You are on page 1of 14

Arch Gynecol Obstet (2014) 290:1079–1092

DOI 10.1007/s00404-014-3433-z

REVIEW

Focus on metabolic and nutritional correlates of polycystic ovary


syndrome and update on nutritional management of these critical
phenomena
Mariangela Rondanelli • Simone Perna •
Milena Faliva • Francesca Monteferrario •

Erica Repaci • Francesca Allieri

Received: 14 February 2014 / Accepted: 22 August 2014 / Published online: 9 September 2014
 Springer-Verlag Berlin Heidelberg 2014

Abstract cornerstone in the treatment of PCOS patients. The opti-


Introduction Polycystic ovary syndrome (PCOS) is mum diet therapy for the PCOS treatment must aim at
associated with numerous metabolic morbidities (insulin achieving specific metabolic goals, such as IR improve-
resistance (IR), central obesity) and various nutritional ment, adipokines secretion and reproductive function.
abnormalities (vitamin D deficit, mineral milieu alterations, These goals must be reached through: accession of the
omega6/omega3 PUFA ratio unbalance). patient to hypocaloric dietary program aimed at achieving
Methods We performed a systematic literature review to and/or maintaining body weight; limiting the consumption
evaluate the till-now evidence regarding: (1) the metabolic of sugar and refined carbohydrates, preferring those with
and nutritional correlates of PCOS; (2) the optimum diet lower glycemic index; dividing the food intake in small and
therapy for the treatment of these abnormalities. This frequent meals, with high caloric intake at breakfast;
review included 127 eligible studies. increasing their intake of fish (4 times/week) or taking
Results In addition to the well-recognized link between omega3 PUFA supplements; taking Vitamin D and chro-
PCOS and IR, the recent literature underlines that in PCOS mium supplementation, if there are low serum levels.
there is an unbalance in adipokines (adiponectin, leptin, Conclusion Lifestyle intervention remains the optimal
visfatin) production and in omega6/omega3 PUFA ratio. treatment strategy for PCOS women. A relatively small
Given the detrimental effect of overweight on these met- weight loss (5 %) can improve IR, hyperandrogenism,
abolic abnormalities, a change in the lifestyle must be the menstrual function, fertility.

Keywords Polycystic ovary syndrome  Dietary


M. Rondanelli (&)  S. Perna  M. Faliva  F. Monteferrario  management  Dietary supplement  Overweight 
E. Repaci  F. Allieri Adipokines  Insulin resistance
Department of Public Health, Experimental and Forensic
Medicine, Section of Human Nutrition, Endocrinology and
Nutrition Unit, University of Pavia, Azienda di Servizi alla
Persona, Pavia, Italy Introduction
e-mail: mariangela.rondanelli@unipv.it
S. Perna Polycystic ovary syndrome (PCOS) is one of the most
e-mail: simoneperna@hotmail.it common female endocrine disorders with a prevalence of
M. Faliva approximately 5–10 % in women of reproductive age [1,
e-mail: milo.milena87@hotmail.it 2], and women with PCOS are at increased risk of repro-
F. Monteferrario ductive abnormalities [3]. PCOS is a heterogeneous syn-
e-mail: francescamontef@libero.it drome characterized by increased ovarian and adrenal
E. Repaci androgen secretion with hyperandrogenic symptoms, such
e-mail: ericarepaci@gmail.com as hirsutism, acne and/or alopecia, menstrual irregularity,
F. Allieri anovulation and infertility [3]. A positive diagnosis is made
e-mail: francesca.allieri@gmail.com when the patient has any two of the three features: oligo-

123
1080 Arch Gynecol Obstet (2014) 290:1079–1092

ovulation/anovulation, clinical and/or biochemical signs of published in the last 20 years; humans; languages: English;
hyperandrogenism, and polycystic ovaries on ultrasound (e) manual search performed by the senior researchers
examination, according to Rotterdam ESHRE/ASRM- experienced in clinical nutrition through the revision of
sponsored PCOS consensus workshop [4]. Besides the reviews and individual articles on microbiota in elderly
endocrinological abnormalities observed in this syndrome, published in journals qualified in the Index Medicus. (4)
profound metabolic alterations have also been documented: Analysis and presentation of the outcomes: the data
insulin resistance (IR), hyperinsulinemia, central obesity extrapolated from the revised studies were collocated in
and metabolic syndrome (MeTS) [5], which increase risks tables; in particular, for each study we specified: the author,
for developing type 2 diabetes (T2DM), cardiovascular the name of the journal where the study was published and
disorders, hypertension, atherosclerosis and dyslipidemia year of publication, study characteristics. (5) The analysis
[6]. Hyperinsulinemia and IR stimulate ovarian androgen was carried out in the form of a narrative review of the
production [7, 8] and decrease serum sex hormone-binding reports. At the beginning of each section, the keywords
globulin (SHBG) concentrations [9, 10], leading to considered and the kind of studies chosen have been
increased levels of circulating free testosterone. This reported. Suitable for the systematic review were studies of
association between IR and ovarian hyperandrogenism any design, which considered women with PCOS diag-
suggests that insulin directly influences ovarian function nosed consistently to The Rotterdam ESHRE/ASRM-
[11]. Women with PCOS have increased ovarian cyto- Sponsored PCOS Consensus Workshop Group, 2004 [4].
chrome P450c17a activity, as evidenced by an elevated
serum 17OHP (17a-hydroxyprogesterone) in response to
stimulation by GnRH (gonadotropin-releasing hormone) Results
agonists [12]; ovarian cytochrome P450c17a appears to be
stimulated by insulin in PCOS. Metabolic correlates in PCOS
Given this background, the aim of the present systematic
review is to summarize the state of the art according to the Obesity and insulin resistance
extant literature about two topics: (1) the metabolic and
nutritional correlates of PCOS and (2) the optimum diet This research has been carried out based on the keywords:
therapy for the treatment of these abnormalities. ‘‘Polycystic Ovary Syndrome’’ AND ‘‘obesity’’ AND
‘‘insulin resistance’’; 1,248 articles were sourced. Among
them, 22 retrospective studies, 12 reviews, 7 cross-over
Methods studies, 3 double-blind studies, 2 retrospective studies and
1 cohort study have been selected and discussed.
The present systematic review was performed following Obesity is not necessarily a defect intrinsic to PCOS [14],
the steps by Egger et al. [13] as follows: (1) configuration but is a significant characteristic of this syndrome [4, 15, 16],
of a working group: three operators skilled in endocrinol- with a pooled estimated prevalence of 49 %, as shown in a
ogy and clinical nutrition, of whom one acting as a meth- recent meta-analysis [17] and, specifically central obesity,
odological operator and two participating as clinical worsens the phenotype [18]. Adiposity-dependent IR is
operators. (2) Formulation of the revision question on the linked with PCOS, given that the prevalence of obesity
basis of considerations made in the abstract: ‘‘the state of among women with PCOS is higher than that of age-matched
the art on metabolic and nutritional correlates of PCOS and healthy women without this syndrome, as determined at
their nutritional treatment’’. (3) Identification of relevant referral centers [13]. IR occurs in approximately 50–70 % of
studies: a research strategy was planned, on PubMed women with PCOS and in 95 % of obese women with PCOS
[Public Medline run by the National Center of Biotech- [19, 20]. The degree of IR in PCOS is greater than that pre-
nology Information (NCBI) of the National Library of dicted by the Body Mass Index (BMI-ratio between weight
Medicine of Bathesda (USA)], as follows: (a) definition of and the square of the height), although 40–50 % of women
the key words (Polycystic Ovary Syndrome, dietary man- with PCOS are not obese [21–23]. The prevalence of MetS
agement, dietary supplement, overweight, adipokines, and IR varies between the different female hyperandrogenic
insulin resistance), allowing the definition of the interest phenotypes [24]. In another study [25], non-hyperandro-
field of the documents to be searched, grouped in inverted genic anovulatory cases of PCOS were found to show only
commas (‘‘…’’) and used separately or in combination; marginal risk. Given this well-documented association
(b) use of: the Boolean (a data type with only two possible between PCOS and IR, insulin-sensitizing agents have been
values: true or false) AND operator that allows the estab- used in the therapy of PCOS. As shown in a 2003 Cochrane,
lishments of logical relations among concepts; (c) research over 50 intervention studies have demonstrated a positive
modalities: advanced search; (d) limits: time limits: papers effect of metformin on both reproductive and metabolic

123
Arch Gynecol Obstet (2014) 290:1079–1092 1081

aspects of PCOS [26]. It is peculiar that numerous random- In summary, IR is a relative common finding in PCOS
ized controlled trials (RCT) showed this positive effect of women, independent of obesity. Assessment of insulin
metformin therapy on endocrine and metabolic variables, not sensitivity may allow identification of patients at higher
only in obese or insulin-resistant PCOS patients, but even in risk for metabolic sequelae and allow the selection of
lean and insulin-sensitive women [27–31]. A recent con- patients most likely to respond to treatment with insulin-
sensus on women’s health aspects of PCOS [32] has defined sensitizing drugs.
that metformin treatment is indicated in those with impaired
glucose tolerance (IGT) who do not respond adequately to Adipokines unbalance in PCOS
calorie restriction and lifestyle changes. As regards the
relationship between IR and menstrual disturbance, PCOS This research has been carried out based on the keywords:
patients with isolated primary or secondary amenorrhea or ‘‘Polycystic Ovary Syndrome’’ AND ‘‘adipokines’’ AND
oligomenorrhea and patients with secondary amenorrhea ‘‘insulin resistance’’ 220 articles were sourced. Among
alternating with regular menstrual cycles had more pro- these, 9 reviews, 29 prospective studies, 17 cross-over
nounced IR than women with regular menstrual cycles [33]. studies and 3 single-blinded studies have been selected and
In contrast, patients with oligomenorrhea alternating with discussed.
secondary amenorrhea or with regular menstrual cycles did Paracrine dysregulation of adipokine production by
not differ in markers of IR from women with regular men- macrophage-secreted cytokines in PCOS favors develop-
strual cycles. Two previous studies reported that patients ment of IR [55]. Considering the frequent clustering of
with PCOS and oligomenorrhea or amenorrhea had more obesity and IR-associated disorders in PCOS patients,
severe IR than patients with PCOS and regular cycles [34, adipokines have been proposed to play a role in the path-
35]. In contrast, in two other more recent studies that ana- ogenesis of PCOS [56]. Adipokines may thus serve as an
lyzed patients with amenorrhea separately from patients with endocrine link between obesity and PCOS [57]. Moreover,
oligomenorrhea, only the former had more pronounced IR a possible role for cytokine products of fat (adipokines) as
than patients with regular menses [36, 37]. As regards a link between reproductive and metabolic abnormalities
intraovarian hyperinsulinemic pathways, hyperinsulinemia has been mooted [58, 59]. Abnormal serum levels of var-
may have preferentially impaired oocyte developmental ious adipokines in PCOS have been reported in the litera-
competence, resulting in reduced rates of fertilization, ture [60].
embryonic development and implantation in PCOS patients
with obesity [38, 39]. Data from in vitro cell culture models Adiponectin Adiponectin is considered to be one of the
suggest that co-incubation of insulin and follicle-stimulating most important adipokines in human physiology and differs
hormone (FSH) with mouse [40] and bovine [41] oocytes from most other adipokines by having a protective effect
promotes FSH-induced up-regulation of GC (granulose on development of obesity. The disruption of adiponectin
cells) LH (luteinizing hormone) receptor mRNA expression and its receptors is a major mechanism that links metabolic
[42–44], inhibiting FSH stimulation of aromatase activity and reproductive dysfunction in women with polycystic
[41], thus reducing the percentage of fertilized oocytes that ovary syndrome [61]. Serum adiponectin levels are
develop into blastocysts [45, 40]. Insulin may induce local decreased in PCOS patients [56, 62–67], yet this result may
androgen production, which results in oocytes of lower be explained by the concurrence of obesity [64], IR [65,
quality, post-maturity [46]. At the molecular level, insulin 66] and/or IGT [67] in these women, whereas other
binds to its receptor, localized on GC and theca cells, and researches reported that serum adiponectin concentrations
oocytes, to stimulate follicle recruitment [45, 47], conse- did not seem to differ between PCOS and controls [68]. A
quently altering expression of multiple genes involved in meta-analysis [69] revealed that serum adiponectin levels
meiotic/mitotic spindle dynamics and centrosome function are lower in women with PCOS compared with BMI-
in PCOS oocytes [48]. This indicates that insulin may be an matched healthy controls and that the more insulin-resis-
important mediator of oocyte developmental competence via tant women with PCOS recruited, the lower serum adipo-
a ligand-receptor regulating system [45]. nectin levels were found. The effects of adiponectin are
Chronic low-grade inflammation has emerged as a key mediated by at least two main receptors that have been
contributor to the pathogenesis of PCOS and emerging data identified recently [70]: adiponectin receptor-1 (AdipoR1)
suggest that this chronic low-grade inflammation underpins that is abundantly expressed in skeletal muscle, and
the development of metabolic aberration, such as IR, and adiponectin receptor-2 (AdipoR2) that is predominantly
ovarian dysfunction in PCOS [49, 50]. Most importantly, expressed in the liver. PCOS patients had lower baseline
there is a strong association between hyperandrogenism serum adiponectin and AdipoR1 compared to healthy
and inflammation in PCOS [51–54]. women after controlling for BMI [71].

123
1082 Arch Gynecol Obstet (2014) 290:1079–1092

Leptin In humans, leptin acts as an afferent satiety signal, orlistat results in a significant reduction in serum leptin
regulating appetite and weight and is mainly produced in levels in both patients with PCOS and controls, and this
the adipocyte of white adipose tissue [72]. However, recent decrease is comparable in the two groups. On the other
studies have confirmed that other tissues also express lep- hand, serum adiponectin, resistin, and visfatin levels are
tin, including placenta, ovaries, skeletal muscle, stomach, not affected by weight loss. Previous short-term
pituitary, and liver [73]. Leptin appears to act as an (1–8 weeks) studies on the effects of hypocaloric diet in
endocrine and paracrine factor for the regulation of puberty PCOS patients showed that serum leptin levels decline in
and reproduction; it affects maternal, placental and fetal parallel with weight loss [98, 100, 101]. This finding is
function, modifies insulin sensitivity in the muscle or liver, somewhat expected because the degree of obesity is the
prevents ectopic lipid deposition, and links the endocrine major determinant of serum leptin levels in patients with
and immune systems [74, 75]. Leptin levels have been PCOS [81, 102, 103].
reported to be increased in women with PCOS in com- In summary, our current understanding of the role for
parison to weight-matched controls [76–79] although this adipokines in PCOS is far from complete. Moreover, there
was not supported by many other studies [80–83]. Wang is a paucity of data on the effects of weight loss on serum
also reported significantly higher mRNA expression of adipokine levels in overweight/obese patients with PCOS.
leptin in subcutaneous adipose tissue of PCOS patients
compared with controls [81–84]. No significant difference Nutritional correlates in PCOS
was found in circulating leptin levels between the ovula-
tory and anovulatory PCOS patients either [83]. Further- Mineral deficiency and its treatment
more, a recent study showed that there was no effect of
PCOS on either adipose leptin expression or plasma leptin This research has been carried out based on the keywords:
levels [85]. As regards relationship with IR, no significant ‘‘Polycystic Ovary Syndrome’’ AND ‘‘mineral’’; 59 articles
differences were observed in serum leptin or leptin receptor were sourced. Among them, 2 reviews, 11 prospective
(LEPR) levels between PCOS IR and PCOS non-IR studies and 5 cross-over studies have been selected and
women [84]. However, Yildizhan [79] observed an asso- discussed.
ciation between serum leptin levels with IR in young
women with PCOS. Magnesium Decreased magnesium level was reported in
women with increased testosterone levels [104] and/or IR
Visfatin Visfatin is a more recently described adipokine [105]. A prospective study demonstrated that the PCOS
that also appears to have insulin-like effects [86]. It has women with IR exhibited significantly lower serum levels
previously been reported that the gene expression and of magnesium than controls and PCOS women without IR
circulating levels of visfatin were increased in women with and that circulating serum magnesium significantly corre-
PCOS compared with age- and BMI-matched controls [87– lated with fasting insulin levels [106]. Hypomagnesemia is
94]. However, several recently published studies did not associated with T2DM and MetS [107].
find a difference in plasma or serum visfatin levels between
patients with PCOS and control groups [95–97] Moreover, Copper Two recent prospective studies showed that
Chan [88] did not observe any correlation between visfatin higher serum copper level is observed in PCOS patients
concentrations and testosterone, insulin, and LH levels in than in the controls [106, 108], which significantly
either PCOS or control groups. A positive correlation, increases with the association of insulin resistance [106].
however, was found between plasma visfatin concentra- Moreover, copper, in addition to its enzymatic roles, sim-
tion, fasting insulin, and homeostasis model assessment ilarly can induce oxidative stress by catalyzing the for-
(HOMA)-IR, as reported by Tan [87]. mation of reactive oxygen species and decreasing
In summary, at present, any indication to measure rou- glutathione levels [77]. Many studies conferred the role of
tinely circulating levels of such adipokines is present in the increased oxidative stress resulting from high generation of
literature, but this topic represents an intriguing topic of reactive oxygen species (ROS) in the pathogenesis of
research in PCOS women. PCOS [53, 109].

Weight loss and adipokines Chrome Chrome picolinate consists of trivalent chro-
mium (Cr3?), an extremely safe [110] and highly tolerable
There is a paucity of data on the effects of weight loss on trace mineral that is present in the normal diet [111],
serum adipokine levels in overweight/obese patients with complexed to picolinic acid to enhance gut absorption.
PCOS [98, 99]. A recent prospective study by Spanos [94] After cleavage of picolinic acid, Cr3? is transported by
demonstrated that weight loss (about 12 %) by diet and transferrin and later, by chromodulin, its binding protein.

123
Arch Gynecol Obstet (2014) 290:1079–1092 1083

Table 1 Metabolic or nutritional correlates in PCOS and recommended treatment


Metabolic or Recommended treatment References Type of study Results after recommended treatment
nutritional
correlates

Insulin Intake of foods with low Graff [151] Cross-sectional study Dietary GI is increased in the classic
resistance glycemic index PCOS phenotype and associated
with a less favorable
anthropometric and metabolic
profile
Mehrabani Single blind clinical trials: hypocaloric Both hypocaloric diets significantly
[150] diet (CHCD) (15 % of daily energy led to reduced body weight and
from protein) and a modified androgen levels in these two groups
hypocaloric diet (MHCD) with a of women with PCOS. The
high-protein, low-glycemic load combination of high-protein and
(30 % of daily energy from protein low-glycemic-load foods in a
plus low-glycemic-load foods) modified diet caused a significant
increase in insulin sensitivity and a
decrease in hsCRP level when
compared with a conventional diet
Overweight or Low caloric diet with high Moran Systematic review Greater weight loss for a
obesity protein and low glycemic [148] monounsaturated fat-enriched diet;
load improved menstrual regularity for a
low-glycemic index diet; increased
free androgen index for a high-
carbohydrate diet; greater
reductions in insulin resistance,
fibrinogen, total, and high-density
lipoprotein cholesterol for a low-
carbohydrate or low-glycemic
index diet; improved quality of life
for a low-glycemic index diet; and
improved depression and self-
esteem for a high-protein diet.
Weight loss improved the
presentation of PCOS regardless of
dietary composition in the majority
of studies. Weight loss should be
targeted in all overweight women
with PCOS through reducing
caloric intake in the setting of
adequate nutritional intake and
healthy food choices irrespective of
diet composition
Alterations in Balanced low-calorie diet Rafraf Intervention study: 720 mg/day Increased serum adiponectin levels,
the secretion with adequate timing of [135] eicosapentaenoic acid and 480 mg/ insulin resistance and lipid profile
of adipokines intake of meals and day docosahexaenoic acid
and cytokines supplements with omega3 Leidy Intervention study: two isocaloric high caloric intake at breakfast with
fatty acids [152] (*1,800 kcal) maintenance diets reduced intake at dinner results in
with different meal timing improved insulin sensitivity indices
distribution: a BF (breakfast diet) and reduced cytochrome P450c17a
(980 kcal breakfast, 640 kcal lunch activity, which ameliorates
and 190 kcal dinner) or a D (dinner hyperandrogenism and improves
diet) group (190 kcal breakfast, ovulation rate
640 kcal lunch and 980 kcal dinner)
Low blood Intake of vitamin D Pal [129] Intervention study: 8,533 UI/day of Androgen and Blood Pressure
levels of 25 supplement vitamin D profiles improved
hydroxy Wehr [130] Intervention study: 20,000 IU Vitamin D treatment might improve
vitamin D cholecalciferol weekly glucose metabolism and menstrual
frequency

123
1084 Arch Gynecol Obstet (2014) 290:1079–1092

Table 1 continued
Metabolic or Recommended treatment References Type of study Results after recommended treatment
nutritional
correlates

Alteration of Intake of chromium picolinate Lydic Intervention study: dietary supplement Improved glucose disposal
the milieu of supplement [117] with 1,000 lg as CrP/day
trace
minerals
GI glycemic index, hsCRP high-sensitivity C-reactive protein

This complex binds the insulin receptor [111]. Active the pathogenesis of insulin resistance and the metabolic
glucose transport is enhanced through tyrosine kinase syndrome in PCOS [119, 120]. Vitamin D deficiency has
phosphorylation, without inhibition of phosphotyrosine been shown to be associated with impaired glucose clear-
phosphatase [84]. Chrome deficiency as a cause of glucose ance and insulin secretion in both human [121] and animal
intolerance was recognized first in 1977 [112]. Chromium models [122]. 25-Hydroxyvitamin D (25-OH-VD) is pos-
appeared to improve the effects of insulin and worked at itively correlated with the insulin sensitivity and negatively
the level of the cell membrane [113]. The interaction with beta-cell function [123]. A recent review by Thomson
between chromium and insulin has been elucidated by the et al. [124] suggests that there is an association between
discovery of low-molecular weight chromium-binding vitamin D status and hormonal and metabolic dysfunctions
substance, which binds chromium and the insulin receptor, in PCOS. The underlying mechanism is, however, yet to be
activating the insulin receptor’s kinase activity [114, 115]. elucidated. One of the plausible mechanisms may be the
Morris and colleagues [116] showed diminished plasma dysregulation of the complex mechanism that regulated
Cr3? levels in T2DM subjects, suggesting that Cr3? losses ovarian apoptosis [125]. Due to its immunomodulatory
diminish insulin sensitivity and worsen T2DM. As dem- functions, hypovitaminosis D might induce a higher
onstrated, Cr3? (1,000 lg as chrome picolinate) improved inflammatory response, which is again associated with IR
glucose disposal in five obese PCOS subjects after [126, 127]. An interesting study on obese and non-obese
2 months [117]. PCOS patients [128] demonstrated that low serum con-
In summary, some studies suggest that PCOS women centrations of 25-OH-VD have been associated with higher
have mostly insufficient magnesium levels, so magnesium BMI and total cholesterol values. This study also confirmed
replacement therapy may have a beneficial effect on the association between abdominal obesity, hyperandrog-
ameliorating the IR. Also, CrP supplementation may enism, and IR. A recent single-arm, open-label trial per-
improve IR in PCOS, as shown in the literature (Table 1). formed for 3 months in 23 PCOS women supplemented
Higher serum copper level is observed in PCOS patients with Vitamin D and calcium suggests potential therapeutic
than the controls, so RCT must be performed to evaluate if benefits of these nutrients supplementation in ameliorating
therapy with copper chelators may be useful in PCOS the hormonal milieu (significant reduction in total testos-
women with elevated blood levels of copper. terone) and PCOS-related sequelae in women deficient in
vitamin D [129]. Another intervention study by Wehr [130]
Nutritional correlates in PCOS: vitamin D deficiency conducted in fifty-seven PCOS women, which received
and its treatment 20,000 IU cholecalciferol weekly for 24 weeks, suggested
that vitamin D treatment might improve glucose metabo-
This research has been carried out based on the keywords: lism and menstrual frequency in PCOS women. The posi-
‘‘Polycystic Ovary Syndrome’’ AND ‘‘vitamin D’’; 59 tive effect on glucose metabolism is in according to the
articles were sourced. Among them, 6 reviews, 7 pro- study by Selimoglu [131] that showed improvement in
spective studies and 1 cross-over study have been selected HOMA indices within 3 weeks of a single oral mega dose
and discussed. of 300,000 IU D3 in a pilot study on 11 PCOS women.
Vitamin D is rapidly emerging as an important bio- In summary, there are data that suggest pathophysio-
molecule with effects beyond the already established role logical relevance of vitamin D deficiency for PCOS. Some
in calcium and phosphorus metabolism. The various studies demonstrated that women with PCOS have mostly
pathways include apoptotic pathway, insulin metabolism, insufficient vitamin D levels, and vitamin D replacement
growth and differentiation [118]. There is some evidence therapy may have a beneficial effect on ameliorating the
suggesting that vitamin D deficiency might be involved in hormonal status (significant reduction in total testosterone

123
Arch Gynecol Obstet (2014) 290:1079–1092 1085

and menstrual disturbances) and PCOS-related sequelae may benefit from LSM through adiposity reduction [140],
(improvement in IR) in women deficient in vitamin D improved ovulatory function [141] and reduction in overall
(Table 1). cardiovascular risk [142]. Previous observational studies
demonstrated that LSM can be associated with clinical
Nutritional correlates in PCOS: imbalance in the n-6/n-3 improvement in PCOS, as shown in Table 2 Kiddy and
PUFA ratio and its treatment colleagues [143] demonstrated that moderate weight loss
(equal to 5 %) during long-term calorie restriction
This research has been carried out based on the keywords: (6 months) is associated with a marked clinical improve-
‘‘Polycystic Ovary Syndrome’’ AND ‘‘plasma fatty acids’’; ment in menstrual function and fertility. Clark et al. [144]
12 articles were sourced. Among them, 1 review, 2 pro- demonstrated in a retrospective study that weight loss (the
spective studies, 2 cross-over studies and 2 double-blind average weight loss in the PCOS group was 6.3 kg, during
studies have been selected and discussed. 6 months of LSM program) is associated with improve-
An interesting study reported that increased dietary ment in ovulation, pregnancy outcome, self-esteem and
polyunsaturated fatty acids (PUFA) intake could be asso- endocrine parameters in women who are infertile and
ciated with improved endocrine and metabolic character- overweight. Thomson demonstrated that in overweight and
istics in women with PCOS [132]. Eicosapentaenoic acid obese women with PCOS and reproductive dysfunction, a
(EPA) and docosahexaenoic acid (DHA) are n-3 long-chain 20-week weight loss intervention (with weight loss equal to
polyunsaturated fatty acids (n-3 LC PUFA) found primarily 9 kg) resulted in significant reduction in fasting insulin,
in fatty fish. In current diets, n-6 fatty acids are predomi- homeostasis model assessment, testosterone and Free
nant PUFA and imbalance in the n-6/n-3 PUFA ratio may Androgen Index (FAI), an increase in sex hormone-binding
be related to chronic disease [133]. Many studies, as well globulin (SHBG) and in improvements in reproductive
documented in the literature, have shown increasing evi- function, but no change in anti-Mullerian hormone levels
dence for antiatherogenic and anti-inflammatory effects of [145]. A significant correlation between the weight
fish oil supplementation and it is conceivable that dietary reduction (5–6 kg lost or lost 5–10 % of starting body
PUFA might also have benefit effects on glycemic control weight is highly recommended) and the improvement in
and lipid profile [133–135]. Moreover, it seems that the metabolic parameters could be attributed to decreased
positive effect of EPA and DHA on metabolic parameters insulin resistance or to other related factors. Clinicians
could be partially due to enhancement of adiponectin prescribing LSM interventions must consider the patient’s
production [114, 136]. A recent RCT has demonstrated that capacity to sustain diet and exercise adherence and weight
a supplementation for 8 weeks with 720 mg/day eicosa- maintenance over time in order for the clinical benefits on
pentaenoic acid and 480 mg/day docosahexaenoic acid PCOS to continue [146].
increases serum adiponectin levels, insulin resistance and In summary, lifestyle modification (dietary intervention
lipid profile in a group of overweight or obese PCOS and increased physical activity) remains the optimal
patients [135, 137]. treatment strategy for overweight/obese women with
In conclusion, the data currently available, although the PCOS. The studies currently available showed that a rel-
research carried out are few, are very promising and show atively small weight loss (equal to 5–10 %), achieved with
that an adequate intake of omega3 fatty acids may be long-term caloric restrictions (5–6 months), can improve
useful in the control and prevention of metabolic compli- IR and hyperandrogenism, menstrual function and fertility,
cations of PCOS patient, as shown in Table 1. as reported in 2011 Cochrane by Moran [147]. Weight loss
can also improve long-term metabolic health.
Management of metabolic and nutritional correlates
of PCOS: lifestyle modification Dietary composition and meal timing

This research has been carried out based on the keywords: The most important determinant of dietary intervention for
‘‘Polycystic Ovary Syndrome’’ AND ‘‘lifestyle’’ AND weight loss in PCOS women is energy balance, though for
‘‘diet’’; 101 articles were sourced. Among them, 7 reviews, many other reasons various types of diet have been pro-
4 prospective studies, 6 cross-over studies 2 single-blinded posed and tested, as shown in a recent review by Moran
studies have been selected and discussed. [148]. This review and meta-analysis reported that weight
Lifestyle modification programs (LSM), comprised of loss should be targeted in all overweight women with
diet and physical activity, are recommended for in high risk PCOS through reducing caloric intake in the setting of
patients (prediabetic) to delay the onset of T2DM [138, adequate nutritional intake and healthy food choices, irre-
139], one of the most serious complications of PCOS. spective of diet composition. Table 1 summarizes the
Additionally, overweight and obese women with PCOS studies investigating the optimum diet therapy and the

123
1086 Arch Gynecol Obstet (2014) 290:1079–1092

optimum dietary supplementation for the treatment of In summary, a weight loss of 5–10 % of original body
PCOS. A crossover study conducted by Douglas demon- weight improves insulin sensitivity and short-term repro-
strated that a moderate low-carbohydrate diet (43 %), ductive fitness in PCOS overweight women and is addi-
conducted for 16 days, reduced the fasting and postchal- tionally crucial for improving short- and long-term
lenge insulin concentrations among PCOS women, which, metabolic health.
over time, may improve reproductive/endocrine outcomes
[149]. A following study conducted by Marsh in 96 PCOS
women confirmed this result: with only modest weight loss Discussion and conclusion
(4–5 % of body weight), the low-carbohydrate diet pro-
vided a threefold greater improvement in whole-body The adipose tissue of women with PCOS is characterized
insulin sensitivity, improved menstrual regularity and bet- by hypertrophic adipocytes and impairments in insulin
ter emotion scores, compared to conventional hypocaloric action. Amenorrhea is associated with more pronounced IR
diet. By contrast, both the low-carbohydrate and the con- and hyperandrogenemia in patients with PCOS. Therefore,
ventional diets led to similar improvements or changes in the type of menstrual cycle abnormality might represent a
blood lipid and androgenic hormones concentrations, useful tool for identifying a more severe metabolic profile
markers of inflammation, and other measures of quality of in PCOS. The expression and secretion of a wide variety of
life. A further recent 12-week single-blind clinical trial, in adipokines implicated in IR are also altered in PCOS.
which a total of 60 PCOS overweight and obese women Collectively, the available data indicate that adipose tissue
were recruited and randomly assigned to (1) a conventional dysfunction plays a pivotal role in the metabolic abnor-
hypocaloric diet (CHCD) (15 % of daily energy from malities observed in PCOS. Whether these abnormalities
protein) and (2) a modified hypocaloric diet (MHCD) with are primary or secondary to hyperandrogenism or other
a high-protein, low-glycemic load (30 % of daily energy abnormalities in PCOS is not yet known. However, a
from protein plus low-glycemic-load foods), demonstrated comprehensive evaluation of the circulating levels of sev-
that both hypocaloric diets significantly led to reduced eral adipokines in patients with PCOS has not been per-
body weight and androgen levels, but the combination of formed to date and the results of the studies that have
high-protein and low-glycemic-load foods in a modified evaluated how the weight loss has resulted in a change of
diet caused a significant increase in insulin sensitivity and a the levels of adipokines, are very few, conducted for short
decrease in high-sensitivity C-reactive protein level, when periods and difficult to interpret. So, new studies in this
compared with a conventional diet [150]. However, in a interesting topic must be conducted. There is well-estab-
recent cross-sectional study, Graff [151] demonstrated that lished evidence for the detrimental effect of overweight
the intake of food with high glycemic index is increased in and obesity in PCOS women. A weight loss of 5–10 % of
the classic PCOS phenotype than in control women and is original body weight improves insulin sensitivity and short-
associated with a less favorable anthropometric and meta- term reproductive activities in PCOS overweight women
bolic profile. and is additionally crucial for improving short- and long-
Concerning protein supplementation, in PCOS patients a term metabolic health. This can be accomplished through
2-month hypocaloric diet, associated with a 240-kcal sup- lifestyle intervention, with the overall aim of energy
plement containing whey protein, reduced body weight, fat expenditure exceeding energy intake over a short or med-
mass, serum cholesterol, and apoprotein B more than the ium period. Following weight loss, metabolic and endo-
same hypocaloric diet associated with a 240-kcal supplement crine variables were improved to a level similar to that of
containing simple sugars instead of whey protein [98]. BMI-matched non-PCOS controls. However, even if the
As regards the meal timing, recently, Jakubowicz and studies performed on specific diets are heterogeneous and
colleagues found in 60 lean PCOS women that high caloric final conclusions can not be drown, it appears useful to
intake at breakfast than high caloric intake at dinner led to limit the consumption of sugar and refined carbohydrates,
greater weight loss, improved glucose metabolism and with preference of products with low glycemic index in
insulin sensitivity indices, which lead to the reduction of order to favor metabolic improvements. In addition, it can
ovarian P450c17a activity and, as a result, to decreased be suggested to divide the food intake in small and frequent
ovarian testosterone synthesis [152]. Thus, the improve- meals, with high caloric intake at breakfast. Moreover, the
ment of insulin resistance indices found after high caloric majority of studies were also of short- to medium-term
intake at breakfast suggests that the high caloric intake in duration, with only one 12-month study. Because the long-
the morning might represent a schedule more synchronized term sustainability of anthropometric and metabolic
with the circadian pacemaker. This clock resetting could be improvements is more important than acute changes, there
protective also against the disruption in hormone secretion is a significant gap in the literature. Additional research is
reported in PCOS patients [153, 154]. warranted that assesses the effect of dietary composition on

123
Arch Gynecol Obstet (2014) 290:1079–1092 1087

Table 2 Effects of LMS program on metabolic correlates of PCOS (modified from Domecq et al. [155])
Author and Age Follow- n Groups BMI Characteristics Main conclusions
year up
(months)

Brown, 2009 18–50 years 6 11 LMS 38 (median) Ex Prog: moderate to intensive, Improved lipoprotein profiles in
[156] 228 min/week for 5–6 m PCOS women
31 C 31 (median) No intervention
Guzik, 1994 32 mean 5 12 LMS 38 (mean) Diet (1,200–1,000 kcal/day ? Ex WL in ob PCOS women reduces
[157] age Prog. (walking 2 miles/day, I and n-SHBGT conc., may
5 days/week) for 3 m restore ovulation
C No intervention
Hoeger, 2004 28 mean 12 27 LMS 39 (mean) Diet (500–1,000 kcal/day) ? Ex WL may restore ovulation in
[158] age Prog. (150 min/week for) 12 m PCOS ob. women
Met 1,700 mg/dia for 12 m
C No intervention
Hoeger, 2008 12–18 years 6 months 32 LMS [25 (above Diet ? Ex Prog. ? Behav. Enhancement of androgen and
[159] the 95th Int. ? oral contraceptives for reduction of SHBGT in ob.
percentile) 6m adolescents PCOS women
Met 1,700 mg/day ? LSM ? oral Reduction of CA, total T and
contraceptives for 6 m increased HDL, no WL
Control Placebo for 6 m
Karimzadeh, 27 mean 6 25 LMS 27 (mean) Diet (delta 500 kcal/day) ? Ex LMS improved LP in PCOS
2010 [160] age Prog (120 min/day from 3 to 5 women
times/week) for 6 m
5 Met 1,500 mg/dia
Palomba, 18–35 years 1.5 64 LMS 32 (mean) Diet ? Ex Prog (3 times/week) for Increased ovulation in ob. PCOS
2010 [161] 1.5 m women resistant to clomiphene
Control No intervention
Qublan, 2007 31 mean 3 46 LMS 32 (mean) Diet (120–1,400 kcal/day) for 3 m Ameliorated hyperinsulinemia
[162] age and hyperandrogenemia
Met 1,700 mg/day for 3 m Improved clinical parameters and
ameliorated reproductive
function in PCOS ob. women
Stener- 30 mean 4 84 LMS 27 (mean) Ex Prog. (30–45 min. 3 day/ Improved reproductive function
Victorin, age week ? 1 day of reinforcement) in PCOS women
2009 [163] for 4 m
Control No intervention
Vigorito, 2 mean age 3 months 90 LMS 29 (mean) Ex. Prog. (30 min/day 3 times/ Improved oxygen peak
2007 [164] week) for 3 m consumption, reduced BMI,
C-reactive protein and ISI
Control No intervention
When not specifically indicated, the value of the age means the mean age; follow-up period is expressed in months after the end of the study;
when not specifically indicated, the value of the BMI means the mean of BMI values
BMI Body Max Index (ratio between weight and the square of the height), C control group, CA central adiposity, Ex Prog exercise program,
I insulin, Diet hypocaloric diet, ISI insulin sensitivity index, LMS life modification program, LP lipid profile, PCOS polycystic ovary syndrome,
T testosterone, m months, Met metformin, ob. obese, SHGT conc. SHGT concentrations, WL weight loss, Y years

weight maintenance or prevention of weight gain in both Finally, as regards minerals, magnesium supplementation
lean and overweight women with PCOS, and the effect of and use of copper chelators may be promising topics.
dietary composition on reproductive, metabolic, and psy- Moreover, also a supplementation of omega3 PUFA (DHA
chological parameters independent of changes in weight in and EPA) improves IR and counteracts the damage derived
PCOS lean women. Finally, a supplementation of vitamin from oxidative stress.
D and chrome improve glycemic control and IR in PCOS Further research is needed, including high-quality, long-
woman, but at the present the specific dose for supple- term RCTs that assess a range of diet compositions,
mentation of these nutrients is not yet well established. including low GI, and use of dietary supplement, in PCOS

123
1088 Arch Gynecol Obstet (2014) 290:1079–1092

for both weight management and optimizing reproductive, 18. Gambineri A, Pelusi C, Vicennati V, Pagotto U, Pasquali R
metabolic, and psychological outcomes. (2002) Obesity and the polycystic ovary syndrome. Int J Obes
Metab Disord 26:883–896
19. DeUgarte CM, Bartolucci AA, Azziz R (2005) Prevalence of
Conflict of interest There are no conflicts of interest. insulin resistance in the polycystic ovary syndrome using the
homeostasis model assessment. Fertil Steril 83:1454–1460
20. Carmina E, Lobo RA (2004) Use of fasting blood to assess the
References prevalence of insulin resistance in women with polycystic ovary
syndrome. Fertil Steril 82:661–665
1. Calvo AM, San Millan RM, Sancho JL, Avila SJ, Escobar- 21. Azziz R, Carmina E, Dewailly D et al (2009) The Androgen
Morreale HF (2000) A prospective study of the prevalence of the Excess and PCOS Society criteria for the polycystic ovary
polycystic ovary syndrome in unselected Caucasian women syndrome: the complete task force report. Fertil Steril
from Spain. J Clin Endocrinol Metab 85:2434–2438 91:456–488
2. Dunaif A (1997) Insulin resistance and the polycystic ovary 22. Carmina E, Bucchieri S, Esposito A (2007) Abdominal fat
syndrome: mechanism and implications for pathogenesis. quantity and distribution in women with polycystic ovary syn-
Endocr Rev 18:774–800 drome and extent of its relation to insulin resistance. J Clin
3. Goodarzi MO, Dumesic DA, Chazenbalk G, Azziz R (2011) Endocrinol Metab 92:2500–2505
Polycystic ovary syndrome: etiology, pathogenesis and diagno- 23. Diamanti-Kandarakis E (2006) Insulin resistance in PCOS.
sis. Nat Rev Endocrinol 7:219–231 Endocrine 30:13–17
4. Rotterdam ESHRE/ASRM-Sponsored PCOS Consensus Work- 24. Geisthövel F (2010) Novel systematics of nomenclature and
shop Group (2003) Revised 2003 consensus on diagnostic cri- classification of female functional androgenization (including
teria and long-term health risks related to polycystic ovary polycystic ovary syndrome and non-classic congenital adrenal
syndrome (PCOS). Hum Reprod 19:41–47 hyperplasia). J Reprod Med Endokrinol 7:6–26
5. Ehrmann DA (2005) Polycystic ovary syndrome. N Engl J Med 25. Goverde AJ, van Koert AJ, Eijkemans MJ, Knauff EA, West-
352:1223–1236 erveld HE, Fauser BC, Broekmans FJ (2009) Indicators for
6. Solomon CG (1999) The epidemiology of polycystic ovary metabolic disturbances in anovulatory women with polycystic
syndrome. Prevalence and associated disease risks. Endocrinol ovary syndrome diagnosed according to the Rotterdam con-
Metab Clin North Am 28:247–263 sensus criteria. Hum Reprod 24:710
7. Dunaif A, Graf M, Mandeli J, Laumas V, Dobrjansky A (1987) 26. Lord JM, Flight IH, Norman RJ (2003) Insulin sensitising drugs
Characterization of groups of hyperandrogenic women with (metformin, troglitazone, rosiglitazone, pioglitazone, D-chiro-
acanthosis nigricans, impaired glucose tolerance, and/or hyper- inositol) for polycystic ovary syndrome. Cochrane Database
insulinemia. J Clin Endocrinol Metab 65:499–507 Syst Rev 3:CD003053
8. Jakubowicz DJ, Nestler JE (1997) 17 alpha-hydro- 27. Palomba S, Orio F Jr, Falbo A, Manguso F, Russo T, Cascella T,
xyprogesterone responses to leuprolide and serum androgens in Tolino A, Carmina E, Colao A, Zullo F (2005) Prospective
obese women with and without polycystic ovary syndrome offer parallel randomized, double-blind, double-dummy controlled
dietary weight loss. J Clin Endocrinol Metab 82:556–560 clinical trial comparing clomiphene citrate and metformin as the
9. Plymate SR, Matej LA, Jones RE, Friedl KE (1998) Inhibition first-line treatment for ovulation induction in nonobese anovu-
of sex hormone-binding globulin production in the human latory women with polycystic ovary syndrome. J Clin Endo-
hepatoma (Hep G2) cell line by insulin and prolactin. J Clin crinol Metab 90(7):4068–4074
Endocrinol Metab 67:460–464 28. Palomba S, Orio F Jr, Nardo LG, Falbo A, Russo T, Corea D,
10. Nestler JE, Powers LP, Matt DW et al (1991) A direct effect of Doldo P, Lombardi G, Tolino A, Colao A, Zullo F (2004)
hyperinsulinemia on serum sex hormone-binding globulin levels Metformin administration versus laparoscopic ovarian dia-
in obese women with the polycystic ovary syndrome. J Clin thermy in clomiphene citrate-resistant women with polycystic
Endocrinol Metab 72:83–89 ovary syndrome: a prospective parallel randomized double-blind
11. Utsunomiya T, Taniguchi I, Sadanaga A, Iwasato K, Mizoguchi placebo-controlled trial. J Clin Endocrinol Metab 89(10):
Y, Terawaki S (1993) Insulin resistance in non-obese patients 4801–4809
with polycystic ovary syndrome. Jpn J Fertil Steril 38:77–81 29. Maciel GA, Soares Júnior JM, Alves da Motta EL, Abi Haidar
12. Rosenfield RL, Barnes RB, Ehrmann DA (1994) Studies of the M, de Lima GR, Baracat EC (2004) Nonobese women with
nature of 17-hydroxyprogesterone hyperresponsiveness to gona- polycystic ovary syndrome respond better than obese women to
dotropin-releasing hormone agonist challenge in functional ovar- treatment with metformin. Fertil Steril 81(2):355–360
ian hyperandrogenism. J Clin Endocrinol Metab 79:1686–1692 30. Kumari AS, Haq A, Jayasundaram R, Abdel-Wareth LO, Al
13. Egger M, Smith GD, Altman DG (2001) Systematic reviews in Haija SA, Alvares M (2005) Metformin monotherapy in lean
health care: meta-analysis in context. BMJ 2001:3–68 women with polycystic ovary syndrome. Reprod Biomed Online
14. Yildiz BO, Knochenhauer ES, Azziz R (2008) Impact of obesity 10(1):100–104
on the risk for polycystic ovary syndrome. J Clin Endocrinol 31. Tan S, Hahn S, Benson S, Dietz T, Lahner H, Moeller LC,
Metab 93:162–168 Schmidt M, Elsenbruch S, Kimmig R, Mann K, Janssen OE
15. Apridonidze T, Essah PA, Iuorno MJ, Nestler JE (2005) Prev- (2007) Metformin improves polycystic ovary syndrome symp-
alence and characteristics of the metabolic syndrome in women toms irrespective of pre-treatment insulin resistance. Eur J
with polycystic ovary syndrome. J Clin Endocrinol Metab Endocrinol 157(5):669–676
90:1929–1935 32. Fauser BC, Tarlatzis BC, Rebar RW, Legro RS, Balen AH,
16. Peppard HR, Marfori J, Iuorno MJ, Nestler JE (2001) Prevalence Lobo R, Carmina E, Chang J, Yildiz BO, Laven JS, Boivin J,
of polycystic ovary syndrome among premenopausal women Petraglia F, Wijeyeratne CN, Norman RJ, Dunaif A, Franks S,
with type 2 diabetes. Diabetes Care 24:1050–1052 Wild RA, Dumesic D, Barnhart K (2012) Consensus on
17. Lim SS, Norman RJ, Davies MJ, Moran LJ (2013) The effect of women’s health aspects of polycystic ovary syndrome (PCOS):
obesity on polycystic ovary syndrome: a systematic review and the Amsterdam ESHRE/ASRM-Sponsored 3rd PCOS Consen-
meta-analysis. Obes Rev 14:25–109 sus Workshop Group. Fertil Steril 97:28–38

123
Arch Gynecol Obstet (2014) 290:1079–1092 1089

33. Panidis D, Tziomalos K, Chatzis P, Papadakis E, Delkos D, resistance in polycystic ovary syndrome. J Clin Endocrinol
Tsourdi EA, Kandaraki EA, Katsikis I (2013) Association Metab 91:1508–1512
between menstrual cycle irregularities and endocrine and met- 50. Piotrowski PC, Rzepczynska IJ, Kwintkiewicz J, Duleba AJ
abolic characteristics of the polycystic ovary syndrome. Eur J (2005) Oxidative stress induces expression of CYP11A, CYP17,
Endocrinol 168(2):145–152 STAR and 3bHSD in rat theca-interstitial cells. J Soc Gynecol
34. Robinson S, Kiddy D, Gelding SV, Willis D, Niththyananthan Invest 12(2 Suppl):319A
R, Bush A, Johnston DG, Franks S (1993) The relationship of 51. González F, Minium J, Rote NS, Kirwan JP (2005) Hypergly-
insulin insensitivity to menstrual pattern in women with hy- cemia alters tumor necrosis factor-a release from mononuclear
perandrogenism and polycystic ovaries. Clin Endocrinol (Oxf) cells in women with polycystic ovary syndrome. J Clin Endo-
39(3):351–355 crinol Metab 90:5336–5342
35. Gill S, Taylor AE, Martin KA, Welt CK, Adams JM, Hall JE 52. González F, Rote NS, Minium J, Kirwan JP (2006) In vitro
(2001) Specific factors predict the response to pulsatile gona- evidence that hyperglycemia stimulates tumor necrosis factor-a
dotropin-releasing hormone therapy in polycystic ovarian syn- release in obese women with polycystic ovary syndrome.
drome. J Clin Endocrinol Metab 86(6):2428–2436 J Endocrinol 188:521–529
36. Cupisti S, Kajaia N, Dittrich R, Duezenli H, Beckmann M W, 53. González F, Rote NS, Minium J, Kirwan JP (2006) Reactive
Mueller A (2008) Body mass index and ovarian function are oxygen species-induced oxidative stress in the development of
associated with endocrine and metabolic abnormalities in insulin resistance and hyperandrogenism in polycystic ovary
women with hyperandrogenic syndrome. Eur J Endocrinol syndrome. J Clin Endocrinol Metab 91:336–340
158(5):711–719. doi:10.1530/EJE-07-0515 54. González F, Rote NS, Minium J, Kirwan JP (2009) Evidence of
37. Strowitzki T, Capp E, von Eye Corleta H (2010) The degree of proatherogenic inflammation in polycystic ovary syndrome.
cycle irregularity correlates with the grade of endocrine and Metabolism 58:954–962
metabolic disorders in PCOS patients. Eur J Obstet Gynecol 55. Chazenbalk G, Trivax BS, Yildiz BO (2010) Regulation of
Reprod Biol 149(2):178–181. doi:10.1016/j.ejogrb.2009.12.024 adiponectin secretion by adipocytes in the polycystic ovary
38. Wang JX, Davies MJ, Norman RJ (2001) Polycystic ovarian syndrome: role of tumor necrosis factor-{alpha}. J Clin Endo-
syndrome and the risk of spontaneous abortion following crinol Metab 95:935–942
assisted reproductive technology treatment. Hum Reprod 56. Carmina E, Orio F, Palomba S (2005) Evidence for altered
16(12):2606–2609 adipocyte function in polycystic ovary syndrome. Eur J Endo-
39. Boomsma CM, Fauser BC, Macklon NS (2008) Pregnancy crinol 152:389–394
complications in women with polycystic ovary syndrome. Se- 57. Chen X, Jia X, Qiao J, Guan Y, Kang J (2013) Adipokines in
min Reprod Med 26(1):72–84. doi:10.1055/s-2007-992927 reproductive function: a link between obesity and polycystic
40. Eppig JJ, O’Brien MJ, Pendola FL, Watanabe S (1998) Factors ovary syndrome. J Mol Endocrinol 50:21–37
affecting the developmental competence of mouse oocytes 58. Carmina E (2013) Obesity, adipokines and metabolic syndrome
grown in vitro: follicle-stimulating hormone and insulin. Biol in polycystic ovary syndrome. Front Horm Res 40:40–50
Reprod 59(6):1445–1453 59. Chabrolle C, Tosca L, Ramé C, Lecomte P, Royère D, Dupont J
41. Galal A, Mitwally MF (2009) Insulin sensitizers for women with (2009) Adiponectin increases insulin-like growth factor
polycystic ovarian syndrome. Expert Rev Endocrinol Metab l-induced progesterone and estradiol secretion in human gran-
4:183–192 ulosa cells. Fertil Steril 92:1988–1996
42. Dumesic DA, Schramm RD, Peterson E, Paprocki AM, Zhou R, 60. Pusalkar M, Meherji P, Gokral J, Savardekar L, Chinnaraj S,
Abbott DH (2002) Impaired developmental competence of Maitra A (2010) Obesity and polycystic ovary syndrome:
oocytes in adult prenatally androgenized female rhesus monkeys association with androgens, leptin and its genotypes. Gynecol
undergoing gonadotropin stimulation for in vitro fertilization. Endocrinol 26:874–882
J Clin Endocrinol Metab 87(3):1111–1119 61. Comim FV, Hardy K, Franks S (2013) Adiponectin and its
43. Tao Z, Yan L (2005) Luteinizing hormone and insulin inducing receptors in the ovary: further evidence for a link between
earlier and excess expression of luteinizing hormone receptor obesity and hyperandrogenism in polycystic ovary syndrome.
messenger ribonucleic acids in granulosa cells of polycystic PLoS ONE 18(8):e80416
ovary syndrome. Fertil Steril 84(Suppl):S426–S427 62. Panidis D, Kourtis A, Farmakiotis D, Mouslech T, Rousso D,
44. Diamanti-Kandarakis E (2008) Polycystic ovarian syndrome: Koliakos G (2003) Serum adiponectin levels in women with
pathophysiology, molecular aspects and clinical implications. polycystic ovary syndrome. Hum Reprod 18:1790–1796
Expert Rev Mol Med 10:e3 63. Ardawi MS, Rouzi AA (2005) Plasma adiponectin and insulin
45. Jabara S, Coutifaris C (2003) In vitro fertilization in the PCOS resistance in women with polycystic ovary syndrome. Fertil
patient: clinical considerations. Semin Reprod Med 21(3): Steril 83:1708–1716
317–324 64. Escobar-Morreale HF, Villuendas G, Botella-Carretero JI,
46. Cano F, Garcı́a-Velasco JA, Millet A, Remohı́ J, Simón C, Alvarez-Blasco F, Sanchón R, Luque-Ramı́rez M, San Millán JL
Pellicer A (1997) Oocyte quality in polycystic ovaries revisited: (2006) Adiponectin and resistin in PCOS: a clinical, biochemi-
identification of a particular subgroup of women. J Assist Re- cal and molecular genetic study. Hum Reprod 21(9):2257–2265
prod Genet 14(5):254–261 65. Pinhas-Hamiel O, Singer S, Pilpel N, Koren I, Boyko V, Hemi
47. Kezele PR, Nilsson EE, Skinner MK (2002) Insulin but not R, Pariente C, Kanety H (2009) Adiponectin levels in adolescent
insulin-like growth factor-1 promotes the primordial to primary girls with polycystic ovary syndrome (PCOS). Clin Endocrinol
follicle transition. Mol Cell Endocrinol 192(1–2):37–43 (Oxf) 71(6):823–827. doi:10.1111/j.1365-2265.2009.03604.x
48. Wood JR, Dumesic DA, Abbott DH, Strauss JF (2007) Molec- 66. Mannerås-Holm L, Leonhardt H, Kullberg J, Jennische E, Odén
ular abnormalities in oocytes from women with polycystic ovary A, Holm G et al (2011) Adipose tissue has aberrant morphology
syndrome revealed by microarray analysis. J Clin Endocrinol and function in PCOS: enlarged adipocytes and low serum
Metab 92:705–713. doi:10.1210/jc.2006-2123 adiponectin, but not circulating sex steroids, are strongly asso-
49. González F, Rote NS, Minium J, Kirwan JP (2006) Increased ciated with insulin resistance. J Clin Endocrinol Metab
activation of nuclear factor kB triggers inflammation and insulin 96(2):E304–E311. doi:10.1210/jc.2010-1290

123
1090 Arch Gynecol Obstet (2014) 290:1079–1092

67. Shin HY, Lee DC, Lee JW (2011) Adiponectin in women with manipulation of dietary protein level and glycaemic index. Br J
polycystic ovary syndrome. Korean J Fam Med 32(4):243–248. Nutr 107:106–119
doi:10.4082/kjfm.2011.32.4.243 85. Svendsen PF, Christiansen M, Hedley PL, Nilas L, Pedersen SB,
68. Lecke SB, Mattei F, Morsch DM, Spritzer PM (2011) Abdom- Madsbad S (2012) Adipose expression of adipocytokines in
inal subcutaneous fat gene expression and circulating levels of women with polycystic ovary syndrome. Fertil Steril
leptin and adiponectin in polycystic ovary syndrome. Fertil 98(1):235–241. doi:10.1016/j.fertnstert.2012.03.056
Steril 95(6):2044–2049. doi:10.1016/j.fertnstert.2011.02.041 86. Stofkova A (2010) Resistin and visfatin: regulators of insulin
69. Toulis KA, Gouli DG, Farmakiotis D, Georgopoulos NA, Ka- sensitivity, inflammation and immunity. Endocr Regul 44:25–36
tsikis I, Tarlatzis BC, Papadima I, Panidis D (2009) Adiponectin 87. Tan BK, Chen J, Digby JE, Keay SD, Kennedy CR, Randeva HS
levels in women with polycystic ovary syndrome: a systematic (2006) Increased visfatin messenger ribonucleic acid and protein
review and a meta-analysis. Hum Reprod Update 15:297–307. levels in adipose tissue and adipocytes in women with poly-
doi:10.1093/humupd/dmp006 cystic ovary syndrome: parallel increase in plasma visfatin.
70. Yamauchi T, Kamon J, Ito Y et al (2003) Cloning of adiponectin J Clin Endocrinol Metab 91(12):5022–5028
receptors that mediate antidiabetic metabolic effects. Nature 88. Chan TF, Chen YL, Chen HH, Lee CH, Jong SB, Tsai EM
423:762–769 (2007) Increased plasma visfatin concentrations in women with
71. Hossam OH (2013) Role of adiponectin and its receptor in polycystic ovary syndrome. Fertil Steril 88(2):401–405
prediction of reproductive outcome of metformin treatment in 89. Kowalska I, Straczkowski M, Nikolajuk A, Adamska A, Kar-
patients with polycystic ovarian syndrome. J Obstet Gynaecol czewska-Kupczewska M, Otziomek E, Wolczynski S, Gorska M
Res 39:1596–1603 (2007) Serum visfatin in relation to insulin resistance and
72. Considine RV, Caro JF (1997) Leptin and the regulation of body markers of hyperandrogenism in lean and obese women with
weight. Int J Biochem Cell Biol 29:1255–1272 polycystic ovary syndrome. Hum Reprod 22(7):1824–1829
73. Muoio DM, Lynis DG (2002) Peripheral metabolic actions of 90. Panidis D, Farmakiotis D, Rousso D, Katsikis I, Delkos D,
leptin. Best Pract Res Clin Endocrinol Metab 16:653–666 Piouka A, Gerou S, Diamanti-Kandarakis E (2008) Plasma
74. Bjorbaek C, Kahn BB (2004) Leptin signaling in the central visfatin levels in normal weight women with polycystic ovary
nervous system and the periphery. Recent Prog Horm Res syndrome. Eur J Intern Med 19(6):406–412. doi:10.1016/j.ejim.
59:305–331 2007.05.014
75. Margetic S, Gazzola C, Pegg GG, Hill RA (2002) Leptin: a 91. Ozkaya M, Cakal E, Ustun Y, Engin-Ustun Y (2010) Effect of
review of its peripheral actions and interactions. Int J Obes Relat metformin on serum visfatin levels in patients with polycystic
Metab Disord 26:1407–1433 ovary syndrome. Fertil Steril 93(3):880–884. doi:10.1016/j.
76. Brzechffa PR, Jakimiuk AJ, Agarwal SK, Weitsman SR, fertnstert.2008.10.058
Buyalos RP, Magoffin DA (1996) Serum immunoreactive leptin 92. Plati E, Kouskouni E, Malamitsi-Puchner A, Boutsikou M,
concentrations in women with polycystic ovary syndrome. Kaparos G, Baka S (2010) Visfatin and leptin levels in women
J Clin Endocrinol Metab 81(11):4166–4169 with polycystic ovaries undergoing ovarian stimulation. Fertil
77. Vicennati V, Gambineri A, Calzoni F, Casimirri F, Macor C, Steril 94(4):1451–1456. doi:10.1016/j.fertnstert.2009.04.055
Vettor R, Pasquali R (1998) Serum leptin in obese women with 93. Seow KM, Hwang JL, Wang PH, Ho LT, Juan CC (2011)
polycystic ovary syndrome is correlated with body weight and Expression of visfatin mRNA in peripheral blood mononuclear
fat distribution but not with androgen and insulin levels. cells is not correlated with visfatin mRNA in omental adipose
Metabolism 47(8):988–992 tissue in women with polycystic ovary syndrome. Hum Reprod
78. Pehlivanov B, Mitkov M (2009) Serum leptin levels correlate 26(10):2869–2873. doi:10.1093/humrep/der267
with clinical and biochemical indices of insulin resistance in 94. Spanos N, Tziomalos K, Macut D, Koiou E, Kandaraki EA,
women with polycystic ovary syndrome. Eur J Contracept Re- Delkos D, Tsourdi E, Panidis D (2012) Adipokines, insulin
prod Health Care 14(2):153–159. doi:10.1080/1362518080 resistance and hyperandrogenemia in obese patients with poly-
2549962 cystic ovary syndrome: cross-sectional correlations and the
79. Yildizhan R, Ilhan GA, Yildizhan B, Kolusari A, Adali E, effects of weight loss. Obes Facts 5(4):495–504. doi:10.1159/
Bugdayci G (2011) Serum retinol binding protein 4, leptin, and 000341579
plasma asymmetric dimethylarginine levels in obese and non- 95. Güdücü N, İşçi H, Görmüş U, Yiğiter AB, Dünder I (2012)
obese young women with polycystic ovary syndrome. Fertil Serum visfatin levels in women with polycystic ovary syn-
Steril 96:246–250 drome. Gynecol Endocrinol 28(8):619–623. doi:10.3109/
80. Chapman IM, Wittert GA, Norman RJ (1997) Circulating leptin 09513590.2011.650749
concentrations in polycystic ovary syndrome: relation to 96. Lajunen TK, Purhonen AK, Haapea M, Ruokonen A, Puukka K,
anthropometric and metabolic parameters. Clin Endocrinol Hartikainen AL et al (2012) Full-length visfatin levels are
(Oxf) 46(2):175–181 associated with inflammation in women with polycystic ovary
81. Rouru J, Anttila L, Koskinen P, Penttilä TA, Irjala K, Huup- syndrome. Eur J Clin Invest 42(3):321–328. doi:10.1111/j.1365-
ponen R, Koulu M (1997) Serum leptin concentrations in 2362.2011.02586.x
women with polycystic ovary syndrome. J Clin Endocrinol 97. Olszanecka-Glinianowicz M, Madej P, Zdun D, Bo_zentowicz-
Metab 82(6):1697–1700 Wikarek M, Sikora J, Chudek J, Skałba P (2012) Are plasma
82. Gennarelli G, Holte J, Wide L, Berne C, Lithell H (1998) Is levels of visfatin and retinol-binding protein 4 (RBP4) associ-
there a role for leptin in the endocrine and metabolic aberrations ated with body mass, metabolic and hormonal disturbances in
of polycystic ovary syndrome? Hum Reprod 13(3):535–541 women with polycystic ovary syndrome? Eur J Obstet Gynecol
83. Carmina E, Bucchieri S, Mansueto P, Rini G, Ferin M, Lobo RA Reprod Biol 162(1):55–61. doi:10.1016/j.ejogrb.2012.01.026
(2009) Circulating levels of adipose products and differences in 98. Kasim-Karakas SE, Almario RU, Cunningham W (2009) Effects
fat distribution in the ovulatory and anovulatory phenotypes of of protein versus simple sugar intake on weight loss in poly-
polycystic ovary syndrome. Fertil Steril 1(4 Suppl):1332–1335. cystic ovary syndrome (according to the National Institutes of
doi:10.1016/j.fertnstert.2008.03.007 Health criteria). Fertil Steril 92:262–270
84. Wang P, Holst C, Astrup A et al (2012) Blood profiling of 99. Moran LJ, Noakes M, Clifton PM, Wittert GA, Belobrajdic DP,
proteins and steroids during weight maintenance with Norman RJ (2007) C-reactive protein before and after weight

123
Arch Gynecol Obstet (2014) 290:1079–1092 1091

loss in overweight women with and without polycystic ovary sensitivity in obese subjects with polycystic ovary syndrome.
syndrome. J Clin Endocrinol Metab 92:2944–2951 Fertil Steril 86:243–246
100. Stamets K, Taylor DS, Kunselman A, Demers LM, Pelkman CL, 118. Verstuyf A, Carmeliet G, Bouillon R, Mathieu C (2010) Vitamin
Legro RS (2004) A randomized trial of the effects of two types D: a pleiotropic hormone. Kidney Int 78:140–145
of short-term hypocaloric diets on weight loss in women with 119. Alvarez JA, Ashraf A (2010) Role of vitamin D in insulin
polycystic ovary syndrome. Fertil Steril 81:630–637 secretion and insulin sensitivity for glucose homeostasis. Int J
101. Van Dam EW, Roelfsema F, Veldhuis JD, Helmerhorst FM, Endocrinol 2010:351–385
Frolich M, Meinders AE, Krans HM, Pijl H (2002) Increase in 120. Ngo DT, Chan WP, Rajendran S et al (2011) Determinants of
daily LH secretion in response to short-term calorie restriction in insulin responsiveness in young women: impact of polycystic
obese women with PCOS. Am J Physiol Endocrinol Metab ovarian syndrome, nitric oxide, and vitamin D. Nitric Oxide
282:E865–E872 25:326–330
102. Laughlin GA, Morales AJ, Yen SS (1997) Serum leptin levels in 121. Ortlepp JR, Metrikat J, Albrecht M, von Korff A, Hanrath P,
women with polycystic ovary syndrome: the role of insulin Hoffmann R (2003) The vitamin D receptor gene variant and
resistance/hyperinsulinemia. J Clin Endocrinol Metab physical activity predicts fasting glucose levels in healthy young
82:1692–1696 men. Diabet Med 20:451–454
103. Mantzoros CS, Dunaif A, Flier JS (1997) Leptin concentrations 122. Bourlon PM, Billaudel B, Faure-Dussert A (1999) Influence of
in the polycystic ovary syndrome. J Clin Endocrinol Metab vitamin D3 deficiency and 1, 25 dihydroxyvitamin D3 on de
82:1687–1691 novo insulin biosynthesis in the islets of the rat endocrine
104. Muneyvirci-Delale O, Nacharaju VL, Altura BM, Altura BT pancreas. J Endocrinol 160:87–95
(1998) Sex steroid hormones modulate serum ionized magne- 123. Chiu KC, Chu A, Go VL, Saad MF (2004) Hypovitaminosis D is
sium and Ca levels throughout the menstrual cycle in women. associated with insulin resistance and beta cell dysfunction. Am
Fertil Steril 69:958–962 J Clin Nutr 79:820–825
105. Rumawas ME, McKeown NM, Rogers G, Meigs JB, Wilson 124. Thomson RL, Spedding S, Buckley JD (2012) Vitamin D in the
PWF, Jacques PF (2006) Magnesium intake is related to aetiology and management of polycystic ovary syndrome. Clin
improved insulin homeostasis in the Framingham offspring Endocrinol 7:343–350
cohort. J Am Coll Nutr 25:486–492 125. Homburg R, Amsterdam A (1998) Polycystic ovary syndrome—
106. Chakraborty P, Ghosh S, Goswami SK, Syed NK, Baidyanath C, loss of the apoptotic mechanism in the ovarian follicles?
Kuladip J (2013) Altered trace mineral milieu might play an J Endocrinol Invest 21:552–557
aetiological role in the pathogenesis of polycystic ovary syn- 126. Bikle D (2009) Nonclassic actions of vitamin D. J Clin Endo-
drome. Biol Trace Elem Res 152:9–15 crinol Metab 94:26–34
107. Kauffman RP, Tullar PE, Nipp RD, Castracane VD (2011) 127. Shoelson SE, Herrero L, Naaz A (2007) Obesity, inflammation,
Serum magnesium concentrations and metabolic variables in and insulin resistance. Gastroenterology 132:2169–2180
polycystic ovary syndrome. Acta Obstet Gynecol Scand 128. Yildizhan R, Kurdoglu M, Adali E et al (2009) Serum 25-hydrox-
90:452–458 yvitamin D concentrations in obese and non-obese women with
108. Kurdoglu Z, Kurdoglu M, Demir H, Sahin HG (2012) Serum polycystic ovary syndrome. Arch Gynecol Obstet 280:559–563
trace elements and heavy metals in polycystic ovary syndrome. 129. Pal L, Berry A, Coraluzzi L (2012) Therapeutic implications of
Hum Exp Toxicol 31(5):452–456. doi:10.1177/096032711 vitamin D and calcium in overweight women with polycystic
1424299 ovary syndrome. Gynecol Endocrinol 28:965–968
109. Kelly CC, Lyall H, Petrie JR, Gould GW, Connell JMC, Sattar 130. Wehr E, Pieber TR, Obermayer-Pietsch B (2011) Effect of
N (2001) Low grade chronic inflammation in women with vitamin D3 treatment on glucose metabolism and menstrual
polycystic ovary syndrome. J Clin Endocrinol Metab frequency in polycystic ovary syndrome women: a pilot study.
86:2453–2455 J Endocrinol Invest 34:757–763
110. Slesinski RS, Clarke JJ, San RH, Gudi R (2005) Lack of 131. Selimoglu H, Duran C, Kiyici S, Ersoy C, Guclu M, Ozkaya G,
mutagenicity of chromium picolinate in the hypoxanthine Tuncel E, Erturk E, Imamoglu S (2010) The effect of vitamin D
phosphoribosyltransferase gene mutation assay in Chinese replacement therapy on insulin resistance and androgen levels in
hamster ovary cells. Mutat Res 585:86–95 women with polycystic ovary syndrome. J Endocrinol Invest
111. Cefalu WT, Hu FB (2004) Role of chromium in human health 33:234–238
and in diabetes. Diabetes Care 11:2741–2751 132. Kasim-Karakas SE, Almario RU, Gregory L, Wong R, Todd H,
112. Jeejeebhoy KN, Chu RC, Marliss EB, Greenberg GR, Bruce- Lasley BL (2004) Metabolic and endocrine effects of a poly-
Robertson A (1977) Chromium deficiency, glucose intolerance, unsaturated fatty acid-rich diet in polycystic ovary syndrome.
and neuropathy reversed by chromium supplementation, in a J Clin Endocrinol Metab 89:615–620
patient receiving long-term total parenteral nutrition. Am J Clin 133. Oh R (2005) Practical applications of fish oil (Omega-3 fatty
Nutr 30:531–538 acids) in primary care. J Am Board Fam Pract 18:28–36
113. Mertz W (1998) Chromium research from a distance: from 1959 134. Ebbesson SO, Risica PM, Ebbesson LO, Kennish JM, Tejero
to 1980. J Am Coll Nutr 17:544–547 ME (2005) Omega-3 fatty acids improve glucose tolerance and
114. Davis CM, Vincent JB (1997) Chromium oligopeptide activates components of the metabolic syndrome in Alaskan Eskimos: the
insulin receptor tyrosine kinase activity. Biochemistry Alaska Siberia project. Int J Circumpolar Health 64:396–408
36:4382–4385 135. Rafraf M, Mohammadi E, Asghari-Jafarabadi M, Farzadi L
115. Vincent JB (1999) Mechanisms of chromium action: low- (2012) Omega-3 fatty acids improve glucose metabolism with-
molecular-weight chromium-binding substance. J Am Coll Nutr out effects on obesity values and serum visfatin levels in women
18:6–12 with polycystic ovary syndrome. J Am Coll Nutr 31:361–368
116. Morris BW, MacNeil S, Hardisty CA, Heller S, Burgin C, Gray 136. Kratz M, Swarbrick MM, Callahan HS, Matthys CC, Havel PJ,
TA (1999) Chromium homeostasis in patients with type II Weigle DS (2008) Effect of dietary n-3 polyunsaturated fatty
(NIDDM) diabetes. J Trace Elem Med Biol 13:57–61 acids on plasma total and high-molecular-weight adiponectin
117. Lydic ML, McNurlan M, Bembo S, Mitchell L, Komaroff E, concentrations in overweight to moderately obese men and
Gelato M (2006) Chromium picolinate improves insulin women. Am J Clin Nutr 87:347–353

123
1092 Arch Gynecol Obstet (2014) 290:1079–1092

137. Mohammadi E, Rafraf M, Farzadi L, Asghari-Jafarabadi M, 152. Leidy HJ, Racki EM (2010) The addition of a protein-rich
Sabour S (2012) Effects of omega-3 fatty acids supplementation breakfast and its effects on acute appetite control and food
on serum adiponectin levels and some metabolic risk factors in intake in ‘breakfast-skipping’ adolescents. Int J Obes
women with polycystic ovary syndrome. Asia Pac J Clin Nutr 34:1125–1133
21(4):511–518 153. Zumoff B, Freeman R, Coupey S, Saenger P, Markowitz M,
138. Gillies CL, Abrams KR, Lambert PC et al (2007) Pharmaco- Kream J (1983) A chronobiologic abnormality in luteinizing
logical and lifestyle interventions to prevent or delay type 2 hormone secretion in teenage girls with the polycystic-ovary
diabetes in people with impaired glucose tolerance: systematic syndrome. N Engl J Med 309:1206–1209
review and meta-analysis. BMJ 334:299 154. Prelevic GM, Wurzburger MI, Balint-Peric L (1993) 24-hour
139. Colberg SR, Albright AL, Blissmer BJ et al (2010) Exercise and serum cortisol profiles in women with polycystic ovary syn-
type 2 diabetes: American College of Sports Medicine and the drome. Gynecol Endocrinol 7:179–184
American Diabetes Association: joint position statement. Exer- 155. Domecq JP, Prutsky G, Mullan RJ, Hazem A, Sundaresh V,
cise and type 2 diabetes. Med Sci Sports Exerc 42:2282–2303 Elamin MB, Phung OJ, Wang A, Hoeger K, Pasquali R, Erwin
140. Lee S, Kuk JL, Davidson LE et al (2005) Exercise without P, Bodde A, Montori VM, Murad MH (2013) Lifestyle modi-
weight loss is an effective strategy for obesity reduction in obese fication programs in polycystic ovary syndrome: Systematic
individuals with and without Type 2 diabetes. J Appl Physiol review and meta-analysis. J Clin Endocrinol Metab
99:1220–1225 98(12):4655–4663. doi:10.1210/jc.2013-2385
141. Moran LJ, Pasquali R, Teede HJ, Hoeger KM, Norman RJ 156. Brown AJ, Setji TL, Sanders LL, Lowry KP, Otvos JD, Kraus
(2009) Treatment of obesity in polycystic ovary syndrome: a WE, Svetkey PL (2009) Effects of exercise on lipoprotein par-
position statement of the Androgen Excess and Polycystic Ovary ticles in women with polycystic ovary syndrome. Med Sci
Syndrome Society. Fertil Steril 92:1966–1982 Sports Exerc 41:497–504
142. Bianchi C, Miccoli R, Penno G, Del Prato S (2008) Primary 157. Guzick DS, Wing R, Smith D, Berga SL, Winters SJ (1994)
prevention of cardiovascular disease in people with dysglyce- Endocrine consequences of weight loss in obese, hyperandro-
mia. Diabetes Care 31:208–214 genic, anovulatory women. Fertility, Sterility 61:598–604
143. Kiddy DS, Hamilton-Fairley D, Bush A et al (1992) Improve- 158. Hoeger KM, Kochman L, Wixom N, Craig K, Miller RK,
ment in endocrine and ovarian function during dietary treatment Guzick DS (2004) A randomized, 48-week, placebo-controlled
of obese women with polycystic ovary syndrome. Clin Endo- trial of intensive lifestyle modification and/or metformin therapy
crinol (Oxf) 36:105–111 in overweight women with polycystic ovary syndrome: a pilot
144. Clark AM, Ledger W, Galletly C et al (1995) Weight loss results study. Fertil Steril 82:421–429
in significant improvement in pregnancy and ovulation rates in 159. Hoeger K, Davidson K, Kochman L, Cherry T, Kopin L, Guzick
anovulatory obese women. Hum Reprod 10:2705–2712 DS (2008) The impact of metformin, oral contraceptives, and
145. Thomson RL, Buckley JD, Moran LJ, Noakes M, Clifton PM, lifestyle modification on polycystic ovary syndrome in obese
Norman RJ, Brinkworth GD (2009) The effect of weight loss on adolescent women in two randomized, placebo-controlled clin-
anti-Müllerian hormone levels in overweight and obese women ical trials. J Clin Endocrinol Metab 93:4299–4306
with polycystic ovary syndrome and reproductive impairment. 160. Karimzadeh MA, Javedani M (2010) An assessment of lifestyle
Hum Reprod 24:1976–1981 modification versus medical treatment with clomiphene citrate,
146. Holte J, Bergh T, Berne C, Wide L, Lithell H (1995) Restored metformin, and clomiphene citrate-metformin in patients with
insulin sensitivity but persistently increased early insulin polycystic ovary syndrome. Fertil Steril 94:216–220
secretion after weight loss in obese women with polycystic 161. Palomba S, Falbo A, Giallauria F, Russo T, Rocca M, Tolino A,
ovary syndrome. J Clin Endocrinol Metab 80:2586–2593 Zullo F, Orio F (2010) Six weeks of structured exercise training
147. Moran LJ, Hutchison SK, Norman RJ, Teede HJ (2011) Life- and hypocaloric diet increases the probability of ovulation after
style changes in women with polycystic ovary syndrome. clomiphene citrate in overweight and obese patients with poly-
Cochrane Database Syst Rev 7:CD007506. doi:10.1002/ cystic ovary syndrome: a randomized controlled trial. Hum
14651858.CD007506 Reprod 25:2783–2791
148. Moran LJ, Ko H, Misso M, Marsh K, Noakes M, Talbot M et al 162. Qublan HS, Yannakoula EK, Al-Qudah MA, El-Uri FI (2007)
(2013) Dietary composition in the treatment of polycystic ovary Dietary intervention versus metformin to improve the repro-
syndrome: a systematic review to inform evidence-based ductive outcome in women with polycystic ovary syndrome. A
guidelines. J Acad Nutr Diet 113(4):520–545. doi:10.1016/j. prospective comparative study. Saudi Med J 28:1694–1699
jand.2012.11.018 163. Stener-Victorin E, Jedel E, Janson PO, Sverrisdottir YB (2009)
149. Douglas CC, Gower BA, Darnell BE, Ovalle F, Oster RA, Azziz Low-frequency electroacupuncture and physical exercise
R (2006) Role of diet in the treatment of polycystic ovary decrease high muscle sympathetic nerve activity in polycystic
syndrome. Fertil Steril 85:679–688 ovary syndrome. Am J Physiol Regul Integr Comp Physiol
150. Mehrabani HH, Salehpour S, Amiri Z, Farahani SJ, Meyer BJ, 297:R387–R395
Tahbaz F (2012) Beneficial effects of a high-protein, low-gly- 164. Vigorito C, Giallauria F, Palomba S, Cascella T, Manguso F,
cemic-load hypocaloric diet in overweight and obese women Lucci R, De Lorenzo A, Tafuri D, Lombardi G, Colao A, Orio F
with polycystic ovary syndrome: a randomized controlled (2007) Beneficial effects of a three-month structured exercise
intervention study. J Am Coll Nutr 31:117–125 training program on cardiopulmonary functional capacity in
151. Graff SK, Mário FM, Alves BC, Spritzer PM (2013) Dietary young women with polycystic ovary syndrome. J Clin Endo-
glycemic index is associated with less favorable anthropometric crinol Metab 92:1379–1384
and metabolic profiles in polycystic ovary syndrome women
with different phenotypes. Fertil Steril 100:1081–1088

123

You might also like