout mice fail to accumu-
late fat even when overfed (223). Visceral fat production of
PAI-1 exceeds that of subcutaneous adipose tissue, especially
in subjects with visceral adiposity (218, 224).
Plasminogen activator inhibitor 2 is sometimes called the
placental PAI, because it is expressed at high levels by tropho-
blastic epithelium and is found in plasma only during preg-
nancy (216). An increase in the PAI-1/PAI-2 ratio in maternal
plasma is one biochemical marker of preeclampsia (225).
Resistin
During their study of the nuclear receptor PPAR-g, using thia-
zolidinedione antidiabetic drugs to study glucose homeostasis,
Steppan et al. found that a gene, later designated as Retn, was
suppressed in murine adipocytes (3, 226). They named the
protein productsecreted as a 94amino acid polypeptide
resistin for resist insulin, owing to its association with
insulin resistance, at least in mice. The product was simulta-
neously discovered by two other groups (227). Serum levels
of resistin are increased in diet-induced obese mice as well
as in those with genetic forms of obesity, e.g., leptin (ob/ob)
and leptin receptor (db/db) decient mice (3, 226, 228).
Further evidence to support a role for resistin as a link be-
tween obesity and insulin resistance includes a decrease in
blood glucose in mice treated with resistin antibodies, an im-
pairment of glucose tolerance in those treated with recombi-
nant resistin, and a decrease in resistin synthesis in response
to the PPAR agonist pioglitazone which improves insulin
sensitivity (226, 229).
Resistin belongs to a family of cysteine-rich resistin-like
molecules that include found in inammatory zone pro-
teins (1, 12, 230). Although localized in adipocytes in
mice, resistin levels in human adipocytes are low and may
not originate from adipocytes but rather from other resident
cells in adipose tissue, including macrophages and stromal
cells (12, 82, 84, 171, 231). Resistin is also produced in pan-
creatic islet cells and possibly muscle (84, 85, 229). More-
over, resistin mRNA levels are greater in human peripheral-
blood mononuclear cells than in adipocytes (229).
Resistin appears to impair insulin sensitivity in rodents, but
its role in human insulin resistance is unclear. Serum levels of
resistin in patients with insulin resistance or type 2 diabetes
mellitus are not consistently different from normal, although
a positive correlation between resistin with body fat mass has
been found (84, 85, 90, 91, 171, 228). Resistin levels in hu-
man peripheral blood mononuclear cells are increased by
proinammatory cytokines, including IL-1, IL-6, and TNF-
a, and are reduced by thiazolidines, providing a possible
link between inammation and insulin resistance (82, 229).
Sex steroids may affect the levels of resistin in adipose tis-
sue (depending on the fat depot), and these levels may or may
not parallel plasma levels. In rats, P increased Retn expres-
sion in retroperitoneal white adipose tissue, with a simulta-
neous increase in plasma concentrations, but not in
parametrial or subcutaneous fat depots. Estrogen decreased
804
Bohler et al. Adipose tissue and reproduction in women Vol. 94, No. 3, August 2010
Retn expression in some fat depots, but without a change in
plasma concentrations (232). On the other hand, Chen et al.
(233) found that 17b-estradiol up-regulated Retn expression
in 3T3-L1 adipocytes, increasing with dose and time.
Pregnancy is associated with an increase in sex steroids as
well as insulin resistance, and some have hypothesized that
resistin may be an insulin antagonist during pregnancy. Se-
rum resistin levels are elevated in pregnancy, may increase
with gestational age, and decline postpartum, and the princi-
pal source of the rising resistin levels may be the placenta
rather than adipose tissue (173, 234236).
Chen et al. (237) reported that when pregnancy is compli-
cated by gestational diabetes, resistin concentrations are
higher than in nondiabetic pregnant control subjects, whereas
Megia et al. (238) found lower resistin levels in gestational
diabetes compared with pregnant women with normal glu-
cose tolerance, indicating a need for further study.
Because there might be a link between resistin, insulin re-
sistance, and PCOS, Seow et al. (239) examined serum and
follicular uid levels of resistin in women with PCOS, includ-
ing those with insulin resistance who were undergoing IVF
cycles, and found no signicant difference from control sub-
jects although resistin mRNAin adipose tissue was increased.
On the other hand, Munir et al. (240) found a 40% increase in
mean serumresistin concentrations in women with PCOS and
a positive correlation with BMI and T. In addition, resistin was
found to increase ovarian androgen production by directly
stimulating ovarian theca cells, more so in the presence of
insulin. Resistin mRNA in adipose tissue as well as serum
T and glucose-stimulated insulin secretion was found to
decrease after laparoscopic ovarian electrocautery (241).
Furthermore, in a case-control study of infertility patients
treated with IVF, serum resistin levels were inversely corre-
lated with the number of oocytes retrieved, but, interestingly,
only in non-PCOS patients, and there was no correlation with
BMI or body weight (242). In summary, resistin is an impor-
tant adipokine, and its role in female reproduction is a subject
of ongoing studies.
FREE FATTY ACIDS
Plasma levels of FFAs are elevated in obesity partly because
of the corresponding increase in TNF-a, which stimulates
lipolysis (36). An increase in FFAs leads to lipid deposition
in skeletal muscle and liver and contributes signicantly to
insulin resistance (52, 203, 243). Consequently, FFAs cause
a decrease in glucose uptake peripherally and an increase
in hepatic glucose production (203, 243). Because of its
portal drainage, visceral fat is thought to contribute a greater
quantity of FFAs to the hepatic circulation (22, 244, 245).
FFAs are re-esteried in the liver, leading to steatosis, and
the resultant inammation further increases insulin resistance
and hyperinsulinemia (244, 246). FFAs also suppress insulin
secretion from pancreatic b-cells (247, 248). Finally, FFAs
added to cultured human granulosa cells decreased cell sur-
vival by causing apoptosis, and thus FFAs could contribute
to anovulation (249).
ADIPOSE TISSUE DISTRIBUTION
Upper body or central obesity refers to excess fat above the
waistline and includes extra-abdominal SAT lying just be-
neath the skin and intra-abdominal, or VAT (52). Fat below
the waistline, obviously, is subcutaneous. Altogether, the to-
tal volume of SATis greater than VAT (53). VATis composed
of mesenteric, omental, and retroperitoneal fat (52). This
includes fat deposits which surround and protect organs
such as kidneys and heart (23, 28). The products of VATenter
the portal blood system and have direct access to the liver,
whereas the venous drainage of SATis into the peripheral cir-
culation (22, 23). VATaccumulation is governed by genetics,
diet, energy expenditure, age, race, steroid hormones, and
gender (22). VAT increases with age and weight gain and
decreases with exercise and weight loss (22, 23). Generally,
men tend to accumulate more upper body fat, including
VAT and women have more lower body fat (27) (Table 4).
African-American women may have less visceral fat than
caucasian women (250255). In the Heritage Family Study,
Stanforth et al. (256) compared fat distribution between black
and white adults aged 1765 years using CT scanning and
found less VAT and more SAT in African-American than in
caucasian women, despite a higher BMI in the African Amer-
icans. Weinsier et al. (251) tracked the effect of weight loss
on VAT and SAT stratied by weight (overweight, normal
weight, and never overweight) in women with a mean age
of 36 years. In all three groups, intra-abdominal fat was
less in black than in white subjects, whereas SATwas greater
in only the black overweight women. In a longitudinal study
with a follow-up to 4 years, in which total body fat was sim-
ilar between white and black women at baseline, VAT was
less in black women at baseline and at follow-up. The in-
crease in VAT was more signicant than the increase in total
body fat over time in both groups (257). Perry et al. (258)
found higher VAT in obese white women compared with
obese black women matched for age, BMI, and percentage
body fat using magnetic resonance imaging (MRI).
In contrast, in the Coronary Artery Risk Development in
Young Adults study, VAT and total fat were greater in young
adult African-American women (259). Lovejoy et al. (250,
260) conducted two studies, one in which the mean age of
women was 35 years, and a second in which black and white
women were middle-aged. In those studies, there was no sig-
nicant racial difference in VAT (although in both studies
VAT was slightly less and SAT signicantly greater in black
women (250, 260). Differences in adipose tissue distribution
have been reported for other racial groups as well. Hispanics,
Filipinos, Japanese, and Ethiopians have dissimilar fat distri-
butions compared with each other and with caucasian women
(261264).
There are several different methods used to measure total
body fat and fat distribution. All have their advantages and
Fertility and Sterility

805
disadvantages, including practicality, complexity, cost, accu-
racy, and the need for an experienced anthropometrist. The
easier methods have been used to predict clinical outcome
and are correlated with the gold standards of measuring
body fat, including the measurement of body density, body
water, and body potassium(265). However, the gold standards
do not always agree and are themselves not direct measures of
body fat (265, 266). Additionally, they are better suited for use
in research than for clinical assessments.
Some of the better known and simplest means for estimat-
ing body fat and fat distribution include measurement of
BMI, waist circumference (WC), WHR, and abdominal sag-
ittal diameter (ASD) (265, 268). BMI is the ratio of weight in
kg to the square of height in meters, and is a reliable method
of estimating body fat percentage, particularly if age and gen-
der are considered (266268). A BMI of R30 denes obesity
by the World Health Organization and the U.S. National
Institutes of Health (266, 269). Because abdominal adiposity
may be more important biologically than total body fat, BMI
may not be as accurate in predicting metabolic disturbances
or reproductive dysfunction as indexes that quantify fat distri-
bution (268, 270, 271). Additionally, the reference standards
for BMI do not apply to all ethnic groups, which affects
research on clinical implications (266, 270, 272).
Waist circumference and ASD may be the most accurate
clinical indexes for estimating intra- and extra-abdominal
fat (273). While the person is standing, WC is measured
with a tape around the smallest portion of the torso, halfway
between the lowest rib and the iliac crest. The abdomen
should be bare, the patient relaxed, and the measurement
taken after an exhalation (274). AWC of R80 cm is associ-
ated with increased morbidity in women, but standards vary
with different ethnic groups (265). When estimating the
quantity of intra-abdominal fat, WC comes closer to the
true value than does WHR (273): Using a tape measure, the
widest part of the hips and buttocks are measured; the waist
is measured as described above. AWHR >.85 for women de-
nes abdominal obesity. ASD is measured with a Holtain-
Kahn abdominal caliper at the level of the iliac crest (275).
The gold standards for measuring intra-abdominal fat or
VAT are CT scanning or MRI (276278) (Fig. 4). Several
studies suggest that only one image is needed, preferably at
the L4-L5 level, to quantify VAT (279281). However, Tul-
loch-Reid et al. (282) concluded that this method is inferior
to measuring VAT volume by using multiple CT slices, be-
cause more detailed imaging correlated with the insulin
sensitivity index, whereas a single slice did not. Because of
radiation exposure by CT and the cost of both methods,
MRI and CT are more valuable as research tools than for
clinical applications.
ADIPOSITY AND PUBERTY
The environment, socioeconomic status, genetic factors,
nutrition, and body fat inuence the onset and progression
of puberty (283285). Fat accumulation accelerates during
puberty in girls and plays a major role in reaching pubertal
milestones (283, 286). Frisch and McArthur (68) were among
the rst to propose that body fat was not only needed for the
inception of menstrual cycles but also for their maintenance
and return in subjects who were amenorrheic with weight
loss. Many subsequent studies have concluded that the earlier
TABLE 4
Comparison between visceral and subcutaneous adipose tissue.
Characteristic
Visceral adipose
tissue (VAT)
Subcutaneous
adipose tissue (SAT)
Location (42) Intra-abdominal Extra-abdominal and peripheral
Drainage (42) Drained by the portal venous
system
Drained by the systemic circulation
Lipolytic activity (22) Higher mobilization of FFAs Lower
Catecholamine-induced
lipolysis (36)
More responsive Less responsive
Antilipolytic effect by insulin (461) Weaker Stronger
Expression of b-adrenoceptors
(lipolytic) (36, 461)
Increased Decreased
Expression of a2-adrenoceptors
(antilipolytic) (36, 462)
Decreased Baseline
Insulin receptor afnity and insulin
receptor substrate 1 (36)
Less than SAT Baseline
LPL activity (42) Lower Baseline
Note: FFAs free fatty acids; LPL lipoprotein lipase.
Bohler. Adipose tissue and reproduction in women. Fertil Steril 2010.
806
Bohler et al. Adipose tissue and reproduction in women Vol. 94, No. 3, August 2010
onset of puberty in girls is linked to nutrition and the increas-
ing prevalence of obesity (287). Moreover, others have shown
that the amount or percentage of body fat may not be as
important as where the fat is distributed (288, 289).
Although the age of menarche declined fromthe early 1800s
to the mid-1950s, it has seemingly reached a plateau in all but
one group, African Americans (290, 291). African-American
girls have continued to experience a decline in menarcheal
age of 5.5 months over the last 30 years and, not surprisingly,
they now have an earlier mean age for thelarche: 8.9 years ver-
sus 10.9 yrs for caucasian girls (283, 290, 292). Body fat is one
factor that may dene the difference in sexual maturation be-
tween African-American girls and their caucasian counterparts
(293). African-American girls on average are heavier than cau-
casian girls, even after adjusting for socioeconomic status, and
tend to have less VAT than caucasians (252, 293, 294).
The mechanisms through which nutrition, growth, and adi-
pose tissue inuence pubertal development are complex and
not fully understood; however, there is substantial evidence
linking leptin to sexual maturation (see above). Leptin levels
in girls increase during the prepubertal years and continue to
rise during puberty in parallel with an increase in BMI
(131). This effect may be partly initiated by aromatase in adi-
pose tissue, which increases estrogen production, which in
turn induces leptin. In contrast, girls decient in leptin due
to rare loss-of-function mutations in the leptin gene or with
leptin receptor gene mutation have delayed puberty and are
gonadotropin decient (102, 295). Low body fat mass due to
extreme exercise, e.g., ballet dancers or gymnasts, oftenresults
in low circulating leptin levels and pubertal delay (118, 296,
297). Puberty is also disrupted and leptin levels are lowin con-
ditions with insufcient nutritional status, such as anorexia
nervosa (107, 298, 299). Leptin levels are higher in black girls
than white even after adjusting for fat mass (300).
When prepubertal female monkeys were treated with sub-
cutaneous recombinant human leptin daily for 18 months, LH
and E
2
were increased and the age of perineal swelling was
advanced compared with untreated control subjects (301).
In male monkeys, on the other hand, no change in daytime
serum leptin levels before the onset of puberty was found.
Furthermore, IV infusion of leptin into agonadal prepubertal
male monkeys for 16 days did not enhance GnRH-stimulated
LH secretion, although GH was increased, suggesting that
circulating leptin does not initiate puberty, at least in males
(302, 303). From these studies, it seems prudent to conclude
that the role of leptin in human pubertal development requires
further investigation.
Premature adrenarche is dened as the development of pu-
bic hair before the age of 8years in girls, and it is characterized
by a premature increase in adrenal androgen production (304,
305). Premature adrenarche is associated with obesity and ac-
celerated growth and with a history of low birth weight (306).
Girls with premature adrenarche are at a higher risk for devel-
oping the metabolic syndrome, type 2 diabetes, and cardiovas-
cular disease when they reach adulthood (304). These girls
have insulin resistance and hyperinsulinemia, low SHBG,
and higher levels of IGF-1, each of which may play a role in
the link between obesity and puberty (304). In girls of low
birth weight who developed precocious adrenarche, metfor-
min improved insulin resistance, reduced androgen excess,
and led to a reduction in total and abdominal adipose tissue
compared with untreated control subjects (307). Approxi-
mately 2%3%of girls with premature adrenarche have a mu-
tation in the CYP21 gene causing nonclassic 21-hydroxylase
deciency and androgen excess (308). Variable effects of lep-
tin on ACTH production have been reported, and although
leptin receptors are found in the adrenal, leptin may interfere
with, rather than activate, ACTH signaling (309, 310).
FIGURE 4
Computerized tomography showing cross-sectional abdominal areas at the umbilicus level in two women
demonstrating variation in fat distribution despite similar BMIs. A. 49-yr-old female with a BMI of 23; B. 40-yr-old
female with a BMI of 24. VAT visceral adipose tissue; SAT subcutaneous adipose tissue. FromWajchenberg
B. Subcutaneous and visceral adipose tissue: their relation to the metabolic syndrome. Endocr Rev 2000;21:
697738.
Bohler. Adipose tissue and reproduction in women. Fertil Steril 2010.
Fertility and Sterility

807
ADIPOSE TISSUE DEFICIENCY AND REPRODUCTION
Amajor functionof the secretoryproducts fromadipocytes is to
signal the adequacy of energy stores to the CNS for reproduc-
tive function. An adipose tissue threshold must be reached
to initiate and continue ovulatory cycles (68). When the fat
mass is too low, gonadotropin secretion is reduced and repro-
ductive capacity is decreased (311, 312). Adeciency of leptin
appears to be the key mediator of this effect (313). There is now
an abundant literature linking leptin to gonadotropin secretion
(147, 314, 315). Notably, leptin activates reproductive function
incongenital leptindeciency and restores gonadotropinsecre-
tion in leptin deciency associated with fasting. Puberty is also
advanced in leptin-transgenic skinny mice, which have no ap-
parent adipose tissue (316). Leptin stimulates the secretion of
GnRH and directly stimulates the pituitary to release LH
(115, 317). There is accumulatingevidence that kisspeptin neu-
rons in the anteroventral periventricular nucleus mediate the
nutritional signal linking leptin to GnRH. Kisspeptin neurons
express the leptin receptor, and leptin treatment normalizes
suppressed KISS-1 expression in rodents with gonadotropin
insufciency and low leptin, including the ob/ob mouse and
the streptozotocin-induced diabetic rat (317). Aleptin-induced
increase could mediate the increase in GnRH pulsatility that
follows the administration of recombinant leptin to low-weight
women with hypothalamic amenorrhea (311).
Lipodystrophy, the complete absence of subcutaneous adi-
pose tissue, is marked by severe insulin resistance, elevated
androgens, low gonadotropins, amenorrhea, elevated andro-
gens, and low leptin levels (318, 319). When these patients
are treated with recombinant leptin glucose tolerance im-
proves, androgen levels fall, and ovulatory menstrual cycles
resume, even before there is a change in weight or fat accumu-
lation (318, 319). Gonadotropin levels were unchanged, but
the response to GnRH was more robust, after therapy (319).
Leptin improved insulin sensitivity and was accompanied
by a decrease in insulin levels and an increase in SHBG(296).
Fat quantity is also severely reduced in patients with an-
orexia nervosa (299, 312). These patients experience a de-
crease in gonadotropins and E
2
and have low leptin levels
(313, 320). Puberty is delayed or arrested, and secondary
amenorrhea is common in those with established menses.
Some women continue to have menstrual cycles, and those
who menstruate have more body fat, especially truncal fat,
and higher leptin levels (312). Interestingly, about 20%of pa-
tients with anorexia nervosa experience amenorrhea before
the onset of weight loss. Menses generally resume when
weight increases to about 90% of ideal body weight, but
amenorrhea may persist when body fat is low or there are
unresolved emotional issues (321).
In fact, calorie restriction of any cause can disturb gonado-
tropin secretion and thereby ovulation (322). Sustained calorie
deprivation sufcient to cause weight loss leads to anovulation
in previously ovulatory women; however, short-term fasting
has a lesser effect on LHsecretion than in men (323, 324). Ad-
ministration of recombinant leptin reverses the fasting-induced
decrease in LH pulsatility in normal women (325).
Strenuous physical exercise may disrupt GnRH secretion
and produce menstrual dysfunction, including amenorrhea
(311). Athletes who experience amenorrhea tend to have re-
duced body fat and lower leptin levels (297, 326). Most often,
calorie intake is insufcient for the level of exercise, produc-
ing a negative energy balance. Treatment with recombinant
leptin can reactivate the reproductive axis when amenorrhea
results from low weight or strenuous exercise (311).
ADIPOSE TISSUE EXCESS AND REPRODUCTION
Obesity
Obesity has reached epidemic proportions in the United
States, affecting all age groups and races, including women
of reproductive age (19). The prevalence of obesity (BMI
R30 kg/m
2
) in women aged 2039 years stratied by ra-
cial/ethnic groups for 19992000, 20012002, and 2003
2004 is shown in Table 5 (19). Although there appears to
have been a decrease in obesity in caucasians, obesity has
consistently increased in African Americans and Mexican
Americans, with the former most affected (19).
Obesity is associated with many comorbidities, including
detrimental effects on reproductive health (327). Anovulation
and infertility occur more frequently in obese women com-
pared with nonobese women (26, 328330). Ovulation induc-
tion with either clomiphene citrate or gonadotropins may
require a higher dose and take longer to induce oocyte matu-
ration (331). With assisted reproduction, there may be
a decreased number of oocytes retrieved and fewer live births
(332, 333). Miscarriages may also be increased, but this
nding has been inconsistent (332, 333, 335).
The negative impact of obesity on reproduction may be re-
lated to a change in a variety of hormones starting as early as
the pubertal transition. In a cross-sectional analysis, obese
peripubertal girls were found to have an increase in total
and free T, a decrease in SHBG, and an increase in insulin
levels compared with normal-weight girls of the same age
(336). Other changes include an increase in DHEAS and
LH hypersecretion. Although there may be other factors re-
sponsible for a reduction in fecundity and successful preg-
nancy, the hormonal changes associated with obesity are an
important cause for interrupting normal ovulatory menstrual
cycles (17).
Hormone Proles and Effects in Upper and Lower Body
Obesity
Global adiposity has an important effect on an individuals
overall health and metabolic status, but compartmental fat
distribution appears to be the most important characteristic
(337). The distinction between UBO and lower body obesity
(LBO) was rst made by Vague in 1947 (20), because of the
changes in sex hormones and the comorbidities that accom-
pany UBO in women. Androgen and estrogen levels may
be increased with UBO and LBO, but UBO is more closely
linked to an androgenic prole; therefore, it is referred to
as android or central obesity (338, 339). Because the
808
Bohler et al. Adipose tissue and reproduction in women Vol. 94, No. 3, August 2010
balance of sex steroids in women is weighted toward estro-
gen, LBO is referred to as gynoid or peripheral obesity
(339).
Kirschner et al. (340) found an increase in the production
rates of T, DHT, and A in women with UBO and LBO com-
pared with nonobese controls, but women with UBO had
higher levels. Women with LBO demonstrated more periph-
eral aromatization and had higher production rates of estrone.
The women in their study had regular menstrual cycles.
The relationship between central obesity and hyperandro-
genemia may be bidirectional (336, 341, 342). In women,
androgens appear to encourage a male-type central obesity
(62, 113). Postmenopausal women, who have a decrease in
ovarian production of estrogen and an increase in androge-
n:estrogen ratio, have less gluteofemoral adiposity and more
upper body fat (53, 343). Additionally, postmenopausal
women given hormone therapy (estrogen) maintain a fat dis-
tribution similar to premenopausal women instead of the
android body type acquired by those not treated (344).
Female-to-male transsexuals given T had similar changes
(345). After 1 year, there was a signicant reduction in sub-
cutaneous fat but no change in visceral fat. They were then
ovariectomized and examined again by MRI at year 3, at
which time visceral fat had increased compared with baseline
values (345). Moreover, the effect was at least partly reversed
by utamide, an androgen receptor antagonist. Visceral fat
was not affected. Testosterone has been shown to reduce cat-
echolamine-induced lipolysis in subcutaneous fat cells from
human subjects by reducing HSL and b
2
-adrenoceptors (59).
Androgen-induced lipogenesis is also due to an effect on
LPL (59, 346). Evidence linking androgens to LPL activity
and lipid storage include a positive correlation between
LPL activity and free T in obese women in whom there is
a negative correlation with E
2
(60, 346, 347). When DHT,
a nonaromatizable androgen, was added to cultures of subcu-
taneous adipose tissue obtained from nondiabetic women
(59), LPL increased and HSL expression decreased. Addi-
tionally, ovariectomized mice given DHT demonstrated an
increase in visceral fat TG levels and, therefore, an increase
in visceral fat quantity (53). Proteins involved in lipogenesis
were elevated, including fatty acid synthase, sterol regulatory
element binding protein, and LPL. Enzymes involved in
lipolysis (AMP-activated protein kinase and acetyl-CoA
carboxylase) were down-regulated (53). Overall, in women,
androgens appear to promote central and suppress peripheral
fat accumulation, and estrogens have the opposite effect.
Central obesity, on the other hand, may contribute to ele-
vated serum androgen levels (17, 341, 348). Circulating an-
drogen levels increase with increasing obesity and decline
when obese hyperandrogenemic women lose weight
(349353). It is possible that insulin resistance and hyperin-
sulinemia, which commonly accompany abdominal obesity
and decline with weight loss, may be the causal link between
obesity and increased androgens, because insulin may serve
as a cogonadotropin to augment the production of T from
the ovaries (354). Insulin also decreases SHBG levels, which
increases the non-SHBG (bio-available) T (355357).
Abdominal obesity and hyperandrogenemia may also be
related to an increase in FFA ux (358). FFAs not only con-
tribute to insulin resistance, compensatory hyperinsulinemia,
and, consequently, elevated androgen levels but may also add
to the androgen pool by increasing the production of andro-
gen precursors fromthe adrenal glands (247, 359). Men given
a short-term 20% lipid infusion had an increase in FFA levels
and an increase in DHEA and A, before any detectable
change in insulin resistance. The evidence suggests that
FFA may affect adrenal androgen production directly and
not through a central (ACTH) mechanism (359).
As previously noted, UBO may also be considered to be
a hyperestrogenic state (340, 360, 361). Estrogenic effects
on fat distribution are partly due to actions on the sympathetic
nervous system and directly on proteins expressed in adipose
tissue. Both estrogen receptor a (ERa) and ERb are ex-
pressed in human subcutaneous adipocytes, but the most im-
portant physiologic effects appear to involve ERa (362364).
Estradiol decreases lipolysis by augmenting the number of
a2A-adrenergic receptors in subcutaneous but not visceral
fat, which may at least partly explain why reproductive-age
women have more body fat than men, because they have
a greater number of a2A-adrenergic receptors (362, 364).
Evidence for estrogen effects on fat deposition and LPL
activity is both complex and contradictory. Studies in this
area include quantifying LPL protein synthesis, activity,
and mRNA levels in vivo and in vitro, in rodents and in hu-
mans after the removal or administration of estrogen. Ovari-
ectomy in rats results in an increase in energy intake, TG
TABLE 5
Prevalence of obesity (BMI R30 kg/m
2
) in women aged 2039 years, % (SE).
Non-Hispanic white Non-Hispanic black Mexican-American
19992000 24.4 (2.6) 45.6 (3.9) 30.6 (5.7)
20012002 25.2 (2.3) 47.2 (3.8) 31.6 (5.3)
20032004 23.8 (3.1) 50.3 (4.6) 35.7 (3.6)
Note: BMI body mass index.
Bohler. Adipose tissue and reproduction in women. Fertil Steril 2010.
Fertility and Sterility

809
accumulation, LPL activity, and obesity (365). These effects
are diminished and almost completely reversed by physio-
logic estrogen replacement and a selective estrogen receptor
modulator with agonist properties (365). LPL activity in-
creases after ovariectomy in adipose tissue biopsies from
rats and is decreased dose-dependently after the administra-
tion of 17b-estradiol (366). Using genetically manipulated
3T3-L1 mouse adipocytes expressing ER, Homma et al.
(60) demonstrated a reduction in fat accumulation and LPL
mRNA levels after estrogen exposure.
In humans, the impact of estrogen on LPL and lipogenesis
appears to be age, region, and gender specic. Lipogenesis
and LPL activity in the gluteal-femoral area of women ap-
pears to follow an increase in estrogen, whereas there may
be an inverse relationship in the abdominal region. There
are several examples. Body fat distribution does not differ
by gender until puberty (367). Menstrual cycling women
have higher LPL activity in the gluteal-femoral region com-
pared with the abdominal region. LPL activity increases even
further during pregnancy but decreases during lactation
(368). Postmenopausal women tend to acquire fat centrally
and lose fat peripherally. Femoral adipocytes from postmen-
opausal women given estrogen-progesterone therapy showed
increased LPL activity (344, 369). Congenital estrogen de-
ciency due to Turner syndrome is accompanied by an in-
crease in visceral fat proportionate to the years of estrogen
deciency (370). Finally, male-to-female transsexuals treated
with estrogen accumulate more peripheral and less abdomi-
nal fat (371).
The results of other studies disagree with the above nd-
ings, however. Transdermal estrogen patches in the gluteal
region decreased LPL protein and activity but not mRNA
levels in adipose tissue biopsies beneath the patch compared
with surrounding areas (372). Toth et al. (373) found that
ovariectomized rats lost rather than gained fat mass, and
women of reproductive age given ethinyl-estradiol for 2
weeks had no change in LPL activity in adipocytes taken
from the buttocks (374). Finally, the effect of estrogen on
LPL activity and thereby TG hydrolysis may be dose related
in that subcutaneous abdominal adipocytes taken before elec-
tive surgery in women exposed to high-dose 17b-estradiol
demonstrated reduced LPL protein expression, whereas low
doses increased LPL expression (347).
ADIPOSITY AND POLYCYSTIC OVARY SYNDROME
The Rotterdam ESHRE/ASRMSponsored Consensus
Workshop Group dened PCOS by at least two of the follow-
ing: 1) oligo- or anovulation; 2) clinical and/or biochemical
signs of hyperandrogenism; and 3) polycystic ovaries; with
the exclusion of other etiologies that mimic PCOS (375).
Other common features include LH hypersecretion, insulin
resistance, and obesity (17, 376, 377). Although weight
gain and central obesity are common in PCOS patients and
often precede the onset of anovulatory cycles, obesity is
not one of the diagnostic criteria for PCOS (378).
The most recent criteria for PCOS actually date back to
Irving Stein and Michael Leventhal (379), who in 1935 in-
cluded amenorrhea, hirsutism, and polycystic ovaries in their
description of the syndrome. Most of their patients were
obese, and recent studies have determined that at least 50%
of PCOS patients are either overweight or obese (380,
381). Moreover, visceral adiposity is a common nding,
even when the subject is not classied as overweight (382).
When 30 nonobese Turkish patients with PCOS were com-
pared with 30 lean women with regular menstrual cycles
and similar BMI (mean 20 kg/m
2
), age, and WHR, the
PCOS group had signicantly more visceral fat but a similar
amount of upper body subcutaneous fat (270). Despite the
lack of obesity, they exhibited the classic features of PCOS,
suggesting that excess visceral fat is an important component
of the syndrome.
Truncal obesity is clearly associated with insulin resistance
(383, 384). Lord et al. (384) recently demonstrated that an
increase in visceral adipose tissue, in particular, was signi-
cantly related to insulin resistance in a prospective cross-sec-
tional analysis. An impressive linear correlation between
visceral fat and insulin resistance (r 0.68; P<.001) was
demonstrated (384).
The prevalence of insulin resistance, dened by a higher-
than-normal concentration of insulin to effect a normal re-
sponse (usually in glucose transport), may be as high as
80% in PCOS and is considered to be an important patho-
physiologic feature of the disorder (377, 385387). Insulin
resistance in PCOS appears to be selective; that is, whereas
skeletal muscle and adipose tissue may be refractory to insu-
lin action, other tissues, including ovarian stroma and liver,
respond or may be even hypersensitive to insulin (388,
389). This may be explained by the multiple intracellular sig-
naling pathways and pleiotropic effects of insulin, which in-
clude not only activating glucose transport but also mitogenic
and steroidogenic actions (390, 391). Insulin resistance leads
to a compensatory increase in insulin secretion, resulting in
hyperinsulinemia (81).
Impaired glucose tolerance (IGT) and type 2 diabetes mel-
litus among women in the U.S. are found in approximately
30% and 7.5%, respectively, of women with PCOS but in-
creases with age (392). Women with PCOS who have IGT
are more obese than PCOS patients with normal glucose tol-
erance (NGT) (393, 394). Moreover, obese women with
PCOS and NGT are hyperinsulinemic compared with thin
women with PCOS and NGT and have higher SHBG and
lower free T levels (394).
Weight loss in previously anovulatory obese women with
PCOS is associated with improvement in insulin sensitivity,
diminished insulin levels, a decrease in T, and increased ovu-
lation rates (349, 395397). Several studies have demon-
strated an increase in ovulatory cycles in PCOS women
when insulin sensitivity is increased by metformin or a thiazo-
lodinedione (15, 398, 399). Long-termtreatment with metfor-
min reduced menstrual abnormalities, abdominal (including
810
Bohler et al. Adipose tissue and reproduction in women Vol. 94, No. 3, August 2010
visceral) fat, and hirsutism when added to a hypocaloric diet
(397).
Fat cells of women with PCOS may have altered lipolysis
and lipogenesis even in the absence of obesity. There is a re-
sistance to catecholamine-induced lipolysis in subcutaneous
cells in vitro, and fasting glycerol levels (a product of lipoly-
sis) are lower in women with PCOS (400). Signaling path-
ways leading to the activation of HSL may be responsible
(400). This was conrmed in a recent study in which lean
women with PCOS or obesity (without PCOS) had signi-
cantly decreased catecholamine-induced lipolysis and HSL
protein expression compared with healthy control subjects
(185).
The link between obesity and androgens outlined in previ-
ously is probably operational in PCOS as well. A male-type
fat distribution dominates the phenotype of women with
PCOS (representing more lipogenesis than lipolysis), per-
haps because of excess androgens (401). Androgens have
the potential to augment fat deposition in subcutaneous and
visceral fat and may do so by stimulating LPL and inhibiting
HSL (402, 403). Testosterone and A production are aug-
mented from ovarian theca cells and stroma in response to
LH hypersecretion and higher-than-normal levels of insulin
(404406).
The adrenal glands also contribute to the total amount of
circulating androgens and perhaps to obesity. In as many as
40%of women with anovulatory PCOS, the adrenal gland ap-
pears to be overactive based on an increase in DHEAS levels
(404, 407, 408). Explanations for adrenal gland overactivity
include an increase in sensitivity to ACTH, dysregulation
of enzymes involved in cortisol metabolism, factors that reg-
ulate 17-hydroxylase/C17,20-lyase, and dyslipidemia with
an increase in FFAs (410412).
The enzymes 5a- and 5b-reductase are expressed in adi-
pose tissue as well as in liver and skin. They reduce cortisol
to 5a- and 5b-dihydrocortisol, and T to DHT (409, 410, 413).
Because the activity of 5a-reductase is enhanced in PCOS,
based on the high ratio of 5a to 5b urinary metabolites and
direct study of T metabolism in skin, there may be a decrease
in negative feedback by cortisol and a compensatory increase
in ACTHsecretion to normalize serumcortisol while increas-
ing androgen levels (409, 414). ACTH levels are normal in
PCOS, however, and explanations for normal ACTH levels
include a resetting of the hypothalamic-pituitary-axis and
an increase in pituitary sensitivity to ACTH (409, 413).
11b-Hydroxysteroid dehydrogenase (11b-HSD), which is
also important in cortisol clearance, is dysregulated in
PCOS (410). 11b-HSD exists as two different isoenzymes,
11b-HSD1 and 11b-HSD2, that catalyze the interconversion
of bioactive cortisol and inactive cortisone (415, 416). Corti-
sol is rapidly oxidized to cortisone by 11b-HSD2 in the kid-
ney, allowing mineralcorticoid receptors to be occupied and
regulated by aldosterone instead of the far more abundant
cortisol; if 11b-HSD2 activity is reduced, the excess cortisol
mimics aldosterone and hypertension and hypokalemia
develop (410). This occurs with inactivating mutations of
the 11-HSD2 gene and when activity is reduced by glycyr-
rhetinic acid in licorice (410).
11b-Hydroxysteroid dehydrogenase 1 converts cortisone
to cortisol (415, 417). The expression and activity of 11b-
HSD1 have emerged as a key factor in obesity, the metabolic
syndrome, and possibly PCOS (410, 418, 419). Overexpres-
sion of this enzyme in rats leads to central obesity and insulin
resistance (419). In human obesity,especially central obesity,
the activity of 11b-HSD1 is tissue specic (420, 421). It is
reduced in the liver but increased in adipocytes (422). De-
creased 11b-HSD1 activity in the liver, a major site of action,
may contribute to the compensatory increase in cortisol pro-
duction, and the enhanced generation of cortisol from
cortisone in adipocytes may be partly responsible for an in-
crease in adipocyte mass and central obesity, because of an
abundance of glucocorticoid receptors in the omentum
(410, 423426). Thus, changes in the production and meta-
bolic clearance of cortisol may play an important role in
PCOS (410, 413) (Figs. 5 and 6).
PCOS AND ADIPOKINES
Because of the interactions between leptin and insulin, leptin
may have a role in the pathophysiology of PCOS. Although
many studies have found similar leptin levels between
PCOS and weight-matched control subjects, others suggest
that leptin is increased (3, 139, 427429). Interestingly, leptin
mRNA levels in subcutaneous fat may be down-regulated in
obese PCOS patients (430).
Mantzoros et al. (139) found a strong correlation between
leptin and BMI in PCOS patients but no relationship with
gonadal hormones or hyperinsulinemia. Moreover, after con-
trolling for BMI, no signicant correlation was found
between serum leptin levels and HOMA or hyperinsulinemia
after glucose challenge in either obese or normal weight
women with PCOS (431). Furthermore, neither troglitazone
nor metformin, which increase insulin sensitivity, affected
serum leptin concentrations (432, 433).
Elevated leptin levels may contribute to the lack of follic-
ular maturation in PCOS (427, 429). Leptin inhibits the IGF-
1induced rise in FSH-stimulated estrogen production by
cultured rat granulosa cells, and the elevated follicular uid
leptin concentrations in PCOS might inhibit aromatase activ-
ity and follicular growth (148, 434). Although the concentra-
tion of leptin receptors was similar in granulosa cells from
women with PCOS and those undergoing IVF because of
tubal disease, the level of phosphorylated signal transducer
and activator of transcription was lower, suggesting that it is
leptin signaling that is disrupted in PCOS (429). Poor IVF
outcome has also been associated with elevated follicular
uid leptin levels (427, 429, 435).
Several studies have examined the potential role of adipo-
nectin in the ovulatory dysfunction and metabolic abnormal-
ities in PCOS (436439). Plasma adiponectin levels are
Fertility and Sterility

811
inversely related to BMI in subjects with PCOS as in subjects
without PCOS, and the degree of insulin resistance appeared
to have little affect on adiponectin levels (439). However,
other studies suggest that there may be at least some correla-
tion between adiponectin levels and insulin sensitivity in
PCOS, independent of adiposity (440, 441). Adiponectin
mRNA levels in adipose tissue have been found to be propor-
tionately reduced with increasing visceral and subcutaneous
fat measured by ultrasound (430).
Adiponectin gene polymorphisms have also been studied in
PCOS. Haap et al. (442) and Zhang et al. (443) found a higher
prevalence of the 45G and 276G/T polymorphism in the
adiponectin gene in women with PCOS compared with con-
trol women. This was not, however, associated with a more in-
sulin-resistant phenotype. On the other hand, Xita et al. (444)
and Escobar-Morreale et al. (438) did not nd any association
between common polymorphisms in the adiponectin gene and
PCOS. Xita et al. (444) reported that genomic variants might
inuence adiponectin levels in PCOS.
Interventions that affect reproductive and metabolic func-
tion in PCOS may also affect adiponectin levels. It has been
postulated that some of the effects of weight loss and insulin-
sensitizing agents in PCOS may be mediated through
changes in adiponectin levels. Ibanez and de Zegher (445)
evaluated the effects of an oral contraceptive or an antiandro-
geninsulin sensitizer combination on adipocytokines and in-
ammatory markers in adolescent and young adult women
with PCOS. They found that adiponectin levels decreased
with ethinylestradiol-drospirenone and increased with uta-
mide-metformin. Furthermore, lean body weight increased
and central adiposity decreased only in the latter group, sug-
gesting that oral contraceptives may not be the optimal treat-
ment for the metabolic abnormalities in PCOS patients.
Adiponectin levels also rose and insulin-stimulated glucose
disposal increased when PCOS patients were treated with
pioglitazone (446).
Several studies have found increased visfatin gene expres-
sion and plasma levels in women with PCOS compared with
BMI-matched control women (447449). In one study, 70
lean and obese women with PCOS and 45 healthy non-PCOS
controls were evaluated using an euglycemic clamp (449).
Both lean and obese PCOS subjects had lower insulin sensitiv-
ity as expected, but serumvisfatin levels were higher than con-
trol subjects only in lean PCOS subjects. Visfatin levels were,
however, negatively correlated with SHBG and positively cor-
related with the free androgen index in the entire PCOS group
(449). PCOS patients undergoing laparoscopy for infertility
also demonstrated an up-regulation of visfatin (447). Visfatin
mRNAand protein were increased in omental as well as subcu-
taneous fat and subcutaneous adipocytes compared with infer-
tile non-PCOS control subjects with unexplained infertility.
FIGURE 5
Cortisol metabolism may be tissue-specic in obesity and polycystic ovary syndrome. In these conditions,
hepatic 11b-HSD1 activity is reduced in the liver resulting in a decrease in the regeneration of cortisol; however,
adipocyte 11b-HSD1 activity is increased (Fig. 6). 5a-R activity is increased in the liver and skin resulting in an
increase in 5a-reduced cortisol metabolites and dihydrotestosterone production from testosterone (not shown).
Inreased production of cortisol in visceral fat because of increased 11bHSD activity may contribute to insulin
reistance and low SHBG. HSD hydroxysteroid dehydrogenase.
Bohler. Adipose tissue and reproduction in women. Fertil Steril 2010.
812
Bohler et al. Adipose tissue and reproduction in women Vol. 94, No. 3, August 2010
Tumor necrosis factor a levels in obese women (average
BMI 35 kg/m
2
) with and without PCOS were similar before
and after weight loss in one study, whereas another study
found higher levels (450, 451). In the latter study, the authors
concluded that the increase in TNF-a may have been due to
excess central fat (451). TNF-a secretion by cultured mono-
nuclear cells fromPCOS patients in response to glucose stim-
ulation was found to be reduced rather than increased as in
women without PCOS (452).
Cytokines have also been studied in follicular cells ob-
tained at the time of oocyte retrieval during IVF. These cells
express mRNA for a variety of cytokines, including IL-6, but
there was no difference between PCOS and non-PCOS pa-
tients (453). Reduced levels of IL-6 were found in those
FIGURE 6
Possible role of VAT and SAT in the pathophysiology of polycystic ovary syndrome. An increase in central obesity
and VAT may result fromelevated serumandrogens and local cortisol production, the latter caused by increased
11-HSD1 activity in adipocytes. 5a-R activity is also dysregulated and contributes to increased cortisol
clearance. The negative feedback of cortisol may be decreased resulting in a compensatory increase in ACTH in
an effort to maintain normal cortisol levels, while increasing adrenal androgen production. FFA in the liver and
skeletal muscle contribute to insulin resistance and hyperinsulinemia, as does altered adipokine levels. Insulin
acts as a co-gonadotropin, amplifying the effect of LH to induce androgen production from the ovary leading to
anovulation. Leptin may play a role in the increase in LHsecretion although other factors and obesity dampen this
effect. VAT visceral adipose tissue; SAT subcutaneous adipose tissue; FFA free fatty acid; HSD
hydroxysteroid dehydrogenase; E cortisone; F cortisol; R reductase; SHBG sex hormone binding
globulin; ACTH adrenocorticotropic hormone; LH luteinizing hormone; GnRH gonadotropin releasing
hormone;TNF tumor necrosis factor; IL interleukin; PAI plasminogen activator inhibitor.
Bohler. Adipose tissue and reproduction in women. Fertil Steril 2010.
Fertility and Sterility

813
patients with high BMI and those who achieved pregnancy,
however. The authors suggested that the angiogenic action
of IL-6 may increase follicular growth by increasing FSH ac-
cess (453). Amato et al. (454), on the other hand, found an in-
crease in serum and follicular uid levels of IL-6 in PCOS
patients undergoing IVF.
The possible increased risk of rst-trimester miscarriages
and gestational diabetes in women with PCOS has been re-
lated to increased PAI-1 activity and elevated plasma levels
of PAI-1 (456, 456). Glueck et al. (455) found more PAI-1
gene 4G polymorphisms and an increase in PAI-1 activity
in women with PCOS compared with control women, a nd-
ing that was also demonstrated in a recent meta-analysis
(457). PAI-1 gene polymorphisms may be important in the
pathogenesis of PCOS, as suggested by the enlarged cystic
ovaries and slightly elevated T levels seen in transgenic
mice overexpressing PAI-1 (458). Other comorbid ndings
associated with an up-regulation of PAI-1 activity and
PCOS include obesity, insulin resistance, and hypertriglycer-
idemia (455). However, amplied PAI-1 plasma levels and
activity are associated with PCOS independent of BMI, and
there is also a positive correlation between PAI-1 activity
and serum T levels (222, 458).
Plasminogen activator inhibitor 1 protein activity as well
as insulin levels decrease in women with PCOS when they
are treated with troglitazone (222). Metformin begun in the
rst trimester and continued throughout pregnancy not only
reduced insulin and PAI-1 levels but also decreased spontane-
ous abortions and other pregnancy complications (165, 455).
Polycystic ovary syndrome has been associated with low-
grade inammation, but whether it is due to PCOS per se or to
UBO is unsettled (451, 459461). Kelly et al. (459) were the
rst to examine low-grade chronic inammatory markers in
PCOS. The PCOS group had signicantly elevated CRP
levels compared with healthy control subjects matched for
BMI which were inversely correlated with insulin sensitivity,
although central adiposity was not evaluated. Other investi-
gators found that the elevated inammatory markers in
PCOS were correlated with BMI and upper abdominal
obesity rather than with the characteristics of PCOS, such
as hyperandrogenemia (451, 461). Evidence of inammation
is usually accompanied by insulin resistance and hyperinsu-
linemia, which together are thought to increase the risk for
hypertension, type 2 diabetes, and heart and vascular disease.
Women with PCOS with elevated hs-CRP levels and insulin
resistance have evidence of endothelial dysfunction, even if
BMI is normal (23 kg/m
2
) (462). Moreover, even in the ab-
sence of inammatory markers, obese young (average age 34
years) insulin-resistant PCOS patients have more evidence of
early coronary atherosclerosis than weight-matched control
subjects (463).
Thus, there are a series of cytokine abnormalities in
women with PCOS, some of which are due to obesity but
others that could be disease specic. Cytokines may play
an important role not only in the fertility disturbance but
also in the many other reproductive problems that PCOS
women experience (Fig. 6).
CONCLUSIONS
Adipose tissue and its chief component, adipocytes, are nec-
essary for survival. They protect the body from trauma, pro-
vide the major source of stored energy, and inuence
a myriad of physiologic functions, including reproduction,
through an array of secreted products collectively known as
adipokines. Deciency or excess of these products disturbs
the dynamics of the reproductive axis and contributes to
a suboptimal environment for pregnancy, if it should occur.
Although the total amount of body fat is important, it is the
selective expression of adipokines in various adipose tissue
compartments that makes the distribution of adipose tissue
critical.
Both thinness and obesity have a negative impact on the re-
productive system. An excess of VAT, in particular, appears to
be the most important contributor to the development of
anovulatory cycles in women with PCOS. To that end it, is
vitally important to understand how obesity and adipokines
predispose to infertility and pregnancy loss. Although pre-
ferred treatments remain controversial, our increasing aware-
ness of the mechanisms linking adipose tissue to reproduction
has dramatically altered the treatment approaches for repro-
ductive endocrinologists.
REFERENCES
1. Kershaw EE, Flier JS. Adipose tissue as an endocrine organ. J Clin
Endocrinol Metab 2004;89:254856.
2. Fischer-Posovszky P, Wabitsch M, Hochberg Z. Endocrinology of
adipose tissuean update. Horm Metab Res 2007;39:31421.
3. Budak E, Fernandez Sanchez M, Bellver J, Cervero A, Simon C,
Pellicer A. Interactions of the hormones leptin, ghrelin, adiponectin,
resistin, and PYY
336
with the reproductive system. Fertil Steril
2006;85:156381.
4. Fietta P. Focus on leptin, a pleiotropic hormone. Minerva Med
2005;96:6575.
5. Kelesidis T, Mantzoros CS. The emerging role of leptin in humans.
Pediatr Endocrinol Rev 2006;3:23948.
6. Badman MK, Flier JS. The adipocyte as an active participant in energy
balance and metabolism. Gastroenterology 2007;132:210315.
7. Zhang Y, Proenca R, Maffei M, Barone M, Leopold L, Friedman JM.
Positional cloning of the mouse obese gene and its human homologue.
Nature 1994;372:42532.
8. Hu E, Liang P, Spiegelman BM. AdipoQ is a novel adipose-specic
gene dysregulated in obesity. J Biol Chem 1996;271:10697703.
9. Fain JN, Madan AK, Hiler ML, Cheema P, Bahouth SW. Comparison
of the release of adipokines by adipose tissue, adipose tissue matrix,
and adipocytes from visceral and subcutaneous abdominal adipose
tissues of obese humans. Endocrinology 2004;145:227382.
10. Haque WA, Garg A. Adipocyte biology and adipocytokines. Clin Lab
Med 2004;24:21734.
11. Trayhurn P, Bing C, Wood IS. Adipose tissue and adipokinesenergy
regulation from the human perspective. J Nutr 2006;136:1935S9S.
12. Rosen ED, Spiegelman BM. Adipocytes as regulators of energy balance
and glucose homeostasis. Nature 2006;444:84753.
13. Goodarzi MO, Azziz R. Diagnosis, epidemiology, and genetics of the
polycystic ovary syndrome. Best Pract Res Clin Endocrinol Metab
2006;20:193205.
814
Bohler et al. Adipose tissue and reproduction in women Vol. 94, No. 3, August 2010
14. Azziz R, Woods KS, Reyna R, Key TJ, Knochenhauer ES, Yildiz BO.
The prevalence and features of the polycystic ovary syndrome in an un-
selected population. J Clin Endocrinol Metab 2004;89:27459.
15. Legro RS, Barnhart HX, Schlaff WD, Carr BR, Diamond MP,
Carson SA, et al. Clomiphene, metformin, or both for infertility in
the polycystic ovary syndrome. N Engl J Med 2007;356:55166.
16. Ehrmann DA, Liljenquist DR, Kasza K, Azziz R, Legro RS, Ghazzi MN.
Prevalence and predictors of the metabolic syndrome in women with
polycystic ovary syndrome. J Clin Endocrinol Metab 2006;91:4853.
17. Barber TM, McCarthy MI, Wass JA, Franks S. Obesity and polycystic
ovary syndrome. Clin Endocrinol (Oxf) 2006;65:13745.
18. Hoeger KM. Obesity and lifestyle management in polycystic ovary syn-
drome. Clin Obstet Gynecol 2007;50:27794.
19. Ogden CL, Carroll MD, Curtin LR, McDowell MA, Tabak CJ,
Flegal KM. Prevalence of overweight and obesity in the United States,
19992004. JAMA 2006;295:154955.
20. Vague J. [Clinical aspects of obesity]. La Vie Medicale 1950;31:915.
French.
21. Krotkiewski M, Bjorntorp P, SjostromL, Smith U. Impact of obesity on
metabolism in men and women. Importance of regional adipose tissue
distribution. J Clin Invest 1983;72:115062.
22. Matsuzawa Y, Shimomura I, Nakamura T, Keno Y, Kotani K,
Tokunaga K. Pathophysiology and pathogenesis of visceral fat obesity.
Obes Res 1995;3(Suppl 2):187S94S.
23. Despres JP, Lemieux I. Abdominal obesity and metabolic syndrome.
Nature 2006;444:8817.
24. Zaadstra BM, Seidell JC, Van Noord PA, te Velde ER, Habbema JD,
Vrieswijk B, et al. Fat and female fecundity: prospective study of effect
of body fat distribution on conception rates. Bmj 1993;306:4847.
25. Hall LF, Neubert AG. Obesity and pregnancy. Obstet Gynecol Surv
2005;60:25360.
26. Nelson SM, Fleming RF. The preconceptual contraception paradigm:
obesity and infertility. Hum Reprod 2007;22:9125.
27. Trujillo ME, Scherer PE. Adipose tissuederived factors: impact on
health and disease. Endocr Rev 2006;27:76278.
28. Albright AL, Stern JS. Adipose tissue. In: Fahey TD, ed. Encyclopedia of
sports medicine and science. Internet Society for Sport Science, May 30,
1998. Available at: http://www.sportsci.org/encyc/adipose/adipose.html
Last accessed June 4, 2008.
29. de Ferranti S, Mozaffarian D. The perfect storm: obesity, adipocyte dys-
function, and metabolic consequences. Clin Chem 2008;54:94555.
30. Trayhurn P. The biology of obesity. Proc Nutr Soc 2005;64:318.
31. Taleb S, Van Haaften R, Henegar C, Hukshorn C, Cancello R,
Pelloux V, et al. Microarray proling of human white adipose tissue
after exogenous leptin injection. Eur J Clin Invest 2006;36:15363.
32. Gallagher D, Kelley DE, Yim JE, Spence N, Albu J, Boxt L, et al. Ad-
ipose tissue distribution is different in type 2 diabetes. Am J Clin Nutr
2009.
33. Trayhurn P. Endocrine and signalling role of adipose tissue: new per-
spectives on fat. Acta Physiol Scand 2005;184:28593.
34. Frayn KN, Karpe F, Fielding BA, Macdonald IA, Coppack SW. Integra-
tive physiology of human adipose tissue. Int J Obes Relat Metab Disord
2003;27:87588.
35. Bartness TJ, Song CK. Thematic review series: adipocyte biology.
Sympathetic and sensory innervation of white adipose tissue. J Lipid
Res 2007;48:165572.
36. Arner P. Human fat cell lipolysis: biochemistry, regulation and clinical
role. Best Pract Res Clin Endocrinol Metab 2005;19:47182.
37. Song CK, Jackson RM, Harris RB, Richard D, Bartness TJ. Melanocor-
tin-4 receptor mRNA is expressed in sympathetic nervous system out-
ow neurons to white adipose tissue. Am J Physiol Regul Integr Comp
Physiol 2005;289:R146776.
38. Brito MN, Brito NA, Baro DJ, Song CK, Bartness TJ. Differential ac-
tivation of the sympathetic innervation of adipose tissues by melanocor-
tin receptor stimulation. Endocrinology 2007;148:533947.
39. Ellacott KL, Cone RD. The central melanocortin system and the
integration of short- and long-term regulators of energy homeostasis.
Recent Prog Horm Res 2004;59:395408.
40. Bowers RR, Festuccia WT, Song CK, Shi H, Migliorini RH,
Bartness TJ. Sympathetic innervation of white adipose tissue and its
regulation of fat cell number. Am J Physiol Regul Integr Comp Physiol
2004;286:R116775.
41. Foster MT, Bartness TJ. Sympathetic but not sensory denervation stim-
ulates white adipocyte proliferation. Am J Physiol Regul Integr Comp
Physiol 2006;291:R16307.
42. Cousin B, Casteilla L, Lafontan M, Ambid L, Langin D, Berthault MF,
et al. Local sympathetic denervation of white adipose tissue in rats in-
duces preadipocyte proliferation without noticeable changes in metab-
olism. Endocrinology 1993;133:225562.
43. Clement K, Vaisse C, Manning BS, Basdevant A, Guy-Grand B, Ruiz J,
et al. Genetic variation in the beta 3-adrenergic receptor and an
increased capacity to gain weight in patients with morbid obesity. N
Engl J Med 1995;333:3524.
44. Dodt C, Lonnroth P, Wellhoner JP, Fehm HL, Elam M. Sympathetic
control of white adipose tissue in lean and obese humans. Acta Physiol
Scand 2003;177:3517.
45. Rossner S, Taylor CL, Byington RP, Furberg CD. Long term proprano-
lol treatment and changes in body weight after myocardial infarction.
BMJ 1990;300:9023.
46. Giordano A, Song CK, Bowers RR, Ehlen JC, Frontini A, Cinti S, et al.
White adipose tissue lacks signicant vagal innervation and immuno-
histochemical evidence of parasympathetic innervation. Am J Physiol
Regul Integr Comp Physiol 2006;291:R124355.
47. Berthoud HR, Fox EA, Neuhuber WL. Vagaries of adipose tissue inner-
vation. Am J Physiol Regul Integr Comp Physiol 2006;291:R12402.
48. Kreier F, Buijs RM. Evidence for parasympathetic innervation of white
adipose tissue, clearing up some vagaries. Am J Physiol Regul Integr
Comp Physiol 2007;293:R5489; author reply R502, discussion
R534.
49. Guilherme A, Virbasius JV, Puri V, Czech MP. Adipocyte dysfunctions
linking obesity to insulin resistance and type 2 diabetes. Nat Rev Mol
Cell Biol 2008;9:36777.
50. Bjorntorp P. Hormonal control of regional fat distribution. HumReprod
1997;12(Suppl 1):215.
51. Jaworski K, Sarkadi-Nagy E, Duncan RE, Ahmadian M, Sul HS. Reg-
ulation of triglyceride metabolism. IV. Hormonal regulation of lipolysis
in adipose tissue. Am J Physiol Gastrointest Liver Physiol 2007;293:
G14.
52. Wajchenberg BL. Subcutaneous and visceral adipose tissue: their rela-
tion to the metabolic syndrome. Endocr Rev 2000;21:697738.
53. McInnes KJ, Corbould A, Simpson ER, Jones ME. Regulation of aden-
osine 5
0
, monophosphateactivated protein kinase and lipogenesis by
androgens contributes to visceral obesity in an estrogen-decient state.
Endocrinology 2006;147:590713.
54. Lafontan M. Advances in adipose tissue metabolism. Int J Obes (Lond)
2008;32(Suppl 7):S3951.
55. Williams RH, Larsen PR. Williams textbook of endocrinology. 10th ed.
Philadelphia: Saunders, 2003.
56. Arner P, Langin D. The role of neutral lipases in human adipose tissue
lipolysis. Curr Opin Lipidol 2007;18:24650.
57. Frayn KN. Adipose tissue as a buffer for daily lipid ux. Diabetologia
2002;45:120110.
58. Wang S, Soni KG, Semache M, Casavant S, Fortier M, Pan L, et al.
Lipolysis and the integrated physiology of lipid energy metabolism.
Mol Genet Metab 2008;95:11726.
59. Anderson LA, McTernan PG, Harte AL, Barnett AH, Kumar S. The reg-
ulation of HSL and LPL expression by DHT and utamide in human
subcutaneous adipose tissue. Diabetes Obes Metab 2002;4:20913.
60. Homma H, Kurachi H, Nishio Y, Takeda T, Yamamoto T, Adachi K,
et al. Estrogen suppresses transcription of lipoprotein lipase gene. Ex-
istence of a unique estrogen response element on the lipoprotein lipase
promoter. J Biol Chem 2000;275:1140411.
61. Mayes JS, Watson GH. Direct effects of sex steroid hormones on
adipose tissues and obesity. Obes Rev 2004;5:197216.
62. Bjorntorp P. The regulation of adipose tissue distribution in humans. Int
J Obes Relat Metab Disord 1996;20:291302.
Fertility and Sterility

815
63. Moller DE. Potential role of TNF-alpha in the pathogenesis of insulin re-
sistance and type 2 diabetes. Trends Endocrinol Metab 2000;11:2127.
64. Hotamisligil GS. Inammation and metabolic disorders. Nature
2006;444:8607.
65. Klaus S. Adipose tissue as a regulator of energy balance. Curr Drug
Targets 2004;5:24150.
66. Trayhurn P. Adipocyte biology. Obes Rev 2007;8(Suppl 1):414.
67. Human energy requirements. Report of a joint FAO/WHO/UNU Expert
Consultation, Rome, October 17-24, 2001. FAO Food and Nutrition
Technical Report Series. Rome: United Nations University, World
Health Organization, Food and Agriculture Organization of the United
Nations, 2004.
68. Frisch RE, McArthur JW. Menstrual cycles: fatness as a determinant of
minimum weight for height necessary for their maintenance or onset.
Science 1974;185:94951.
69. Grill HJ, Kaplan JM. The neuroanatomical axis for control of energy
balance. Front Neuroendocrinol 2002;23:240.
70. Zigman JM, Elmquist JK. Minireview: from anorexia to obesitythe
yin and yang of body weight control. Endocrinology 2003;144:
374956.
71. Morton GJ, Cummings DE, Baskin DG, Barsh GS, Schwartz MW. Cen-
tral nervous system control of food intake and body weight. Nature
2006;443:28995.
72. Spiegelman BM, Flier JS. Obesity and the regulation of energy balance.
Cell 2001;104:53143.
73. Friedman JM, Halaas JL. Leptin and the regulation of body weight in
mammals. Nature 1998;395:76370.
74. Morrison CD, Morton GJ, Niswender KD, Gelling RW, Schwartz MW.
Leptin inhibits hypothalamic Npy and Agrp gene expressionvia a mech-
anism that requires phosphatidylinositol 3-OH-kinase signaling. Am J
Physiol Endocrinol Metab 2005;289:E10517.
75. Coll AP, Farooqi IS, ORahilly S. The hormonal control of food intake.
Cell 2007;129:25162.
76. Morton GJ, Blevins JE, Williams DL, Niswender KD, Gelling RW,
Rhodes CJ, et al. Leptin action in the forebrain regulates the hindbrain
response to satiety signals. J Clin Invest 2005;115:70310.
77. Schwartz MW, Baskin DG, Bukowski TR, Kuijper JL, Foster D,
Lasser G, et al. Specicity of leptin action on elevated blood glucose
levels and hypothalamic neuropeptide Y gene expression in ob/ob
mice. Diabetes 1996;45:5315.
78. Di Marzo V, Goparaju SK, Wang L, Liu J, Batkai S, Jarai Z, et al.
Leptin-regulated endocannabinoids are involved in maintaining food
intake. Nature 2001;410:8225.
79. Kristensen P, Judge ME, Thim L, Ribel U, Christjansen KN, Wulff BS,
et al. Hypothalamic CART is a new anorectic peptide regulated by lep-
tin. Nature 1998;393:726.
80. Abel ED, Peroni O, Kim JK, Kim YB, Boss O, Hadro E, et al. Adipose-
selective targeting of the GLUT4 gene impairs insulin action in muscle
and liver. Nature 2001;409:72933.
81. Kahn BB. Lilly lecture 1995. Glucose transport: pivotal step in insulin
action. Diabetes 1996;45:164454.
82. Kaser S, Kaser A, Sandhofer A, Ebenbichler CF, Tilg H, Patsch JR. Re-
sistin messenger-RNA expression is increased by proinammatory cy-
tokines in vitro. Biochem Biophys Res Commun 2003;309:28690.
83. Pietilainen KH, Kannisto K, Korsheninnikova E, Rissanen A, Kaprio J,
Ehrenborg E, et al. Acquired obesity increases CD68 and tumor necro-
sis factor-alpha and decreases adiponectin gene expression in adipose
tissue: a study in monozygotic twins. J Clin Endocrinol Metab
2006;91:277681.
84. Nien JK, Mazaki-Tovi S, Romero R, Kusanovic JP, Erez O, Gotsch F,
et al. Resistin: a hormone which induces insulin resistance is increased
in normal pregnancy. J Perinat Med 2007;35:51321.
85. Nagaev I, Smith U. Insulin resistance and type 2 diabetes are not related
to resistin expression in human fat cells or skeletal muscle. Biochem
Biophys Res Commun 2001;285:5614.
86. Nien JK, Mazaki-Tovi S, Romero R, Erez O, Kusanovic JP, Gotsch F,
et al. Plasma adiponectin concentrations in nonpregnant, normal and
overweight pregnant women. J Perinat Med 2007;35:52231.
87. Brannstrom M, Friden BE, Jasper M, Norman RJ. Variations in periph-
eral blood levels of immunoreactive tumor necrosis factor alpha
(TNFalpha) throughout the menstrual cycle and secretion of TNFalpha
from the human corpus luteum. Eur J Obstet Gynecol Reprod Biol
1999;83:2137.
88. Fukuhara A, Matsuda M, Nishizawa M, Segawa K, Tanaka M,
Kishimoto K, et al. Visfatin: a protein secreted by visceral fat that
mimics the effects of insulin. Science 2005;307:42630.
89. Samal B, Sun Y, Stearns G, Xie C, Suggs S, McNiece I. Cloning and
characterization of the cDNA encoding a novel human preB-cell col-
onyenhancing factor. Mol Cell Biol 1994;14:14317.
90. Steppan CM, Lazar MA. Resistin and obesity-associated insulin resis-
tance. Trends Endocrinol Metab 2002;13:1823.
91. Silha JV, Krsek M, Skrha JV, Sucharda P, Nyomba BL, Murphy LJ.
Plasma resistin, adiponectin and leptin levels in lean and obese subjects:
correlations with insulin resistance. Eur J Endocrinol 2003;149:3315.
92. Fain JN, Buehrer B, Bahouth SW, Tichansky DS, Madan AK. Compar-
ison of messenger RNA distribution for 60 proteins in fat cells vs the
nonfat cells of human omental adipose tissue. Metabolism 2008;57:
100515.
93. Guzik TJ, Mangalat D, Korbut R. Adipocytokinesnovel link between
inammation and vascular function? J Physiol Pharmacol 2006;57:
50528.
94. Campos DB. The benecial adipokines in reproduction and fertility.
Int J Obes (Lond) 2008;32:22331.
95. Halberg N, Wernstedt-AsterholmI, Scherer PE. The adipocyte as an en-
docrine cell. Endocrinol Metab Clin North Am 2008;37:75368.
96. Smith-Kirwin SM, OConnor DM, De Johnston J, Lancey ED,
Hassink SG, Funanage VL. Leptin expression in human mammary ep-
ithelial cells and breast milk. J Clin Endocrinol Metab 1998;83:18103.
97. Masuzaki H, Ogawa Y, Sagawa N, Hosoda K, Matsumoto T, Mise H,
et al. Nonadipose tissue production of leptin: leptin as a novel pla-
centa-derived hormone in humans. Nat Med 1997;3:102933.
98. Bado A, Levasseur S, Attoub S, Kermorgant S, Laigneau JP,
Bortoluzzi MN, et al. The stomach is a source of leptin. Nature
1998;394:7903.
99. Mantzoros CS. The role of leptin in human obesity and disease: a review
of current evidence. Ann Intern Med 1999;130:67180.
100. Sahu A. Minireview: a hypothalamic role in energy balance with special
emphasis on leptin. Endocrinology 2004;145:261320.
101. Chan JL, Mantzoros CS. Role of leptin in energy-deprivation states:
normal human physiology and clinical implications for hypothalamic
amenorrhoea and anorexia nervosa. Lancet 2005;366:7485.
102. Clement K, Vaisse C, Lahlou N, Cabrol S, Pelloux V, Cassuto D, et al. A
mutation in the human leptin receptor gene causes obesity and pituitary
dysfunction. Nature 1998;392:398401.
103. Montague CT, Farooqi IS, Whitehead JP, Soos MA, Rau H,
Wareham NJ, et al. Congenital leptin deciency is associated with
severe early-onset obesity in humans. Nature 1997;387:9038.
104. Sinha MK, Opentanova I, Ohannesian JP, Kolaczynski JW,
Heiman ML, Hale J, et al. Evidence of free and bound leptin in human
circulation. Studies in lean and obese subjects and during short-term
fasting. J Clin Invest 1996;98:127782.
105. Tartaglia LA. The leptin receptor. J Biol Chem 1997;272:60936.
106. Lee MJ, Wang Y, Ricci MR, Sullivan S, Russell CD, Fried SK. Acute
and chronic regulation of leptin synthesis, storage, and secretion by in-
sulin and dexamethasone in human adipose tissue. Am J Physiol Endo-
crinol Metab 2007;292:E85864.
107. Ahima RS. Body fat, leptin, and hypothalamic amenorrhea. N Engl J
Med 2004;351:95962.
108. Sahu A. Leptin signaling in the hypothalamus: emphasis on energy ho-
meostasis and leptin resistance. Front Neuroendocrinol 2003;24:
22553.
109. Tartaglia LA, Dembski M, Weng X, Deng N, Culpepper J, Devos R,
et al. Identication and expression cloning of a leptin receptor, OB-R.
Cell 1995;83:126371.
110. Papathanassoglou E, El-Haschimi K, Li XC, Matarese G, Strom T,
Mantzoros C. Leptin receptor expression and signaling in lymphocytes:
816
Bohler et al. Adipose tissue and reproduction in women Vol. 94, No. 3, August 2010
kinetics during lymphocyte activation, role in lymphocyte survival, and
response to high fat diet in mice. J Immunol 2006;176:774552.
111. Seufert J, Kieffer TJ, Leech CA, Holz GG, Moritz W, Ricordi C, et al.
Leptin suppression of insulin secretion and gene expression in human
pancreatic islets: implications for the development of adipogenic diabe-
tes mellitus. J Clin Endocrinol Metab 1999;84:6706.
112. Sierra-Honigmann MR, Nath AK, Murakami C, Garcia-Cardena G,
Papapetropoulos A, Sessa WC, et al. Biological action of leptin as an
angiogenic factor. Science 1998;281:16836.
113. Chang PL, Lindheim SR, Lowre C, Ferin M, Gonzalez F, Berglund L,
et al. Normal ovulatory women with polycystic ovaries have hyperan-
drogenic pituitary-ovarian responses to gonadotropin-releasing hor-
mone-agonist testing. J Clin Endocrinol Metab 2000;85:9951000.
114. Ciof JA, Van Blerkom J, Antczak M, Shafer A, Wittmer S,
Snodgrass HR. The expression of leptin and its receptors in pre-ovula-
tory human follicles. Mol Hum Reprod 1997;3:46772.
115. Popovic V, Damjanovic S, Dieguez C, Casanueva FF. Leptin and the
pituitary. Pituitary 2001;4:714.
116. Spiegelman BM, Flier JS. Adipogenesis and obesity: rounding out the
big picture. Cell 1996;87:37789.
117. Licinio J, Negrao AB, Mantzoros C, Kaklamani V, Wong ML,
Bongiorno PB, et al. Synchronicity of frequently sampled, 24-h concen-
trations of circulating leptin, luteinizing hormone, and estradiol in
healthy women. Proc Natl Acad Sci U S A 1998;95:25416.
118. Laughlin GA, Yen SS. Hypoleptinemia in women athletes: absence of
a diurnal rhythm with amenorrhea. J Clin Endocrinol Metab 1997;82:
31821.
119. Iuorno MJ, Islam LZ, Veldhuis PP, Boyd DG, Farhy LS, Johnson ML,
et al. Leptin secretory burst mass correlates with body mass index and
insulin in normal women but not in women with polycystic ovary syn-
drome. Metabolism 2007;56:15615.
120. Ahima RS, Prabakaran D, Mantzoros C, Qu D, Lowell B, Maratos-
Flier E, et al. Role of leptin in the neuroendocrine response to fasting.
Nature 1996;382:2502.
121. Munzberg H, Myers MG Jr. Molecular and anatomical determinants of
central leptin resistance. Nat Neurosci 2005;8:56670.
122. Hamilton BS, Paglia D, Kwan AY, Deitel M. Increased obese mRNA
expression in omental fat cells from massively obese humans. Nat
Med 1995;1:9536.
123. Elbers JM, Asscheman H, Seidell JC, Frolich M, Meinders AE,
Gooren LJ. Reversal of the sex difference in serum leptin levels upon
cross-sex hormone administration in transsexuals. J Clin Endocrinol
Metab 1997;82:326770.
124. Shimizu H, Shimomura Y, Nakanishi Y, Futawatari T, Ohtani K, Sato N,
et al. Estrogen increases in vivo leptin production in rats and human
subjects. J Endocrinol 1997;154:28592.
125. Van Harmelen V, Reynisdottir S, Eriksson P, Thorne A, Hoffstedt J,
Lonnqvist F, et al. Leptin secretion from subcutaneous and visceral ad-
ipose tissue in women. Diabetes 1998;47:9137.
126. Casabiell X, Pineiro V, Vega F, De La Cruz LF, Dieguez C,
Casanueva FF. Leptin, reproduction and sex steroids. Pituitary
2001;4:939.
127. Schwartz MW, Peskind E, Raskind M, Boyko EJ, Porte D Jr. Cerebro-
spinal uid leptin levels: relationship to plasma levels and to adiposity
in humans. Nat Med 1996;2:58993.
128. Wabitsch M, Blum WF, Muche R, Braun M, Hube F, Rascher W, et al.
Contribution of androgens to the gender difference in leptin production
in obese children and adolescents. J Clin Invest 1997;100:80813.
129. Seufert J, Kieffer TJ, Habener JF. Leptin inhibits insulin gene transcrip-
tion and reverses hyperinsulinemia in leptin-decient ob/ob mice. Proc
Natl Acad Sci U S A 1999;96:6749.
130. Kieffer TJ, Heller RS, Habener JF. Leptin receptors expressed on pan-
creatic beta-cells. Biochem Biophys Res Commun 1996;224:5227.
131. Saladin R, De Vos P, Guerre-Millo M, Leturque A, Girard J, Staels B,
et al. Transient increase in obese gene expression after food intake or
insulin administration. Nature 1995;377:5279.
132. Kolaczynski JW, Considine RV, Ohannesian J, Marco C, Opentanova I,
Nyce MR, et al. Responses of leptin to short-term fasting and refeeding
in humans: a link with ketogenesis but not ketones themselves. Diabetes
1996;45:15115.
133. Boden G, Chen X, Kolaczynski JW, Polansky M. Effects of prolonged
hyperinsulinemia on serum leptin in normal human subjects. J Clin
Invest 1997;100:110713.
134. Kieffer TJ, Habener JF. The adipoinsular axis: effects of leptin on pan-
creatic beta-cells. Am J Physiol Endocrinol Metab 2000;278:E114.
135. Ahmed ML, Ong KK, Morrell DJ, Cox L, Drayer N, Perry L, et al. Lon-
gitudinal study of leptin concentrations during puberty: sex differences
and relationship to changes in body composition. J Clin Endocrinol
Metab 1999;84:899905.
136. Clayton PE, Gill MS, Hall CM, Tillmann V, Whatmore AJ, Price DA.
Serum leptin through childhood and adolescence. Clin Endocrinol
(Oxf) 1997;46:72733.
137. Suter KJ, Pohl CR, Wilson ME. Circulating concentrations of nocturnal
leptin, growth hormone, and insulin-like growth factor-I increase before
the onset of puberty in agonadal male monkeys: potential signals for the
initiation of puberty. J Clin Endocrinol Metab 2000;85:80814.
138. Shalitin S, Phillip M. Role of obesity and leptin in the pubertal process
and pubertal growtha review. Int J Obes Relat Metab Disord 2003;27:
86974.
139. Mantzoros CS, Flier JS, Rogol AD. A longitudinal assessment of hor-
monal and physical alterations during normal puberty in boys. V. Rising
leptin levels may signal the onset of puberty. J Clin Endocrinol Metab
1997;82:106670.
140. Grumbach MM. The neuroendocrinology of human puberty revisited.
Horm Res 2002;57(Suppl 2):214.
141. Blum WF, Englaro P, Hanitsch S, Juul A, Hertel NT, Muller J, et al.
Plasma leptin levels in healthy children and adolescents: dependence
on body mass index, body fat mass, gender, pubertal stage, and testos-
terone. J Clin Endocrinol Metab 1997;82:290410.
142. Geisthovel F, Jochmann N, Widjaja A, Horn R, Brabant G. Serum pat-
tern of circulating free leptin, bound leptin, and soluble leptin receptor
in the physiological menstrual cycle. Fertil Steril 2004;81:398402.
143. Cervero A, Horcajadas JA, Mart J, Pellicer A, Simon C. The leptin sys-
tem during human endometrial receptivity and preimplantation devel-
opment. J Clin Endocrinol Metab 2004;89:244251.
144. Quinton ND, Laird SM, Okon MA, Li TC, Smith RF, Ross RJ, et al. Se-
rum leptin levels during the menstrual cycle of healthy fertile women.
Br J Biomed Sci 1999;56:169.
145. Goumenou AG, Matalliotakis IM, Koumantakis GE, Panidis DK. The
role of leptin in fertility. Eur J Obstet Gynecol Reprod Biol
2003;106:11824.
146. Agarwal SK, Vogel K, Weitsman SR, Magofn DA. Leptin antagonizes
the insulin-like growth factor-I augmentation of steroidogenesis in
granulosa and theca cells of the human ovary. J Clin Endocrinol Metab
1999;84:10726.
147. Caprio M, Fabbrini E, Isidori AM, Aversa A, Fabbri A. Leptin in repro-
duction. Trends Endocrinol Metab 2001;12:6572.
148. Huang HF, Wang B, Yang XF, Luo Q, Sheng JZ. Nitric oxide mediates
inhibitory effect of leptin on insulin-like growth factor I augmentation
of 17beta-estradiol production in human granulosa cells. Biol Reprod
2005;72:1026.
149. Spicer LJ, Chamberlain CS, Francisco CC. Ovarian action of leptin: ef-
fects on insulin-like growth factor-I-stimulated function of granulosa
and thecal cells. Endocrine 2000;12:539.
150. Schubring C, Prohaska F, Prohaska A, Englaro P, Blum W, Siebler T,
et al. Leptin concentrations in maternal serum and amniotic uid
during the second trimenon: differential relation to fetal gender and
maternal morphometry. Eur J Obstet Gynecol Reprod Biol 1999;86:
1517.
151. Scherer PE, Williams S, Fogliano M, Baldini G, Lodish HF. Anovel se-
rum protein similar to C1q, produced exclusively in adipocytes. J Biol
Chem 1995;270:267469.
152. Maeda K, Okubo K, Shimomura I, Funahashi T, Matsuzawa Y,
Matsubara K. cDNA cloning and expression of a novel adipose specic
collagen-like factor, apM1 (adipose most abundant gene transcript 1).
Biochem Biophys Res Commun 1996;221:2869.
Fertility and Sterility

817
153. Nakano Y, Tobe T, Choi-Miura NH, Mazda T, Tomita M. Isolation and
characterization of GBP28, a novel gelatin-binding protein puried
from human plasma. J Biochem (Tokyo) 1996;120:80312.
154. Guerre-Millo M. Adiponectin: an update. Diabetes Metab 2008;34:
128.
155. Gavrila A, Peng CK, Chan JL, Mietus JE, Goldberger AL,
Mantzoros CS. Diurnal and ultradian dynamics of serum adiponectin
in healthy men: comparison with leptin, circulating soluble leptin re-
ceptor, and cortisol patterns. J Clin Endocrinol Metab 2003;88:
283843.
156. Hotta K, Funahashi T, Arita Y, Takahashi M, Matsuda M, Okamoto Y,
et al. Plasma concentrations of a novel, adipose-specic protein, adipo-
nectin, in type 2 diabetic patients. Arterioscler Thromb Vasc Biol
2000;20:15959.
157. Bottner A, Kratzsch J, Muller G, Kapellen TM, Bluher S, Keller E, et al.
Gender differences of adiponectin levels develop during the progression
of puberty and are related to serum androgen levels. J Clin Endocrinol
Metab 2004;89:405361.
158. Nishizawa H, Shimomura I, Kishida K, Maeda N, Kuriyama H,
Nagaretani H, et al. Androgens decrease plasma adiponectin, an insu-
lin-sensitizing adipocyte-derived protein. Diabetes 2002;51:273441.
159. Page ST, Herbst KL, Amory JK, Coviello AD, Anawalt BD,
Matsumoto AM, et al. Testosterone administration suppresses adipo-
nectin levels in men. J Androl 2005;26:8592.
160. Katsuki A, Suematsu M, Gabazza EC, Murashima S, Nakatani K,
Togashi K, et al. Increased oxidative stress is associated with decreased
circulating levels of adiponectin in Japanese metabolically obese, nor-
mal-weight men with normal glucose tolerance. Diabetes Res Clin
Pract 2006;73:3104.
161. Pawlak DB, Kushner JA, Ludwig DS. Effects of dietary glycaemic in-
dex on adiposity, glucose homoeostasis, and plasma lipids in animals.
Lancet 2004;364:77885.
162. Yamauchi T, Kamon J, Ito Y, Tsuchida A, Yokomizo T, Kita S, et al.
Cloning of adiponectin receptors that mediate antidiabetic metabolic
effects. Nature 2003;423:7629.
163. Kubota N, Terauchi Y, Yamauchi T, Kubota T, Moroi M, Matsui J, et al.
Disruption of adiponectin causes insulin resistance and neointimal for-
mation. J Biol Chem 2002;277:258636.
164. Yamauchi T, Nio Y, Maki T, Kobayashi M, Takazawa T, Iwabu M, et al.
Targeted disruption of AdipoR1 and AdipoR2 causes abrogation of adi-
ponectin binding and metabolic actions. Nat Med 2007;13:3329.
165. Lagaly DV, Aad PY, Grado-Ahuir JA, Hulsey LB, Spicer LJ. Role of
adiponectin in regulating ovarian theca and granulosa cell function.
Mol Cell Endocrinol 2008;284:3845.
166. Rodriguez-Pacheco F, Martinez-Fuentes AJ, Tovar S, Pinilla L, Tena-
Sempere M, Dieguez C, et al. Regulation of pituitary cell function by
adiponectin. Endocrinology 2007;148:40110.
167. Kadowaki T, Yamauchi T, Kubota N, Hara K, Ueki K, Tobe K. Adipo-
nectin and adiponectin receptors in insulin resistance, diabetes, and the
metabolic syndrome. J Clin Invest 2006;116:178492.
168. Arita Y, Kihara S, Ouchi N, Takahashi M, Maeda K, Miyagawa J, et al.
Paradoxical decrease of an adipose-specic protein, adiponectin, in
obesity. Biochem Biophys Res Commun 1999;257:7983.
169. Yang WS, Lee WJ, Funahashi T, Tanaka S, Matsuzawa Y, Chao CL,
et al. Weight reduction increases plasma levels of an adipose-derived
antiinammatory protein, adiponectin. J Clin Endocrinol Metab
2001;86:38159.
170. Ledoux S, Campos DB, Lopes FL, Dobias-Goff M, Palin MF,
Murphy BD. Adiponectin induces periovulatory changes in ovarian fol-
licular cells. Endocrinology 2006;147:517886.
171. Mitchell M, Armstrong DT, Robker RL, Norman RJ. Adipokines: im-
plications for female fertility and obesity. Reproduction 2005;130:
58397.
172. Kapoor N, Sankaran S, Hyer S, Shehata H. Diabetes in pregnancy: a re-
view of current evidence. Curr Opin Obstet Gynecol 2007;19:58690.
173. Cortelazzi D, Corbetta S, Ronzoni S, Pelle F, Marconi A, Cozzi V, et al.
Maternal and foetal resistin and adiponectin concentrations in normal
and complicated pregnancies. Clin Endocrinol (Oxf) 2007;66:44753.
174. Kyriakakou M, Malamitsi-Puchner A, Militsi H, Boutsikou T,
Margeli A, Hassiakos D, et al. Leptin and adiponectin concentrations
in intrauterine growth restricted and appropriate for gestational age fe-
tuses, neonates, and their mothers. Eur J Endocrinol 2008;158:3438.
175. Cust AE, Kaaks R, Friedenreich C, Bonnet F, Laville M, Lukanova A,
et al. Plasma adiponectin levels and endometrial cancer risk in pre- and
postmenopausal women. J Clin Endocrinol Metab 2007;92:25563.
176. Fukuhara A, Matsuda M, Nishizawa M, Segawa K, Tanaka M,
Kishimoto K, et al. Retraction. Science 2007;318:565.
177. Varma V, Yao-Borengasser A, Rasouli N, Bodles AM, Phanavanh B,
Lee MJ, et al. Human visfatin expression: relationship to insulin sensi-
tivity, intramyocellular lipids, and inammation. J Clin Endocrinol
Metab 2007;92:66672.
178. Korner A, Garten A, Bluher M, Tauscher R, Kratzsch J, Kiess W. Mo-
lecular characteristics of serum visfatin and differential detection by
immunoassays. J Clin Endocrinol Metab 2007;92:478391.
179. Oki K, Yamane K, Kamei N, Nojima H, Kohno N. Circulating visfatin
level is correlated with inammation, but not with insulin resistance.
Clin Endocrinol (Oxf) 2007;67:796800.
180. Mastorakos G, Valsamakis G, Papatheodorou DC, Barlas I, Margeli A,
Boutsiadis A, et al. The role of adipocytokines in insulin resistance in
normal pregnancy: visfatin concentrations in early pregnancy predict
insulin sensitivity. Clin Chem 2007;53:147783.
181. Lewandowski KC, Stojanovic N, Press M, Tuck SM, Szosland K,
Bienkiewicz M, et al. Elevated serumlevels of visfatin in gestational di-
abetes: a comparative study across various degrees of glucose tolerance.
Diabetologia 2007;50:10337.
182. Haider DG, Handisurya A, Storka A, Vojtassakova E, Luger A,
Pacini G, et al. Visfatin response to glucose is reduced in women
with gestational diabetes mellitus. Diabetes Care 2007;30:188991.
183. Hu W, Wang Z, Wang H, Huang H, Dong M. Serum visfatin levels in
late pregnancy and pre-eclampsia. Acta Obstet Gynecol Scand
2008;87:4138.
184. Fasshauer M, Bluher M, Stumvoll M, Tonessen P, Faber R, Stepan H.
Differential regulation of visfatin and adiponectin in pregnancies with
normal and abnormal placental function. Clin Endocrinol (Oxf)
2007;66:4349.
185. Carswell EA, Old LJ, Kassel RL, Green S, Fiore N, Williamson B. An
endotoxin-induced serum factor that causes necrosis of tumors. Proc
Natl Acad Sci U S A 1975;72:366670.
186. Orlinick JR, Chao MV. TNF-related ligands and their receptors. Cell
Signal 1998;10:54351.
187. Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL,
Ferrante AW Jr. Obesity is associated with macrophage accumulation
in adipose tissue. J Clin Invest 2003;112:1796808.
188. Hotamisligil GS, Arner P, Caro JF, Atkinson RL, Spiegelman BM. In-
creased adipose tissue expression of tumor necrosis factor-alpha in hu-
man obesity and insulin resistance. J Clin Invest 1995;95:240915.
189. Xu H, Sethi JK, Hotamisligil GS. Transmembrane tumor necrosis factor
(TNF)-alpha inhibits adipocyte differentiation by selectively activating
TNF receptor 1. J Biol Chem 1999;274:2628795.
190. Prins JB. Adipose tissue as an endocrine organ. Best Pract Res Clin
Endocrinol Metab 2002;16:63951.
191. Ryden M, Jocken J, van Harmelen V, Dicker A, Hoffstedt J, Wiren M,
et al. Comparative studies of the role of hormone-sensitive lipase and
adipose triglyceride lipase in human fat cell lipolysis. Am J Physiol En-
docrinol Metab 2007;292:E184755.
192. Blum CA, Muller B, Huber P, Kraenzlin M, Schindler C, De Geyter C,
et al. Low-grade inammation and estimates of insulin resistance dur-
ing the menstrual cycle in lean and overweight women. J Clin Endocri-
nol Metab 2005;90:32305.
193. Bismar H, Diel I, Ziegler R, Pfeilschifter J. Increased cytokine secretion
by human bone marrow cells after menopause or discontinuation of
estrogen replacement. J Clin Endocrinol Metab 1995;80:33515.
194. Fried SK, Bunkin DA, Greenberg AS. Omental and subcutaneous adi-
pose tissues of obese subjects release interleukin-6: depot difference
and regulation by glucocorticoid. J Clin Endocrinol Metab 1998;83:
84750.
818
Bohler et al. Adipose tissue and reproduction in women Vol. 94, No. 3, August 2010
195. Hotamisligil GS, Murray DL, Choy LN, Spiegelman BM. Tumor necro-
sis factor alpha inhibits signaling from the insulin receptor. Proc Natl
Acad Sci U S A 1994;91:48548.
196. Fernandez-Real JM, Ricart W. Insulin resistance and chronic cardiovas-
cular inammatory syndrome. Endocr Rev 2003;24:278301.
197. Hotamisligil GS, Shargill NS, Spiegelman BM. Adipose expression of
tumor necrosis factor-alpha: direct role in obesity-linked insulin resis-
tance. Science 1993;259:8791.
198. Garg A. Regional adiposity and insulin resistance. J Clin Endocrinol
Metab 2004;89:420610.
199. Watanobe H, Hayakawa Y. Hypothalamic interleukin-1 beta and tumor
necrosis factor-alpha, but not interleukin-6, mediate the endotoxin-in-
duced suppression of the reproductive axis in rats. Endocrinology
2003;144:486875.
200. Fernandez-Real JM, Broch M, Vendrell J, Ricart W. Insulin resistance,
inammation, and serum fatty acid composition. Diabetes Care
2003;26:13628.
201. Boulanger MJ, Chow DC, Brevnova EE, Garcia KC. Hexameric struc-
ture and assembly of the interleukin-6/IL-6 alpha-receptor/gp130 com-
plex. Science 2003;300:21014.
202. Scheller J, Ohnesorge N, Rose-John S. Interleukin-6 trans-signalling in
chronic inammation and cancer. Scand J Immunol 2006;63:3219.
203. Bergman RN, Ader M. Free fatty acids and pathogenesis of type 2 di-
abetes mellitus. Trends Endocrinol Metab 2000;11:3516.
204. Tsigos C, Papanicolaou DA, Kyrou I, Defensor R, Mitsiadis CS,
Chrousos GP. Dose-dependent effects of recombinant human interleu-
kin-6 on glucose regulation. J Clin Endocrinol Metab 1997;82:416770.
205. Willis C, Morris JM, Danis V, Gallery ED. Cytokine production by
peripheral blood monocytes during the normal human ovulatory
menstrual cycle. Hum Reprod 2003;18:11738.
206. Van der Hoek KH, Woodhouse CM, BrannstromM, Norman RJ. Effects
of interleukin (IL)-6 on luteinizing hormone and IL-1betainduced
ovulation and steroidogenesis in the rat ovary. Biol Reprod 1998;58:
126671.
207. OBrien SM, Fitzgerald P, Scully P, Landers A, Scott LV, Dinan TG. Im-
pact of gender and menstrual cycle phase on plasma cytokine concen-
trations. Neuroimmunomodulation 2007;14:8490.
208. Jilma B, Dirnberger E, Loscher I, Rumplmayr A, Hildebrandt J,
Eichler HG, et al. Menstrual cycle-associated changes in blood levels
of interleukin-6, alpha1 acid glycoprotein, and C-reactive protein. J
Lab Clin Med 1997;130:6975.
209. Al-Harthi L, Wright DJ, Anderson D, Cohen M, Matity Ahu D, Cohn J,
et al. The impact of the ovulatory cycle on cytokine production: evalu-
ation of systemic, cervicovaginal, and salivary compartments. J Inter-
feron Cytokine Res 2000;20:71924.
210. Mikuni M. [Effect of interleukin-2 and interleukin-6 on ovary in the
ovulatory periodestablishment of a new ovarian perfusion system
and inuence of interleukins on ovulation rate and steroid secretion].
Hokkaido Igaku Zasshi 1995;70:56172. Japanese.
211. Deura I, Harada T, Taniguchi F, Iwabe T, Izawa M, Terakawa N. Reduc-
tion of estrogen production by interleukin-6 in a human granulosa tu-
mor cell line may have implications for endometriosis-associated
infertility. Fertil Steril 2005;83(Suppl 1):108692.
212. Russell SH, Small CJ, Stanley SA, Franks S, Ghatei MA, Bloom SR.
The in vitro role of tumour necrosis factor-alpha and interleukin-6 in
the hypothalamic-pituitary gonadal axis. J Neuroendocrinol 2001;13:
296301.
213. Harada T, Yoshioka H, Yoshida S, Iwabe T, Onohara Y, Tanikawa M,
et al. Increased interleukin-6 levels in peritoneal uid of infertile pa-
tients with active endometriosis. Am J Obstet Gynecol 1997;176:
5937.
214. Yoshida S, Harada T, Iwabe T, Taniguchi F, Mitsunari M, Yamauchi N,
et al. A combination of interleukin-6 and its soluble receptor impairs
sperm motility: implications in infertility associated with endometri-
osis. Hum Reprod 2004;19:18215.
215. Tsudo T, Harada T, Iwabe T, Tanikawa M, Nagano Y, Ito M, et al.
Altered gene expression and secretion of interleukin-6 in stromal cells
derived from endometriotic tissues. Fertil Steril 2000;73:20511.
216. Praus M, Wauterickx K, Collen D, Gerard RD. Reduction of tumor cell
migration and metastasis by adenoviral gene transfer of plasminogen
activator inhibitors. Gene Ther 1999;6:22736.
217. Aso Y. Plasminogen activator inhibitor (PAI)-1 in vascular inamma-
tion and thrombosis. Front Biosci 2007;12:295766.
218. Alessi MC, Poggi M, Juhan-Vague I. Plasminogen activator inhibitor-1,
adipose tissue and insulin resistance. Curr Opin Lipidol 2007;18:2405.
219. Ebisch IM, Thomas CM, Wetzels AM, Willemsen WN, Sweep FC,
Steegers-Theunissen RP. Review of the role of the plasminogen activa-
tor system and vascular endothelial growth factor in subfertility. Fertil
Steril 2008.
220. Glueck CJ, Wang P, Kobayashi S, Phillips H, Sieve-Smith L. Metformin
therapy throughout pregnancy reduces the development of gestational
diabetes in women with polycystic ovary syndrome. Fertil Steril
2002;77:5205.
221. Tarkun I, Canturk Z, Arslan BC, Turemen E, Tarkun P. The plasmino-
gen activator system in young and lean women with polycystic ovary
syndrome. Endocr J 2004;51:46772.
222. Ehrmann DA, Schneider DJ, Sobel BE, Cavaghan MK, Imperial J,
Roseneld RL, et al. Troglitazone improves defects in insulin action,
insulin secretion, ovarian steroidogenesis, and brinolysis in women
with polycystic ovary syndrome. J Clin Endocrinol Metab 1997;82:
210816.
223. Ma LJ, Mao SL, Taylor KL, Kanjanabuch T, Guan Y, Zhang Y, et al.
Prevention of obesity and insulin resistance in mice lacking plasmino-
gen activator inhibitor 1. Diabetes 2004;53:33646.
224. Alessi MC, Peiretti F, Morange P, Henry M, Nalbone G, Juhan-Vague I.
Production of plasminogen activator inhibitor 1 by human adipose
tissue: possible link between visceral fat accumulation and vascular
disease. Diabetes 1997;46:8607.
225. Chappell LC, Seed PT, Briley A, Kelly FJ, Hunt BJ, Charnock-
Jones DS, et al. Alongitudinal study of biochemical variables in women
at risk of preeclampsia. Am J Obstet Gynecol 2002;187:12736.
226. Steppan CM, Bailey ST, Bhat S, Brown EJ, Banerjee RR, Wright CM,
et al. The hormone resistin links obesity to diabetes. Nature 2001;409:
30712.
227. Kim KH, Lee K, Moon YS, Sul HS. A cysteine-rich adipose tissue-spe-
cic secretory factor inhibits adipocyte differentiation. J Biol Chem
2001;276:112526.
228. Yannakoulia M, Yiannakouris N, Bluher S, Matalas AL, Klimis-
Zacas D, Mantzoros CS. Body fat mass and macronutrient intake in re-
lation to circulating soluble leptin receptor, free leptin index, adiponec-
tin, and resistin concentrations in healthy humans. J Clin Endocrinol
Metab 2003;88:17306.
229. Tilg H, Moschen AR. Adipocytokines: mediators linking adipose tis-
sue, inammation and immunity. Nat Rev Immunol 2006;6:77283.
230. Morash BA, Willkinson D, Ur E, Wilkinson M. Resistin expression and
regulation in mouse pituitary. FEBS Lett 2002;526:2630.
231. Patel L, Buckels AC, Kinghorn IJ, Murdock PR, Holbrook JD,
Plumpton C, et al. Resistin is expressed in human macrophages and di-
rectly regulated by PPAR gamma activators. Biochem Biophys Res
Commun 2003;300:4726.
232. Caja S, Puerta M. Control by reproduction-related hormones of resistin
expression and plasma concentration. HormMetab Res 2007;39:5016.
233. Chen YH, Lee MJ, Chang HH, Hung PF, Kao YH. 17 beta-estradiol
stimulates resistin gene expression in 3T3-L1 adipocytes via the
estrogen receptor, extracellularly regulated kinase, and CCAAT/
enhancer binding protein-alpha pathways. Endocrinology 2006;147:
4496504.
234. Palik E, Baranyi E, Melczer Z, Audikovszky M, Szocs A, Winkler G,
et al. Elevated serum acylated (biologically active) ghrelin and resistin
levels associate with pregnancy-induced weight gain and insulin resis-
tance. Diabetes Res Clin Pract 2007;76:3517.
235. Chen D, Dong M, Fang Q, He J, Wang Z, Yang X. Alterations of serum
resistin in normal pregnancy and pre-eclampsia. Clin Sci (Lond)
2005;108:814.
236. Zhou Y, Zhang M, Guo W, Yu M, Xue K, Huang S, et al. Expression of
resistin protein in normal human subcutaneous adipose tissue and
Fertility and Sterility

819
pregnant women subcutaneous adipose tissue and placenta. J Huazhong
Univ Sci Technolog Med Sci 2006;26:28891.
237. Chen D, Fang Q, Chai Y, Wang H, Huang H, Dong M. Serum resistin in
gestational diabetes mellitus and early postpartum. Clin Endocrinol
(Oxf) 2007;67:20811.
238. Megia A, Vendrell J, Gutierrez C, Sabate M, Broch M, Fernandez-
Real JM, et al. Insulin sensitivity and resistin levels in gestational
diabetes mellitus and after parturition. Eur J Endocrinol 2008;158:
1738.
239. Seow KM, Juan CC, Hsu YP, Ho LT, Wang YY, Hwang JL. Serum and
follicular resistin levels in women with polycystic ovarian syndrome
during IVF-stimulated cycles. Hum Reprod 2005;20:11721.
240. Munir I, Yen HW, Baruth T, Tarkowski R, Azziz R, Magofn DA, et al.
Resistin stimulation of 17alpha-hydroxylase activity in ovarian theca
cells in vitro: relevance to polycystic ovary syndrome. J Clin Endocrinol
Metab 2005;90:48527.
241. Seow KM, Juan CC, Ho LT, Hsu YP, Lin YH, Huang LW, et al. Adipo-
cyte resistin mRNA levels are down-regulated by laparoscopic ovarian
electrocautery in both obese and lean women with polycystic ovary syn-
drome. Hum Reprod 2007;22:11006.
242. Chen YC, Tsai EM, Chen HS, Liu YH, Lee CH, Chou FH, et al. Serum
resistin level is a predictor of ovarian response in in vitro fertilisation
cycle. Acta Obstet Gynecol Scand 2007;86:9637.
243. Borst SE. The role of TNF-alpha in insulin resistance. Endocrine
2004;23:17782.
244. Svedberg J, Stromblad G, Wirth A, Smith U, Bjorntorp P. Fatty acids in
the portal vein of the rat regulate hepatic insulin clearance. J Clin Invest
1991;88:20548.
245. Bjorntorp P. [Metabolic difference between visceral fat and subcutane-
ous abdominal fat]. Diabetes Metab 2000;26(Suppl 3):102.
246. Svedberg J, Bjorntorp P, Smith U, Lonnroth P. Free-fatty acid inhibition
of insulin binding, degradation, and action in isolated rat hepatocytes.
Diabetes 1990;39:5704.
247. Delarue J, Magnan C. Free fatty acids and insulin resistance. Curr Opin
Clin Nutr Metab Care 2007;10:1428.
248. Haber EP, Procopio J, Carvalho CR, Carpinelli AR, Newsholme P,
Curi R. New insights into fatty acid modulation of pancreatic beta-
cell function. Int Rev Cytol 2006;248:141.
249. Mu YM, Yanase T, Nishi Y, Tanaka A, Saito M, Jin CH, et al. Saturated
FFAs, palmitic acid and stearic acid, induce apoptosis in human granu-
losa cells. Endocrinology 2001;142:35907.
250. Lovejoy JC, de la Bretonne JA, Klemperer M, Tulley R. Abdominal fat
distribution and metabolic risk factors: effects of race. Metabolism
1996;45:111924.
251. Weinsier RL, Hunter GR, Gower BA, Schutz Y, Darnell BE,
Zuckerman PA. Body fat distribution in white and black women: differ-
ent patterns of intraabdominal and subcutaneous abdominal adipose tis-
sue utilization with weight loss. Am J Clin Nutr 2001;74:6316.
252. Goran MI, Nagy TR, Treuth MS, Trowbridge C, Dezenberg C,
McGloin A, et al. Visceral fat in white and African American prepuber-
tal children. Am J Clin Nutr 1997;65:17038.
253. Albu JB, Kovera AJ, Johnson JA. Fat distribution and health in obesity.
Ann N YAcad Sci 2000;904:491501.
254. Albu JB, Murphy L, Frager DH, Johnson JA, Pi-Sunyer FX. Visceral fat
and race-dependent health risks in obese nondiabetic premenopausal
women. Diabetes 1997;46:45662.
255. Yanovski JA, Yanovski SZ, Filmer KM, Hubbard VS, Avila N, Lewis B,
et al. Differences in body composition of black and white girls. Am J
Clin Nutr 1996;64:8339.
256. Stanforth PR, Jackson AS, Green JS, Gagnon J, Rankinen T, Despres JP,
et al. Generalized abdominal visceral fat prediction models for black
and white adults aged 1765 y: the HERITAGE Family Study. Int J
Obes Relat Metab Disord 2004;28:92532.
257. Lara-Castro C, Weinsier RL, Hunter GR, Desmond R. Visceral adipose
tissue in women: longitudinal study of the effects of fat gain, time, and
race. Obes Res 2002;10:86874.
258. Perry AC, Applegate EB, Jackson ML, Deprima S, Goldberg RB,
Ross R, et al. Racial differences in visceral adipose tissue but not an-
thropometric markers of health-related variables. J Appl Physiol
2000;89:63643.
259. Hill JO, Sidney S, Lewis CE, Tolan K, Scherzinger AL, Stamm ER. Ra-
cial differences in amounts of visceral adipose tissue in young adults:
the CARDIA (Coronary Artery Risk Development in Young Adults)
study. Am J Clin Nutr 1999;69:3817.
260. Lovejoy JC, Smith SR, Rood JC. Comparison of regional fat distribu-
tion and health risk factors in middle-aged white and African American
women: the Healthy Transitions study. Obes Res 2001;9:106.
261. Araneta MR, Barrett-Connor E. Ethnic differences in visceral adipose
tissue and type 2 diabetes: Filipino, African-American, and white
women. Obes Res 2005;13:145865.
262. Park YW, Allison DB, Heymseld SB, Gallagher D. Larger amounts of
visceral adipose tissue in Asian Americans. Obes Res 2001;9:3817.
263. Okosun IS, Tedders SH, Choi S, Dever GE. Abdominal adiposity values
associated with established body mass indexes in white, black and His-
panic Americans. Astudy from the Third National Health and Nutrition
Examination Survey. Int J Obes Relat Metab Disord 2000;24:127985.
264. Tanaka S, Horimai C, Katsukawa F. Ethnic differences in abdominal
visceral fat accumulation between Japanese, African-Americans, and
caucasians: a meta-analysis. Acta Diabetol 2003;40(Suppl 1):S3024.
265. Deurenberg P, Deurenberg-Yap M. Differences in body-composition
assumptions across ethnic groups: practical consequences. Curr Opin
Clin Nutr Metab Care 2001;4:37783.
266. Deurenberg P, Yap M. The assessment of obesity: methods for measur-
ing body fat and global prevalence of obesity. Baillieres Best Pract Res
Clin Endocrinol Metab 1999;13:111.
267. Garrow JS, Webster J. Quetelets index (W/H2) as a measure of fatness.
Int J Obes 1985;9:14753.
268. Prentice AM, Jebb SA. Beyond body mass index. Obes Rev 2001;2:
1417.
269. National Institutes of Health. Clinical guidelines on the identication,
evaluation, and treatment of overweight and obesity in adultsthe
evidence report. Obes Res 1998;6(Suppl 2):51S209S.
270. Yildirim B, Sabir N, Kaleli B. Relation of intra-abdominal fat distribu-
tion to metabolic disorders in nonobese patients with polycystic ovary
syndrome. Fertil Steril 2003;79:135864.
271. Kahn SE, Prigeon RL, Schwartz RS, Fujimoto WY, Knopp RH,
Brunzell JD, et al. Obesity, body fat distribution, insulin sensitivity
and islet beta-cell function as explanations for metabolic diversity.
J Nutr 2001;131:354S60S.
272. Deurenberg P, Deurenberg-Yap M. Validity of body composition methods
across ethnic population groups. Acta Diabetol 2003;40(Suppl. 1):S2469.
273. Pouliot MC, Despres JP, Lemieux S, Moorjani S, Bouchard C,
Tremblay A, et al. Waist circumference and abdominal sagittal diame-
ter: best simple anthropometric indexes of abdominal visceral adipose
tissue accumulation and related cardiovascular risk in men and women.
Am J Cardiol 1994;73:4608.
274. Taylor RW, Keil D, Gold EJ, Williams SM, Goulding A. Body mass in-
dex, waist girth, and waist-to-hip ratio as indexes of total and regional
adiposity in women: evaluation using receiver operating characteristic
curves. Am J Clin Nutr 1998;67:449.
275. Harris TB, Visser M, Everhart J, Cauley J, Tylavsky F, Fuerst T, et al.
Waist circumference and sagittal diameter reect total body fat better
than visceral fat in older men and women. The Health, Aging and
Body Composition study. Ann N YAcad Sci 2000;904:46273.
276. Gray DS, Fujioka K, Colletti PM, Kim H, Devine W, Cuyegkeng T,
et al. Magnetic-resonance imaging used for determining fat distribution
in obesity and diabetes. Am J Clin Nutr 1991;54:6237.
277. van der Kooy K, Seidell JC. Techniques for the measurement of visceral
fat: a practical guide. Int J Obes Relat Metab Disord 1993;17:18796.
278. Grenier L. Apple shape puts overweight folks on fast track for disease. Re-
search Magazine, Spring/Summer 2005. Available at: http://www.
research.vt.edu/resmag/2005resmag1/apple.html. Last accessed July 28,
2008.
279. Demerath EW, Shen W, Lee M, Choh AC, Czerwinski SA,
Siervogel RM, et al. Approximation of total visceral adipose tissue
witha single magnetic resonance image. AmJ ClinNutr 2007;85:3628.
820
Bohler et al. Adipose tissue and reproduction in women Vol. 94, No. 3, August 2010
280. Shen W, Punyanitya M, Wang Z, Gallagher D, St-Onge MP, Albu J,
et al. Visceral adipose tissue: relations between single-slice areas and
total volume. Am J Clin Nutr 2004;80:2718.
281. Staten MA, Totty WG, Kohrt WM. Measurement of fat distribution by
magnetic resonance imaging. Invest Radiol 1989;24:3459.
282. Tulloch-Reid MK, Hanson RL, Sebring NG, Reynolds JC,
Premkumar A, Genovese DJ, et al. Both subcutaneous and visceral ad-
ipose tissue correlate highly with insulin resistance in african ameri-
cans. Obes Res 2004;12:13529.
283. Slyper AH. Childhood obesity, adipose tissue distribution, and the
pediatric practitioner. Pediatrics 1998;102:e4.
284. Kaplowitz P. Pubertal development in girls: secular trends. Curr Opin
Obstet Gynecol 2006;18:48791.
285. Slyper AH. The pubertal timing controversy in the USA, and a reviewof
possible causative factors for the advance in timing of onset of puberty.
Clin Endocrinol (Oxf) 2006;65:18.
286. Wang Y. Is obesity associated with early sexual maturation? A compar-
ison of the association in American boys versus girls. Pediatrics
2002;110:90310.
287. Kaplowitz PB. Link between body fat and the timing of puberty. Pedi-
atrics 2008;121(Suppl 3):S20817.
288. de Ridder CM, Thijssen JH, Bruning PF, Van den Brande JL,
Zonderland ML, Erich WB. Body fat mass, body fat distribution,
and pubertal development: a longitudinal study of physical and hor-
monal sexual maturation of girls. J Clin Endocrinol Metab 1992;75:
4426.
289. Lassek WD, Gaulin SJ. Brief communication: menarche is related to fat
distribution. Am J Phys Anthropol 2007;133:114751.
290. Diaz A, Laufer MR, Breech LL. Menstruation in girls and adolescents:
using the menstrual cycle as a vital sign. Pediatrics 2006;118:224550.
291. Chumlea WC, Schubert CM, Roche AF, Kulin HE, Lee PA, Himes JH,
et al. Age at menarche and racial comparisons in US girls. Pediatrics
2003;111:1103.
292. Herman-Giddens ME, Slora EJ, Wasserman RC, Bourdony CJ,
Bhapkar MV, Koch GG, et al. Secondary sexual characteristics and
menses in young girls seen in ofce practice: a study from the Pediatric
Research in Ofce Settings network. Pediatrics 1997;99:50512.
293. Wong WW, Butte NF, Ellis KJ, Hergenroeder AC, Hill RB, Stuff JE,
et al. Pubertal African-American girls expend less energy at rest and
during physical activity than caucasian girls. J Clin Endocrinol Metab
1999;84:90611.
294. Kumanyika SK. Obesity in minority populations: an epidemiologic
assessment. Obes Res 1994;2:16682.
295. Farooqi IS, Wangensteen T, Collins S, Kimber W, Matarese G,
Keogh JM, et al. Clinical and molecular genetic spectrum of congenital
deciency of the leptin receptor. N Engl J Med 2007;356:23747.
296. Warren MP, Perlroth NE. The effects of intense exercise on the female
reproductive system. J Endocrinol 2001;170:311.
297. Thong FS, McLean C, Graham TE. Plasma leptin in female athletes:
relationship with body fat, reproductive, nutritional, and endocrine
factors. J Appl Physiol 2000;88:203744.
298. Warren MP, Voussoughian F, Geer EB, Hyle EP, Adberg CL,
Ramos RH. Functional hypothalamic amenorrhea: hypoleptinemia
and disordered eating. J Clin Endocrinol Metab 1999;84:8737.
299. Yager J, Andersen AE. Clinical practice. Anorexia nervosa. N Engl J
Med 2005;353:14818.
300. Wong WW, Nicolson M, Stuff JE, Butte NF, Ellis KJ, Hergenroeder AC,
et al. Serum leptin concentrations in Caucasian and African-American
girls. J Clin Endocrinol Metab 1998;83:35747.
301. Wilson ME, Fisher J, Chikazawa K, Yoda R, Legendre A, Mook D, et al.
Leptin administration increases nocturnal concentrations of luteinizing
hormone and growth hormone in juvenile female rhesus monkeys.
J Clin Endocrinol Metab 2003;88:487483.
302. Plant TM, Durrant AR. Circulating leptin does not appear to provide
a signal for triggering the initiation of puberty in the male rhesus mon-
key (Macaca mulatta). Endocrinology 1997;138:45058.
303. Barker-Gibb ML, Sahu A, Pohl CR, Plant TM. Elevating circulating
leptin in prepubertal male rhesus monkeys (Macaca mulatta) does
not elicit precocious gonadotropin-releasing hormone release, assessed
indirectly. J Clin Endocrinol Metab 2002;87:497683.
304. Saenger P, Dimartino-Nardi J. Premature adrenarche. J Endocrinol
Invest 2001;24:72433.
305. Ghizzoni L, Milani S. The natural history of premature adrenarche.
J Pediatr Endocrinol Metab 2000;13(Suppl 5):124751.
306. Maclaren NK, Gujral S, Ten S, Motagheti R. Childhood obesity and
insulin resistance. Cell Biochem Biophys 2007;48:738.
307. Ibanez L, Ong K, Valls C, Marcos MV, Dunger DB, de Zegher F.
Metformin treatment to prevent early puberty in girls with precocious
pubarche. J Clin Endocrinol Metab 2006;91:288891.
308. Dacou-Voutetakis C, Dracopoulou M. High incidence of molecular de-
fects of the CYP21 gene in patients with premature adrenarche. J Clin
Endocrinol Metab 1999;84:15704.
309. Hsu HT, Chang YC, Chiu YN, Liu CL, Chang KJ, Guo IC. Leptin inter-
feres with adrenocorticotropin/3
0
,5
0
-cyclic adenosine monophosphate
(cAMP) signaling, possibly through a Janus kinase 2phosphatidylino-
sitol 3-kinase/Aktphosphodiesterase 3cAMP pathway, to down-
regulate cholesterol side-chain cleavage cytochrome P450 enzyme in
human adrenocortical NCI-H295 cell line. J Clin Endocrinol Metab
2006;91:27619.
310. Jethwa PH, Smith KL, Small CJ, Abbott CR, Darch SJ, Murphy KG,
et al. Neuromedin U partially mediates leptin-induced hypothalamo-
pituitary adrenal (HPA) stimulation and has a physiological role in
the regulation of the HPA axis in the rat. Endocrinology 2006;147:
288692.
311. Welt CK, Chan JL, Bullen J, Murphy R, Smith P, DePaoli AM, et al.
Recombinant human leptin in women with hypothalamic amenorrhea.
N Engl J Med 2004;351:98797.
312. Miller KK, Grinspoon S, Gleysteen S, Grieco KA, Ciampa J, Breu J,
et al. Preservation of neuroendocrine control of reproductive function
despite severe undernutrition. J Clin Endocrinol Metab 2004;89:
44348.
313. Bluher S, Mantzoros CS. Leptin in reproduction. Curr Opin Endocrinol
Diabetes Obes 2007;14:45864.
314. Yu WH, Kimura M, Walczewska A, Karanth S, McCann SM. Role of
leptin in hypothalamic-pituitary function. Proc Natl Acad Sci U S A
1997;94:10238.
315. Chan JL, Mantzoros CS. Leptin and the hypothalamic-pituitary regula-
tion of the gonadotropin-gonadal axis. Pituitary 2001;4:8792.
316. Yura S, Ogawa Y, Sagawa N, Masuzaki H, Itoh H, Ebihara K, et al. Ac-
celerated puberty and late-onset hypothalamic hypogonadismin female
transgenic skinny mice overexpressing leptin. J Clin Invest 2000;105:
74955.
317. Tena-Sempere M, Pinilla L, Gonzalez LC, Navarro J, Dieguez C,
Casanueva FF, et al. In vitro pituitary and testicular effects of the lep-
tin-related synthetic peptide leptin (116130) amide involve actions
both similar to and distinct from those of the native leptin molecule
in the adult rat. Eur J Endocrinol 2000;142:40610.
318. Oral EA, Simha V, Ruiz E, Andewelt A, Premkumar A, Snell P, et al.
Leptin-replacement therapy for lipodystrophy. N Engl J Med
2002;346:5708.
319. Musso C, Cochran E, Javor E, Young J, Depaoli AM, Gorden P. The
long-termeffect of recombinant methionyl human leptin therapy on hy-
perandrogenism and menstrual function in female and pituitary func-
tion in male and female hypoleptinemic lipodystrophic patients.
Metabolism 2005;54:25563.
320. Tomova A, Makker K, Kirilov G, Agarwal A, Kumanov P. Disturbances
in gonadal axis in women with anorexia nervosa. Eat Weight Disord
2007;12:e927.
321. Golden NH, Jacobson MS, Schebendach J, Solanto MV, Hertz SM,
Shenker IR. Resumption of menses in anorexia nervosa. Arch Pediatr
Adolesc Med 1997;151:1621.
322. Warren MP. Effects of undernutrition on reproductive function in the
human. Endocr Rev 1983;4:36377.
323. Pirke KM, Schweiger U, Lemmel W, Krieg JC, Berger M. The inuence
of dieting on the menstrual cycle of healthy young women. J Clin
Endocrinol Metab 1985;60:11749.
Fertility and Sterility

821
324. Berga SL, Loucks TL, Cameron JL. Endocrine and chronobiological
effects of fasting in women. Fertil Steril 2001;75:92632.
325. Chan JL, Matarese G, Shetty GK, Raciti P, Kelesidis I, Auero D, et al.
Differential regulation of metabolic, neuroendocrine, and immune
function by leptin in humans. Proc Natl Acad Sci U S A 2006;103:
84816.
326. Golden NH, Carlson JL. The pathophysiology of amenorrhea in the ad-
olescent. Ann N YAcad Sci 2008;1135:16378.
327. Nutrition and reproduction in women. Hum Reprod Update 2006;12:
193207.
328. Ramsay JE, Greer I, Sattar N. ABC of obesity. Obesity and reproduc-
tion. BMJ 2006;333:115962.
329. Pasquali R, Pelusi C, Genghini S, Cacciari M, Gambineri A. Obesity
and reproductive disorders in women. Hum Reprod Update 2003;9:
35972.
330. Gray AD, Power ML, Zinberg S, Schulkin J. Assessment and manage-
ment of obesity. Obstet Gynecol Surv 2006;61:7428.
331. Balen AH, Platteau P, Andersen AN, Devroey P, Sorensen P,
Helmgaard L, et al. The inuence of body weight on response to ovu-
lation induction with gonadotrophins in 335 women with World Health
Organization group II anovulatory infertility. BJOG 2006;113:
1195202.
332. Styne-Gross A, Elkind-Hirsch K, Scott RT Jr. Obesity does not impact
implantation rates or pregnancy outcome in women attempting concep-
tion through oocyte donation. Fertil Steril 2005;83:162934.
333. Fedorcsak P, Storeng R, Dale PO, Tanbo T, AbyholmT. Obesity is a risk
factor for early pregnancy loss after IVF or ICSI. Acta Obstet Gynecol
Scand 2000;79:438.
334. Bellver J, Melo MA, Bosch E, Serra V, Remohi J, Pellicer A. Obesity
and poor reproductive outcome: the potential role of the endometrium.
Fertil Steril 2007.
335. Winter E, Wang J, Davies MJ, Norman R. Early pregnancy loss follow-
ing assisted reproductive technology treatment. Hum Reprod 2002;17:
32203.
336. McCartney CR, Prendergast KA, Chhabra S, Eagleson CA, Yoo R,
Chang RJ, et al. The association of obesity and hyperandrogenemia dur-
ing the pubertal transition in girls: obesity as a potential factor in the
genesis of postpubertal hyperandrogenism. J Clin Endocrinol Metab
2006;91:171422.
337. Barber TM, Golding SJ, Alvey C, Wass JA, Karpe F, Franks S, et al.
Global adiposity rather than abnormal regional fat distribution charac-
terises women with polycystic ovary syndrome. J Clin Endocrinol
Metab 2007.
338. Pi-Sunyer FX. Obesity: criteria and classication. Proc Nutr Soc
2000;59:5059.
339. Hollmann M, Runnebaum B, Gerhard I. Impact of waist-hip-ratio and
body-mass-index on hormonal and metabolic parameters in young,
obese women. Int J Obes Relat Metab Disord 1997;21:47683.
340. Kirschner MA, Samojlik E, Drejka M, Szmal E, Schneider G, Ertel N.
Androgen-estrogen metabolism in women with upper body versus
lower body obesity. J Clin Endocrinol Metab 1990;70:4739.
341. Gambineri A, Pelusi C, Vicennati V, Pagotto U, Pasquali R. Obesity and
the polycystic ovary syndrome. Int J Obes Relat Metab Disord 2002;26:
88396.
342. Pasquali R. Obesity and androgens: facts and perspectives. Fertil Steril
2006;85:131940.
343. Genazzani AR, Gambacciani M. Effect of climacteric transition and
hormone replacement therapy on body weight and body fat distribution.
Gynecol Endocrinol 2006;22:14550.
344. Haarbo J, Marslew U, Gotfredsen A, Christiansen C. Postmenopausal
hormone replacement therapy prevents central distribution of body
fat after menopause. Metabolism 1991;40:13236.
345. Elbers JM, Asscheman H, Seidell JC, Megens JA, Gooren LJ. Long-
term testosterone administration increases visceral fat in female to
male transsexuals. J Clin Endocrinol Metab 1997;82:20447.
346. Iverius PH, Brunzell JD. Relationship between lipoprotein lipase activ-
ity and plasma sex steroid level in obese women. J Clin Invest 1988;82:
110612.
347. Palin SL, McTernan PG, Anderson LA, Sturdee DW, Barnett AH,
Kumar S. 17beta-Estradiol and antiestrogen ICI: compound 182,780reg-
ulate expressionof lipoproteinlipase andhormone-sensitive lipase iniso-
lated subcutaneous abdominal adipocytes. Metabolism 2003;52:3838.
348. Pasquali R, Gambineri A, Pagotto U. The impact of obesity on repro-
duction in women with polycystic ovary syndrome. BJOG 2006;113:
114859.
349. Guzick DS, Wing R, Smith D, Berga SL, Winters SJ. Endocrine conse-
quences of weight loss in obese, hyperandrogenic, anovulatory women.
Fertil Steril 1994;61:598604.
350. Kiddy DS, Hamilton-Fairley D, Bush A, Short F, Anyaoku V, Reed MJ,
et al. Improvement in endocrine and ovarian function during dietary
treatment of obese women with polycystic ovary syndrome. Clin Endo-
crinol (Oxf) 1992;36:10511.
351. Reinehr T, de Sousa G, Roth CL, Andler W. Androgens before and after
weight loss in obese children. J Clin Endocrinol Metab 2005;90:
558895.
352. Wabitsch M, Hauner H, Heinze E, Bockmann A, Benz R, Mayer H,
et al. Body fat distribution and steroid hormone concentrations in obese
adolescent girls before and after weight reduction. J Clin Endocrinol
Metab 1995;80:346975.
353. Phillips GB, Jing T, Heymseld SB. Does insulin resistance, visceral
adiposity, or a sex hormone alteration underlie the metabolic syn-
drome? Studies in women. Metabolism 2008;57:83844.
354. Pasquali R, Antenucci D, Casimirri F, Venturoli S, Paradisi R, Fabbri R,
et al. Clinical and hormonal characteristics of obese amenorrheic hyper-
androgenic women before and after weight loss. J Clin Endocrinol
Metab 1989;68:1739.
355. Baillargeon JP, Carpentier A. Role of insulin in the hyperandrogenemia
of lean women with polycystic ovary syndrome and normal insulin sen-
sitivity. Fertil Steril 2007.
356. Poretsky L. On the paradox of insulin-induced hyperandrogenism in
insulin-resistant states. Endocr Rev 1991;12:313.
357. Franks S, Kiddy DS, Hamilton-Fairley D, Bush A, Sharp PS, Reed MJ.
The role of nutrition and insulin in the regulation of sex hormone bind-
ing globulin. J Steroid Biochem Mol Biol 1991;39:8358.
358. Jensen MD, Haymond MW, Rizza RA, Cryer PE, Miles JM. Inuence
of body fat distribution on free fatty acid metabolism in obesity. J Clin
Invest 1989;83:116873.
359. Mai K, Bobbert T, Kullmann V, Andres J, Rochlitz H, Osterhoff M,
et al. Free fatty acids increase androgen precursors in vivo. J Clin
Endocrinol Metab 2006;91:15017.
360. Nimrod A, Ryan KJ. Aromatization of androgens by human abdominal
and breast fat tissue. J Clin Endocrinol Metab 1975;40:36772.
361. Simpson ER. Aromatization of androgens in women: current concepts
and ndings. Fertil Steril 2002;77(Suppl 4):S610.
362. Pedersen SB, Kristensen K, Hermann PA, Katzenellenbogen JA,
Richelsen B. Estrogen controls lipolysis by up-regulating alpha2A-ad-
renergic receptors directly in human adipose tissue through the estrogen
receptor alpha. Implications for the female fat distribution. J Clin Endo-
crinol Metab 2004;89:186978.
363. Mizutani T, Nishikawa Y, Adachi H, Enomoto T, Ikegami H, Kurachi H,
et al. Identication of estrogen receptor in human adipose tissue and
adipocytes. J Clin Endocrinol Metab 1994;78:9504.
364. Pedersen SB, Bruun JM, Hube F, Kristensen K, Hauner H, Richelsen B.
Demonstration of estrogen receptor subtypes alpha and beta in human
adipose tissue: inuences of adipose cell differentiation and fat depot
localization. Mol Cell Endocrinol 2001;182:2737.
365. Picard F, Deshaies Y, Lalonde J, Samson P, Labrie C, Belanger A, et al.
Effects of the estrogen antagonist EM-652.HCl on energy balance and
lipid metabolism in ovariectomized rats. Int J Obes Relat Metab Disord
2000;24:83040.
366. Hamosh M, Hamosh P. The effect of estrogen on the lipoprotein lipase
activity of rat adipose tissue. J Clin Invest 1975;55:11325.
367. Enzi G, Gasparo M, Biondetti PR, Fiore D, Semisa M, Zurlo F. Subcu-
taneous and visceral fat distribution according to sex, age, and over-
weight, evaluated by computed tomography. Am J Clin Nutr 1986;44:
73946.
822
Bohler et al. Adipose tissue and reproduction in women Vol. 94, No. 3, August 2010
368. Rebuffe-Scrive M, Enk L, Crona N, Lonnroth P, Abrahamsson L,
Smith U, et al. Fat cell metabolismin different regions in women. Effect
of menstrual cycle, pregnancy, and lactation. J Clin Invest 1985;75:
19736.
369. Rebuffe-Scrive M, Lonnroth P, Marin P, Wesslau C, Bjorntorp P,
Smith U. Regional adipose tissue metabolism in men and postmeno-
pausal women. Int J Obes 1987;11:34755.
370. Ostberg JE, Thomas EL, Hamilton G, Attar MJ, Bell JD, Conway GS.
Excess visceral and hepatic adipose tissue in Turner syndrome deter-
mined by magnetic resonance imaging: estrogen deciency associated
with hepatic adipose content. J Clin Endocrinol Metab 2005;90:
26315.
371. Elbers JM, Asscheman H, Seidell JC, Gooren LJ. Effects of sex steroid
hormones on regional fat depots as assessed by magnetic resonance im-
aging in transsexuals. Am J Physiol 1999;276:E31725.
372. Price TM, OBrien SN, Welter BH, George R, Anandjiwala J,
Kilgore M. Estrogen regulation of adipose tissue lipoprotein lipase
possible mechanism of body fat distribution. Am J Obstet Gynecol
1998;178:1017.
373. Toth MJ, Poehlman ET, Matthews DE, Tchernof A, MacCoss MJ. Ef-
fects of estradiol and progesterone on body composition, protein syn-
thesis, and lipoprotein lipase in rats. Am J Physiol Endocrinol Metab
2001;280:E496501.
374. Applebaum DM, Goldberg AP, Pykalisto OJ, Brunzell JD,
Hazzard WR. Effect of estrogen on post-heparin lipolytic activity. Se-
lective decline in hepatic triglyceride lipase. J Clin Invest 1977;59:
6018.
375. Rotterdam ESHRE/ASRMSponsored Consensus Workshop Group.
Revised 2003 consensus on diagnostic criteria and long-term health
risks related to polycystic ovary syndrome. Fertil Steril 2004;81:1925.
376. Berga SL, Guzick DS, Winters SJ. Increased luteinizing hormone and
alpha-subunit secretion in women with hyperandrogenic anovulation.
J Clin Endocrinol Metab 1993;77:895901.
377. Ovalle F, Azziz R. Insulin resistance, polycystic ovary syndrome, and
type 2 diabetes mellitus. Fertil Steril 2002;77:1095105.
378. Ehrmann DA. Polycystic ovary syndrome. N Engl J Med 2005;352:
122336.
379. Stein IF, Leventhal ML. Amenorrhea associated with bilateral polycys-
tic ovaries. Am J Obstet Gynecol 1935;29:18191.
380. Azziz R, Ehrmann D, Legro RS, Whitcomb RW, Hanley R,
Fereshetian AG, et al. Troglitazone improves ovulation and hirsutism
in the polycystic ovary syndrome: a multicenter, double blind, pla-
cebo-controlled trial. J Clin Endocrinol Metab 2001;86:162632.
381. Goldzieher JW, Green JA. The polycystic ovary. I. Clinical and histo-
logic features. J Clin Endocrinol Metab 1962;22:32538.
382. Douchi T, Ijuin H, Nakamura S, Oki T, Yamamoto S, Nagata Y. Body fat
distribution in women with polycystic ovary syndrome. Obstet Gynecol
1995;86:5169.
383. Holte J, Bergh T, Berne C, Wide L, Lithell H. Restored insulin sensitiv-
ity but persistently increased early insulin secretion after weight loss in
obese women with polycystic ovary syndrome. J Clin Endocrinol Metab
1995;80:258693.
384. Lord J, Thomas R, Fox B, Acharya U, Wilkin T. The central issue? Vis-
ceral fat mass is a good marker of insulin resistance and metabolic dis-
turbance in women with polycystic ovary syndrome. Bjog 2006;113:
12039.
385. Carmina E, Lobo RA. Use of fasting blood to assess the prevalence of
insulin resistance in women with polycystic ovary syndrome. Fertil
Steril 2004;82:6615.
386. Mlinar B, Marc J, Janez A, Pfeifer M. Molecular mechanisms of insulin
resistance and associated diseases. Clin Chim Acta 2007;375:2035.
387. Nestler JE. Insulin resistance syndrome and polycystic ovary syndrome.
Endocr Pract 2003;9(Suppl 2):869.
388. Corbould A, Zhao H, Mirzoeva S, Aird F, Dunaif A. Enhanced mito-
genic signaling in skeletal muscle of women with polycystic ovary syn-
drome. Diabetes 2006;55:7519.
389. Book CB, Dunaif A. Selective insulin resistance in the polycystic ovary
syndrome. J Clin Endocrinol Metab 1999;84:31106.
390. Dunaif A. Insulin resistance and the polycystic ovary syndrome: mech-
anism and implications for pathogenesis. Endocr Rev 1997;18:
774800.
391. Corbould A, Kim YB, Youngren JF, Pender C, Kahn BB, Lee A, et al.
Insulin resistance in the skeletal muscle of women with PCOS involves
intrinsic and acquired defects in insulin signaling. Am J Physiol Endo-
crinol Metab 2005;288:E104754.
392. Legro RS, Kunselman AR, Dodson WC, Dunaif A. Prevalence and pre-
dictors of risk for type 2 diabetes mellitus and impaired glucose toler-
ance in polycystic ovary syndrome: a prospective, controlled study in
254 affected women. J Clin Endocrinol Metab 1999;84:1659.
393. Legro RS, Gnatuk CL, Kunselman AR, Dunaif A. Changes in glucose
tolerance over time in women with polycystic ovary syndrome: a con-
trolled study. J Clin Endocrinol Metab 2005;90:323642.
394. Gambineri A, Pelusi C, Manicardi E, Vicennati V, Cacciari M, Mor-
selli-Labate AM, et al. Glucose intolerance in a large cohort of mediter-
ranean women with polycystic ovary syndrome: phenotype and
associated factors. Diabetes 2004;53:23538.
395. Huber-Buchholz MM, Carey DG, Norman RJ. Restoration of reproduc-
tive potential by lifestyle modication in obese polycystic ovary syn-
drome: role of insulin sensitivity and luteinizing hormone. J Clin
Endocrinol Metab 1999;84:14704.
396. Clark AM, Ledger W, Galletly C, Tomlinson L, Blaney F, Wang X, et al.
Weight loss results in signicant improvement in pregnancy and ovula-
tion rates in anovulatory obese women. HumReprod 1995;10:270512.
397. Pasquali R, Gambineri A, Biscotti D, Vicennati V, Gagliardi L,
Colitta D, et al. Effect of long-term treatment with metformin added
to hypocaloric diet on body composition, fat distribution, and androgen
and insulin levels in abdominally obese women with and without the
polycystic ovary syndrome. J Clin Endocrinol Metab 2000;85:276774.
398. Nestler JE, Jakubowicz DJ, Evans WS, Pasquali R. Effects of metfor-
min on spontaneous and clomiphene-induced ovulation in the polycys-
tic ovary syndrome. N Engl J Med 1998;338:187680.
399. Liu KE, Tataryn IV, Sagle M. Use of metformin for ovulation induction
in women who have polycystic ovary syndrome with or without evi-
dence of insulin resistance. J Obstet Gynaecol Can 2006;28:5959.
400. Faulds G, Ryden M, Ek I, Wahrenberg H, Arner P. Mechanisms behind
lipolytic catecholamine resistance of subcutaneous fat cells in the poly-
cystic ovarian syndrome. J Clin Endocrinol Metab 2003;88:226973.
401. Azziz R. Androgen excess is the key element in polycystic ovary syn-
drome. Fertil Steril 2003;80:2524.
402. Dicker A, Ryden M, Naslund E, Muehlen IE, Wiren M, Lafontan M,
et al. Effect of testosterone on lipolysis in human pre-adipocytes from
different fat depots. Diabetologia 2004;47:4208.
403. Zang H, Ryden M, Wahlen K, Dahlman-Wright K, Arner P, Linden
Hirschberg A. Effects of testosterone and estrogen treatment on lipoly-
sis signaling pathways in subcutaneous adipose tissue of postmeno-
pausal women. Fertil Steril 2007;88:1006.
404. Carmina E. Ovarian and adrenal hyperandrogenism. Ann N YAcad Sci
2006;1092:1307.
405. Barbieri RL, Makris A, Randall RW, Daniels G, Kistner RW, Ryan KJ.
Insulin stimulates androgen accumulation in incubations of ovarian
stroma obtained from women with hyperandrogenism. J Clin Endocri-
nol Metab 1986;62:90410.
406. Gilling-Smith C, Willis DS, Beard RW, Franks S. Hypersecretion of an-
drostenedione by isolated thecal cells from polycystic ovaries. J Clin
Endocrinol Metab 1994;79:115865.
407. Lobo RA, Paul WL, Goebelsmann U. Serum levels of DHEAS in gyne-
cologic endocrinopathy and infertility. Obstet Gynecol 1981;57:
60712.
408. Ehrmann DA, Roseneld RL, Barnes RB, Brigell DF, Sheikh Z. Detec-
tion of functional ovarian hyperandrogenism in women with androgen
excess. N Engl J Med 1992;327:15762.
409. Stewart PM, Shackleton CH, Beastall GH, Edwards CR. 5 alpha-Re-
ductase activity in polycystic ovary syndrome. Lancet 1990;335:4313.
410. Rodin A, Thakkar H, Taylor N, Clayton R. Hyperandrogenism in poly-
cystic ovary syndrome. Evidence of dysregulation of 11 beta-hydroxys-
teroid dehydrogenase. N Engl J Med 1994;330:4605.
Fertility and Sterility

823
411. Rainey WE, Carr BR, Sasano H, Suzuki T, Mason JI. Dissecting human
adrenal androgen production. Trends Endocrinol Metab 2002;13:
2349.
412. Macut D, Panidis D, Glisic B, Spanos N, Petakov M, Bjekic J, et al.
Lipid and lipoprotein prole in women with polycystic ovary syn-
drome. Can J Physiol Pharmacol 2008;86:199204.
413. Tsilchorozidou T, Honour JW, Conway GS. Altered cortisol metabo-
lism in polycystic ovary syndrome: insulin enhances 5alpha-reduction
but not the elevated adrenal steroid production rates. J Clin Endocrinol
Metab 2003;88:590713.
414. Gompel A, Wright F, Kuttenn F, Mauvais-Jarvis P. Contribution of
plasma androstenedioneto5alpha-androstanediol glucuronide inwomen
with idiopathic hirsutism. J Clin Endocrinol Metab 1986;62:4414.
415. Walker BR, AndrewR. Tissue production of cortisol by 11beta-hydrox-
ysteroid dehydrogenase type 1 and metabolic disease. Ann N YAcad
Sci 2006;1083:16584.
416. Wake DJ, Walker BR. Inhibition of 11beta-hydroxysteroid dehydroge-
nase type 1 in obesity. Endocrine 2006;29:1018.
417. Seckl JR, Walker BR. Minireview: 11beta-hydroxysteroid dehydroge-
nase type 1a tissue-specic amplier of glucocorticoid action. Endo-
crinology 2001;142:13716.
418. Katz JR, Mohamed-Ali V, Wood PJ, Yudkin JS, Coppack SW. An
in vivo study of the cortisol-cortisone shuttle in subcutaneous abdomi-
nal adipose tissue. Clin Endocrinol (Oxf) 1999;50:638.
419. Masuzaki H, Paterson J, Shinyama H, Morton NM, Mullins JJ,
Seckl JR, et al. Atransgenic model of visceral obesity and the metabolic
syndrome. Science 2001;294:216670.
420. Stewart PM, Boulton A, Kumar S, Clark PM, Shackleton CH. Cortisol
metabolismin human obesity: impaired cortisone/cortisol conversion
in subjects with central adiposity. J Clin Endocrinol Metab 1999;84:
10227.
421. Marin P, Darin N, Amemiya T, Andersson B, Jern S, Bjorntorp P. Cor-
tisol secretion in relation to body fat distribution in obese premeno-
pausal women. Metabolism 1992;41:8826.
422. Rask E, Walker BR, Soderberg S, Livingstone DE, Eliasson M,
Johnson O, et al. Tissue-specic changes in peripheral cortisol metab-
olism in obese women: increased adipose 11beta-hydroxysteroid dehy-
drogenase type 1 activity. J Clin Endocrinol Metab 2002;87:33306.
423. Bujalska IJ, Kumar S, Stewart PM. Does central obesity reect Cush-
ings disease of the omentum? Lancet 1997;349:12103.
424. Pedersen SB, Jonler M, Richelsen B. Characterization of regional and
gender differences in glucocorticoid receptors and lipoprotein lipase ac-
tivity in human adipose tissue. J Clin Endocrinol Metab 1994;78:13549.
425. Stewart PM. Tissue-specic Cushings syndrome, 11beta-hydroxyste-
roid dehydrogenases and the redenition of corticosteroid hormone
action. Eur J Endocrinol 2003;149:1638.
426. Rask E, Olsson T, Soderberg S, Andrew R, Livingstone DE, Johnson O,
et al. Tissue-specic dysregulation of cortisol metabolism in human
obesity. J Clin Endocrinol Metab 2001;86:141821.
427. Moschos S, Chan JL, Mantzoros CS. Leptin and reproduction: a review.
Fertil Steril 2002;77:43344.
428. Sepilian VP, Crochet JR, Nagamani M. Serum soluble leptin receptor
levels and free leptin index in women with polycystic ovary syndrome:
relationship to insulin resistance and androgens. Fertil Steril 2006;85:
14417.
429. Mei-Gen L, Guo-Lian D, Xi-Jing C, Xi-Ping L, Li-Jun D, Min-Yue D,
et al. Association of serum and follicular uid leptin concentrations
with granulosa cell p-STAT3 expression in fertile patients with polycys-
tic ovarian syndrome. J Clin Endocrinol Metab 2007.
430. Carmina E, Chu MC, Moran C, Tortoriello D, Vardhana P, Tena G, et al.
Subcutaneous and omental fat expression of adiponectin and leptin in
women with polycystic ovary syndrome. Fertil Steril 2008;89:6428.
431. Erturk E, Kuru N, Savci V, Tuncel E, Ersoy C, Imamoglu S. Serum lep-
tin levels correlate with obesity parameters but not with hyperinsulin-
ism in women with polycystic ovary syndrome. Fertil Steril 2004;82:
13648.
432. Romualdi D, Campagna G, Selvaggi L Jr, Cento R, Proto C, Lanzone A,
et al. Metformin treatment does not affect total leptin levels and free
leptin index in obese patients with polycystic ovary syndrome. Fertil
Steril 2008;89:12736.
433. Mantzoros CS, Dunaif A, Flier JS. Leptin concentrations in the polycys-
tic ovary syndrome. J Clin Endocrinol Metab 1997;82:168791.
434. Zachow RJ, Magofn DA. Direct intraovarian effects of leptin: impair-
ment 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 1997;138:84750.
435. Mantzoros CS, Cramer DW, Liberman RF, Barbieri RL. Predictive
value of serum and follicular uid leptin concentrations during assisted
reproductive cycles in normal women and in women with the polycystic
ovarian syndrome. Hum Reprod 2000;15:53944.
436. Sieminska L, Marek B, Kos-Kudla B, Niedziolka D, Kajdaniuk D,
Nowak M, et al. Serum adiponectin in women with polycystic ovarian
syndrome and its relation to clinical, metabolic and endocrine parame-
ters. J Endocrinol Invest 2004;27:52834.
437. Sepilian V, Nagamani M. Adiponectin levels in women with polycystic
ovary syndrome and severe insulin resistance. J Soc Gynecol Investig
2005;12:12934.
438. Escobar-Morreale HF, Villuendas G, Botella-Carretero JI, Alvarez-
Blasco F, Sanchon R, Luque-Ramirez M, et al. Adiponectin and resistin
in PCOS: a clinical, biochemical and molecular genetic study. Hum
Reprod 2006;21:225765.
439. Orio F Jr, Palomba S, Cascella T, Milan G, Mioni R, Pagano C, et al.
Adiponectin levels in women with polycystic ovary syndrome. J Clin
Endocrinol Metab 2003;88:261923.
440. Spranger J, Mohlig M, Wegewitz U, Ristow M, Pfeiffer AF, Schill T,
et al. Adiponectin is independently associated with insulin sensitivity
in women with polycystic ovary syndrome. Clin Endocrinol (Oxf)
2004;61:73846.
441. Gulcelik NE, Aral Y, Serter R, Demir Y, Culha C. Adiponectin is an in-
dependent determinant of insulin resistance in women with polycystic
ovary syndrome. Gynecol Endocrinol 2006;22:5115.
442. Haap M, Machicao F, Stefan N, Thamer C, Tschritter O, Schnuck F,
et al. Genetic determinants of insulin action in polycystic ovary syn-
drome. Exp Clin Endocrinol Diabetes 2005;113:27581.
443. Zhang N, Shi YH, Hao CF, Gu HF, Li Y, Zhao YR, et al. Association of
45G15G(T/G) and 276(G/T) polymorphisms in the ADIPOQ gene
with polycystic ovary syndrome among Han Chinese women. Eur J
Endocrinol 2008;158:25560.
444. Xita N, Georgiou I, Chatzikyriakidou A, Vounatsou M,
Papassotiriou GP, Papassotiriou I, et al. Effect of adiponectin gene
polymorphisms on circulating adiponectin and insulin resistance in-
dexes in women with polycystic ovary syndrome. Clin Chem
2005;51:41623.
445. Ibanez L, de Zegher F. Ethinylestradiol-drospirenone, utamide-met-
formin, or both for adolescents and women with hyperinsulinemic
hyperandrogenism: opposite effects on adipocytokines and body adi-
posity. J Clin Endocrinol Metab 2004;89:15927.
446. Glintborg D, Frystyk J, Hojlund K, Andersen KK, Henriksen JE,
Hermann AP, et al. Total and high molecular weight (HMW) adiponec-
tin levels and measures of glucose and lipid metabolism following pio-
glitazone treatment in a randomized placebo-controlled study in
polycystic ovary syndrome. Clin Endocrinol (Oxf) 2008;68:16574.
447. Tan BK, Chen J, Digby JE, Keay SD, Kennedy CR, Randeva HS.
Increased visfatin messenger ribonucleic acid and protein levels in
adipose tissue and adipocytes in women with polycystic ovary syn-
drome: parallel increase in plasma visfatin. J Clin Endocrinol Metab
2006;91:50228.
448. Chan TF, Chenb Sc YL, Chen HH, Lee CH, Jong SB, Tsai EM. In-
creased plasma visfatin concentrations in women with polycystic ovary
syndrome. Fertil Steril 2007.
449. Kowalska I, Straczkowski M, Nikolajuk A, Adamska A, Karczewska-
Kupczewska M, Otziomek E, et al. Serum visfatin in relation to insulin
resistance and markers of hyperandrogenism in lean and obese women
with polycystic ovary syndrome. Hum Reprod 2007;22:18249.
450. Moran LJ, Noakes M, Clifton PM, Wittert GA, Belobrajdic DP,
Norman RJ. C-reactive protein before and after weight loss in
824
Bohler et al. Adipose tissue and reproduction in women Vol. 94, No. 3, August 2010
overweight women with and without polycystic ovary syndrome. J Clin
Endocrinol Metab 2007;92:294451.
451. Puder JJ, Varga S, Kraenzlin M, De Geyter C, Keller U, Muller B. Cen-
tral fat excess in polycystic ovary syndrome: relation to low-grade in-
ammation and insulin resistance. J Clin Endocrinol Metab 2005;90:
601421.
452. Gonzalez F, Minium J, Rote NS, Kirwan JP. Hyperglycemia alters tu-
mor necrosis factor-alpha release from mononuclear cells in women
with polycystic ovary syndrome. J Clin Endocrinol Metab 2005;90:
533642.
453. Wu R, Fujii S, Ryan NK, Van der Hoek KH, Jasper MJ, Sini I, et al.
Ovarian leukocyte distribution and cytokine/chemokine mRNA expres-
sion in follicular uid cells in women with polycystic ovary syndrome.
Hum Reprod 2007;22:52735.
454. Amato G, Conte M, Mazziotti G, Lalli E, Vitolo G, Tucker AT, et al.
Serum and follicular uid cytokines in polycystic ovary syndrome dur-
ing stimulated cycles. Obstet Gynecol 2003;101:117782.
455. Glueck CJ, Sieve L, Zhu B, Wang P. Plasminogen activator inhibitor ac-
tivity, 4G5G polymorphism of the plasminogen activator inhibitor 1
gene, and rst-trimester miscarriage in women with polycystic ovary
syndrome. Metabolism 2006;55:34552.
456. Mathur R, Alexander CJ, Yano J, Trivax B, Azziz R. Use of metformin
in polycystic ovary syndrome. Am J Obstet Gynecol 2008;199:
596609.
457. Bagos PG. Plasminogen Activator Inhibitor-1 4G/5G and 5,10-methy-
lene-tetrahydrofolate reductase C677T polymorphisms in Polycystic
Ovary Syndrome. Mol Hum Reprod 2008.
458. Devin JK, Johnson JE, Eren M, Gleaves LA, Bradham WS,
Bloodworth JR Jr, et al. Transgenic overexpression of plasminogen ac-
tivator inhibitor-1 promotes the development of polycystic ovarian
changes in female mice. J Mol Endocrinol 2007;39:916.
459. Kelly CC, Lyall H, Petrie JR, Gould GW, Connell JM, Sattar N. Low
grade chronic inammation in women with polycystic ovarian syn-
drome. J Clin Endocrinol Metab 2001;86:24535.
460. Guzelmeric K, Alkan N, Pirimoglu M, Unal O, Turan C. Chronic in-
ammation and elevated homocysteine levels are associated with in-
creased body mass index in women with polycystic ovary syndrome.
Gynecol Endocrinol 2007;23:50510.
461. Mohlig M, Spranger J, Osterhoff M, Ristow M, Pfeiffer AF, Schill T,
et al. The polycystic ovary syndrome per se is not associated with in-
creased chronic inammation. Eur J Endocrinol 2004;150:52532.
462. Tarkun I, Arslan BC, Canturk Z, Turemen E, Sahin T, Duman C. Endo-
thelial dysfunction in young women with polycystic ovary syndrome:
relationship with insulin resistance and low-grade chronic inamma-
tion. J Clin Endocrinol Metab 2004;89:55926.
463. Shroff R, Kerchner A, Maifeld M, Van Beek EJ, Jagasia D, Dokras A.
Young obese women with polycystic ovary syndrome have evidence of
earlycoronaryatherosclerosis. J ClinEndocrinol Metab2007;92:460914.
Fertility and Sterility

825

You might also like