ISSN: 1734-1922
Archives of Medical Science Special Issues
Current issue Archive Archives of Medical Science
1/2009
 
Share:
Share:

Invited review
Adolescent polycystic ovary syndrome: pathophysiology and implications of the disease

Sajal Gupta
,
Dinah Chen
,
Katherine O’Flynn O’Brien
,
Anjali Chandra
,
Lauren Metterle
,
Shubhangi Kesavan
,
Ashok Agarwal

Arch Med Sci 2009; 5, 1A: S115–S131
Online publish date: 2009/06/10
Article file
Get citation
 
 

Introduction
Polycystic Ovary Syndrome (PCOS) is one of the most prevalent female reproductive endocrinopathies, affecting 5 to 10% of women worldwide [1]. A relatively widespread disorder, PCOS has remained an important topic of research. Along with endocrine and genetic factors, obesity is a key complication related to PCOS. Of the women clinically diagnosed with this syndrome, 50% are overweight or obese and between 50 and 60% reportedly exhibit insulin resistance [2, 3]. Many studies have reported a link between obesity, insulin resistance, and hyperinsulinemia in women with PCOS. Obesity has become an epidemic in developed countries, not only in adults but adolescents as well. In fact, one study estimates that 15% of adolescents are overweight, a percentage that has tripled over the past two decades [4]. Therefore, concern is increasing over the possible effect on obesity related syndromes, such as PCOS, especially in adolescents. Consequently, adolescent obesity and PCOS is currently a topic of intense concern.
Polycystic ovary syndrome is the most common cause of menstrual dysfunction in teenage girls. Although only a few studies focusing on the prevalence of PCOS among adolescents have been conducted, one reported the adolescent incidence to be 3% [5, 6]. More than 50% of adolescents exhibiting the features of this disorder (hirsutism, acne, hyperandrogenemia, etc.) are obese and at high risk for developing type II diabetes [7, 8].
Attempts at standarization
The diagnosis of PCOS remains a subject of contention. Several attempts have been made to standardize its definition and diagnostic criteria, most notably at the National Institutes of Health (NIH) conference of 1990, the Rotterdam conference of 2003, and the Androgen Excess Society (AES) conference held in 2006. The AES requires the specific presence of clinical and/or biochemical hyperandrogenism in combination with either polycystic ovary morphology and/or oligo- anovulation [9]. The NIH consensus of 1990 defined PCOS as the presence of both hyperandrogenism and ovarian dysfunction, with no mention of ovarian morphology. However, the Rotterdam consensus included the presence of polycystic ovary morphology along with hyperandrogenism and oligo/anovulation [10]. Both the NIH and AES criteria also stipulate that other causes of hyperandrogenism, such as Cushing’s syndrome or hyperprolactinemia, be excluded [11]. There are three sets of criteria that have been proposed to establish the diagnosis in adult women: the NIH criteria, the Rotterdam criteria, and the Androgen Excess Society guideline.
All three criteria agree that the exclusion of other disorders is necessary. The 2003 Rotterdam criteria require the presence two of three conditions: (1) oligo- or anovulation, (2) clinical and/or biochemical signs of hyperandrogenism, and/or (3) polycystic ovaries on transvaginal ultrasound (12 or more follicles in each ovary measuring 2-9 mm or increased ovarian volume 10 ml of one ovary). This definition would result in four subsets of PCOS: (1) irregular menstrual pattern plus polycystic ovary morphology plus (clinical or laboratory) hyperandrogenism, (2) irregular menses plus hyperandrogenism, (3) hyperandrogenism plus polycystic ovary morphology, and (4) irregular menses plus polycystic ovary morphology. The last category remains in dispute in regards to whether these patients should be diagnosed under the broad rubric of PCOS, because of some similar metabolic findings, or whether such labeling is premature. The Andro-gen Excess Society, published 2006, underscored the need to define PCOS.
Currently, there is no defined standard to judge and diagnose PCOS in adolescents for a variety of reasons. First, ovulation is often irregular in early menarche and thus, anovulation cannot be considered a definite indication of the existence of the syndrome [12]. Second, multifollicular ovaries, which normally may be present in adolescent girls, are difficult to differentiate from polycystic ones [12]. Third, transvaginal ultrasounds are not routinely performed in adolescents, which inhibits the visualization of the ovaries and therefore precludes any invasive diagnosis of polycystic morphology [12]. Finally, there is little information regarding normal levels of androgens in adolescents and a lack of consensus on the chemical levels of hyperandrogenemia. Therefore, determining whether levels of androgens are abnormally high is a complex task [12]. It appears that a diagnosis of PCOS in this age group is mainly based upon the criteria of clinical hyperandrogenism and menstrual dysfunction [6].
Associated complications of polycystic ovary syndrome
Because irregular ovulation or anovulation is typical in adolescents, the current basis of diagnosis of PCOS in adolescents relies heavily upon clinical signs of hyperandrogenism. In a recent study, 42% of adolescent girls with oligomenorrhea – 22% of whom also had polycystic ovaries – were found to be positive for PCOS [13, 14]. This finding suggests menstrual dysfunction in addition to polycystic morphology is strong indicators of PCOS in adolescents.
Women and adolescents with PCOS present with certain common physical characteristics. Many experience excess male-pattern hair growth. This symptom, termed hirsutism, is a marker of hyperandrogenism and usually becomes apparent at menarche. The patient may have acne as well as dark pigmentation of the skin, usually around the neck, which is known as acanthosis nigricans. Polycystic ovary morphology is present in some, but not all, women with PCOS. These ovaries are enlarged and contain an increased number of developmentally arrested, small antral follicles [15].
Obesity, and in particular, central adiposity, is another characteristic commonly associated with the syndrome. Obesity increases the severity of insulin resistance and propels hyperandrogenism through various mechanisms [4]. All of these traits are implicated in the internal conditions of the PCOS woman.
Insulin resistance
The effect of insulin resistance on PCOS is complicated. Women with PCOS tend to have more severe insulin resistance than their normal counterparts. Insulin resistance is found in 50 to 60% [3] of PCOS women, and 31% [16] present with glucose intolerance. Insulin resistance is often accompanied by hyperinsulinemia and hyperandrogenemia. Consequently, both hyperinsulinemia and hyperandrogensm are often present in women with PCOS. As documented, there is a cyclic relationship between hyperinsulinemia and hyperandrogenism in PCOS women; insulin induces the production of androgen, which in turn propels visceral fat accumulation and eventually exacerbates insulin resistance, leading to severe hyperinsulinemia [14].
Hyperandrogenism is compelled by the actions of insulin through various mechanisms. One such mechanism is the dysregulation of sex hormone binding globulin (SHBG). Hyperinsulinemia is known to suppress the function of SHBG [16]. Once concentrations of this globulin fall, it can no longer bind to testosterone. Subsequently, levels of free circulating testosterone rise and androgen production increases. Studies support a negative correlation between the concentration of SHBG and insulin [2].
A second mechanism by which insulin resistance affects the production of androgens is through the activation of gonadotrophin releasing hormone (GnRH). Activation of GnRH leads to elevated levels of lutenizing hormone (LH) [12], which eventually causes the production of androgens at the theca cell level. Conversely, the presence of hyperandrogenemia has been indicated as a possible cause of increased secretion of LH [16]. Once again, the vicious cycle linking insulin resistance to hyperandrogenism manifests itself in this positive feedback system.
Androgen synthesis is further increased through the actions of insulin on the ovary and pituitary gland. The insulin dysfunction characteristic of women with PCOS may occur due to dysfunction of the insulin receptor or defects in the signaling pathways involving insulin [15]. High concentrations of insulin increase androgen secretion by overpowering insulin receptors, which subsequently reduces SHBG levels [15]. Additional androgen production is propelled by the effects of excess insulin, which serves to inhibit the synthesis of insulin growth factor (IGF) binding proteins. This allows IGF-1 to bind on the ovaries and increase androgen production [17].
A study by Sawathiparnich indicates that the level of insulin resistance is elevated in girls with PCOS and is independent of obesity [18]. Glucose intolerance, which has been found to increase the severity of insulin resistance and hyperinsulinemia [19], is also a key component of PCOS. In a study conducted by Palmert et al., 33% of adolescent girls with PCOS exhibited impaired glucose tolerance [18, 20]. Furthermore, another study demonstrated that glucose intolerance was present at higher percentages among obese PCOS adolescents than in PCOS patients with normal body weights [14]. Thus, not only are PCOS adolescents already predisposed to severe insulin resistance, but obese PCOS adolescents suffer the effects of an exacerbated level of glucose intolerance compounded with all other associated symptoms.
Visceral fat/adiposity
Another common phenotypic trait of PCOS women is visceral fat. Women with PCOS demonstrate a comparatively high level of central adiposity, even amongst those who do not present with obesity [21]. The accumulation of fat in the abdominal area has been correlated with an increased presence of menstrual dysfunction, acanthosis nigricans, and severe hirsutism [22]. Studies have further correlated the level of androgens and the amount of visceral fat present [14].
In adult patients and adolescents alike, 50 to 70% have increased central adiposity accompanying PCOS [23, 25]. The consequences of adiposity may be at hand even in adolescence [24], especially in a society in which obesity is rapidly becoming the norm [11, 25]. Increased central adiposity has been shown to be directly proportional to the level of insulin in the body and also to the severity of insulin resistance [14, 26]. Visceral adiposity can incite insulin resistance and metabolic syndrome [11, 25].
Central adiposity causes both androgen production and is stimulated by it, feeding a vicious cycle [27, 28]. Like their adult counterparts, PCOS adolescents are believed to be at an elevated risk level for developing diabetes mellitus or metabolic dysregularities [29] due to androgen excess [11]. Currently, the diagnosis of PCOS does not consider adiposity as an identifier of the syndrome. However, the pathogenic effect of adiposity on hormonal activity cannot be denied [11, 30].
Relationship of obesity and polycystic ovary syndrome
It is generally acknowledged that the existence and proliferation of PCOS is closely tied to obesity. Not only does obesity itself pose a risk in obvious ways, but it also complicates and compels the development of hyperinsulinemia and hyperandrogenism. Women who are obese or overweight naturally exhibit higher levels of endocrine dysfunction. The presence of obesity exacerbates these endocrinal disturbances. Thus, it appears that being overweight both induces and sustains PCOS.
There is an initiatory relationship between obesity and the production of androgens [10]. In a study by McCartney et al., it was determined that obese girls exhibited higher levels of hyperandrogenemia. Furthermore, the production of androgens in itself could be a cause of PCOS [11, 31]. This finding highlights obesity’s dynamic role in PCOS: it is apparent that obesity, which is caused by elevated levels of androgens, also increases the production of androgens that lead to the development of PCOS.
Obesity also exacerbates the abnormalities concerning gonadotropin and sex steroid secretion in women with PCOS. Further SHBG down- regulation, a globulin already present in lower levels in women with PCOS, is propelled by obesity [32]. Additionally, obesity negatively affects the ovary by further disrupting the ratio of LH to follicle stimulating hormone (FSH). One study determined that LH levels were increased in obese women with PCOS [4, 33]. This increased LH secretion will also affect the production of androsteriodione in theca cells, leading to an increase in androgen levels.
Similarly to adult PCOS patients, obese adolescent girls with PCOS appear to have a higher rate of menstrual disturbance as well as elevated levels of testosterone and free androgens [4]. Levels of SHBG were lower in overweight adolescent females than in PCOS adolescents who were not overweight [24]. Moreover, as a function of overweight and obesity, adolescent PCOS females also exhibit higher levels of insulin which promotes the production of androgen by the ovaries [34]. Ultimately, obesity serves to both exacerbate and instigate the problems of the syndrome. Consequently, it is suggested that the earlier the onset of obesity, the greater the consequences for the young patient [11].
Ovarian dysfunction
Polycystic ovary syndrome impairs the theca and granulosa cells of the developing follicle. Each cell operates under the direction of LH or FSH to produce steroids. The theca cell contains receptors for LH, which stimulates the production of androstenedione from cholesterol when bound. Androstenedione then interacts within the granulosa cell. The periphery of the granulosa cells contains receptors for FSH, which bind and stimulate the aromatase enzyme to convert androgens to estrogens. In the PCOS woman, the roles of each cell are impaired, and the result of this malfunction is hyperandrogenism.
Theca cells
Patients with PCOS exhibit signs of increased steroidogenesis and enhanced androgen activity due to a greater number of theca cells. It has been suggested that overproduction of androgens is facilitated by the action of insulin on theca cells. Insulin functions to upregulate the expression of the P450c17 gene in theca cells, in addition to enhancing the LH receptor activity of the cell [35, 36]. The P450c17 gene encodes the enzyme 17-hydroxylase; this enzyme is responsible for androstenedione production, which eventually is converted to testosterone. Therefore, insulin ultimately leads to the presence of excess testosterone. Additionally, it is thought that insulin may stimulate ovarian steroidogenesis by mimicking LH activity [37]. Thus, hyperinsulinemia plays a key role at the level of the ovaries in PCOS.
Granulosa cells
Aromatase inhibition within granulosa cells plays a critical role in the development of hyperandrogenemia. In women with PCOS, an imbalance in the ratio of LH to FSH persists due to elevated concentration of LH. Follicle stimulating hormone is essential in stimulating the aromatase enzyme for the conversion of androgen to estrogen. However, the increased production of androgens within the theca cells overpowers the capacity of the granulosa cells. Thus, the concentration of androgen remains elevated.
Moreover, Mullerian inhibiting substances (MIS) contribute to the accumulation of androgens and the eventual arrest of follicular development. Mullerian inhibiting substances are thought to inhibit aromatase function [38]. Grossman et al. [39] concluded that the presence of MIS in the granulosa cells leads to a notable decrease in estradiol production [39]. According to a study conducted by Wachs, women with PCOS have 3.4 times the typical amount of MIS [40]. The results of these studies coincide with the belief that MIS promotes androgen excess.
Fetal disposition
Polycystic ovary syndrome manifests during adolescence, suggesting a pre-pubertal or pre-natal predispostion [1, 6, 31, 41]. According to one hypothesis, PCOS occurs when a fetus is overexposed to androgens during its development. Many studies on female animal models support this concept. An analysis of sheep that were exposed to excess androgens in-utero ascertained that the GnRH pulse generator had been reprogrammed, resulting in a decrease in sensitivity to estradiol negative feedback [42, 43]. This alteration of hormone levels leads to elevated LH levels, causing issues in follicular development. Additionally, the exposure seemed to be linked to an increase in follistatin and a decrease in activin, which suppress FSH and obstruct follicular growth [42]. Another study subjected female rhesus monkeys to high levels of testosterone during in-utero development. Later in life, these monkeys displayed hyperandrogenism, ovulatory dysfunction, and defective insulin secretion, all features associated with PCOS. Increased levels of LH appeared only in monkeys exposed to testosterone during early gestation, which suggests that there is a critical time period during which exposure leads to PCOS [44, 45].
Evidence for pre-natal and pre-pubertal predisposition has also been supported by clinical studies in humans. Both Hague et al. and Barnes et al. conducted a study of women who were prenatally exposed to high androgen levels as evidenced either by congenital adrenal hyperplasia or congenital adrenal virilizing tumors. After birth, androgen levels normalized; however, many women exhibited PCOS later in life [46, 47]. Another study demonstrated an association amongst prenatal androgen exposure, loss of P450 aromatase or SHBG gene function, and PCOS [48, 49]. Unfortunately, the precise mechanism of excessive androgens during fetal development still remains unclear.
Polycystic ovary syndrome in pregnant mothers is a potential cause of PCOS in offspring [50]. Various studies have demonstrated that pre-existing elevated androgen levels in combination with the hyperinsulinemia in pregnant women with PCOS overexposes a fetus to high androgens levels. Under normal conditons, placental aromatase converts excess androgens to estrogens. In the presence of hyperinsulinemia, aromatase activity is inhibited. Consequently, pregnant women with PCOS create a fetal environment with increased exposure to androgens [50-53]. Additionally, exposure to high levels of insulin has been implicated as a cause of low birth weight, and birth weight is a potential marker of PCOS [1].
Extreme birth weight, either low or high birth weight, can result from altered developmental pathways, leading to severe adiposity in conjunction with unhealthy diet patterns. This, in turn, predisposes to hyperinsulinemia, hyperandrogenism, and, after puberty, ovulatory dysfunction due to issues with follicle growth and development. Poor in-utero nutrition results in low birth weight and subsequent fetal growth restriction. As a means of survival, the fetus undergoes permanent physiological and structural changes [54-57]. However, in the post-natal period, with subsequent exposure to adequate nutrition and excessive adiposity, PCOS features can develop [23, 31].
Conversely, women with elevated birth weights maintain high levels of adiposity throughout early childhood due to poor dietary patterns thereby increasing their risk for PCOS [58-60]. Interestingly, women with high birth weights more often exhibit all three major criteria for PCOS diagnosis: hyperandrogenism, anovulation, and polycystic ovaries; whereas women with low birth weight typically do not present with polycystic ovaries [58].
Childhood obesity, independent of fetal nutrition, represents another potential factor in PCOS development. It is associated with decreased SHBG levels resulting in increased free testosterone and insulin resistance with compensatory hyperinsulinemia. Studies demonstrate obese adolescents exhibit insulin resistance more often than their normal weight counterparts. Increased insulin levels promote ovarian androgen production, leading to hyperandrogenism. Hyperandrogenism subsequently distorts the hypothalamic pituitary ovarian axis, resulting in a persistently rapid pulse from the GnRH pulse generator, making the body resistant to estrogenic negative feedback. Consequently, adolescent women with PCOS have higher LH levels and lower levels of FSH. Altered hormone levels inhibit proper follicular development and lead to the expression of PCOS during adulthood [23, 31, 61, 62].
The role of obesity in the pathogenesis of PCOS has been demonstrated in many studies. The McCartney et al. study of 76 peripubertal girls found that those who were overweight girls showed greater rates of hyperandrogenemia when compared to normal weight girls [31]. Another study reported that total testosterone in obese pre-pubertal girls was 40 to 50% higher than that in age-matched equivalents, although the difference was not statistically significant because of the small sample size [63]. This was also demonstrated in a study of pre-pubertal German girls, which found that obese girls had lower SHBG levels and a median testosterone concentration that was four times greater than that of the normal weight girls [61]. There is currently no definite causal relationship between androgens and adiposity. However, pre-pubertal obesity consistently seems promote hyperandrogenism. Various studies have demonstrated correlations between adiposity and higher levels of testosterone, however some studies conclude the two are independently related and the correlation is attributed to an inverse relationship between SHBG and adiposity [31].
In a recent study, Leibel et al. concluded that the presence of metabolic syndrome in the father of a woman with PCOS seemed to be more related to the maternal PCOS. The prevalence of metabolic syndrome was 1.5 to 2 fold greater in the fathers of daughters with PCOS than in the average population [64]. However, this association needs further investigation.
GnRH pulse generator
The GnRH pulse generator is a group of hypothalamic neurons that release GnRH in a pulsatile manner. GnRH stimulates the secretion of LH and FSH. Varying levels of LH and FSH control follicular growth and development, effectively regulating the menstrual cycle [65]. The frequency of the GnRH pulse generator modulates the differential production of LH and FSH [65, 66]. Rapid pulses favor the production of LH while slower pulses favor the production of FSH [67-69]. Kirk et al. concluded this occurs as a result of rapid GnRH pulses increasing the expression of follistatin. Follistatin acts to inhibit the effects of FSH by binding to intragonadotrope activin, therefore favoring the production of LH [70]. In women with normal ovulatory function, the GnRH pulses gradually increase during the follicular phase and peak in the later half, increasing LH production and decreasing FSH production. Simultaneously, estradiol production reaches a threshold due to positive feedback, permitting the release of the LH, known as the mid-cycle LH surge; this induces ovulation. Ovulation is followed by the development of the corpus luteum, which manufactures progesterone. The progesterone subsequently slows the GnRH pulse generator, emitting pulses that favor FSH, and creating what is known as an FSH window. This is responsible for initiating the next wave of follicular growth and development [71-75].
Women with PCOS, however, have invariably rapid GnRH pulse frequencies in favor of LH [76]. Thus, these women typically present with persistently elevated LH levels and LH:FSH ratios [77]. LH acts as a stimulatant for ovarian androgen production. Women with PCOS, therefore, present with hyperandrogenemia. Blank et al. postulated that the systemic issues in women with PCOS may be the result of failure to suppress GnRH pulsatility [66].
Long-term consequences of polycystic ovary syndrome
Over time, PCOS can cause serious health conditions. It is critical for physicians treating PCOS patients to understand how the varying pathophysiologies of the disease can combine to cause additional health issues. Women with PCOS often develop hyperandrogenism, hirsutism, and insulin resistance and experience excessive weight gain. The synergy of these symptoms can result in health issues such as metabolic syndrome, cardiovascular disease, type II diabetes mellitus, cancer, sub-fertility, and psychological distress [2, 9, 19, 29, 78-93].
Metabolic syndrome
Metabolic syndrome is characterized by several risk factors that can lead to cardiovascular disease, which is a major long-term consequence associated with PCOS. Characteristics of metabolic syndrome are abdominal obesity, increased levels of triglycerides, hypertension, increased plasma levels of fasting glucose, hyperinsulinemia, and decreased levels of high density lipopro- tein cholesterol (HDL-c) [17, 94]. The condition is particularly related to PCOS because of the prevalence of insulin resistance [17]. Furthermore, adolescent girls with PCOS are at higher risk for developing metabolic syndrome because they already exhibit these risk factors at a young age [88]. Coviello et al. reported that the prevalence of metabolic syndrome in young girls with PCOS was 37 vs. 5% in a control group of normal girls. The study also found that obesity and insulin resistance were risk factors for metabolic syndrome but that hyperandrogenemia was the only constant predictor of the syndrome. After adjusting for body mass index (BMI) and insulin resistance, adolescent girls with PCOS had almost four times the risk of metabolic syndrome for every 25% increase in free testosterone in comparison with the control group [95]. Body mass index and waist circumference, a measure of abdominal fat, were found to be predictors of cardiovascular disease [96]. In the Coviello study, the prevalence of metabolic syndrome was 62% in obese women with PCOS and 63% in extremely obese women with PCOS [95].
Adolescence is a critical time to recognize the risks associated with metabolic syndrome because its onset during adolescence is correlated with the development of cardiovascular disease in adulthood [97]. The syndrome also contributes to hyperandrogenemia, further exacerbating the cycle of accumulating risk factors for the disease [95]. The combination of metabolic syndrome and PCOS promotes higher levels of androgens in the body [98], which can act on androgen receptors located on adipocytes, causing further accumulation of intra-abdominal fat [99]. Central adiposity is a factor in both PCOS and MS, which contributes to the vicious cycle of hyperinsulinemia and hyperandrogenemia. Patients with PCOS who suffer from metabolic syndrome also have lower high density lipoprotein cholesterol (HDL-c) levels, higher low density lipoprotein cholesterol (LDL-c) levels, and higher levels of triglycerides than girls who only have PCOS [95]. These factors present an added cardiovascular risk in young PCOS patients [97].
Young PCOS patients should be counseled about ways to prevent metabolic syndrome in order to avoid cardiovascular disease and type II diabetes mellitus in adulthood [17]. Data collected about abdominal fat and triglyceride levels in girls aged 9 through 11 was found to be predictive of whether a patient would develop the syndrome before entering their twenties, demonstrating the necessity to emphasize the importance of a healthy lifestyle early in life [100]. Furthermore, rates of metabolic syndrome in adolescence increase concurrently with body weight, reaching a prevalence of 50% in excessively obese children [20, 25, 101, 102]. Consequently, it is critical to recognize that the symptoms of PCOS can exacerbate the development of metabolic syndrome and that it is essential for patients with PCOS to initiate proper diet and exercise at an early age to reduce the likelihood of developing the syndrome [95].
Another complication associated with PCOS is nonalcoholic fatty liver disease (NAFLD), which is usually considered the hepatic component of MS [103]. One of the most common causes of liver disease in western countries [104, 105], NAFLD is characterized by steatosis or fibrosis of the liver [105]. A more aggressive form of the disease is nonalcoholic steatohepatitis (NASH), which is characterized by “necroinflammatory changes” and pericellular fibrous degeneration [106]. Patients with NASH are at a greater risk of developing serious conditions such as “advanced fibrosis, cirrhosis, and hepatocellular carcinoma” [107]. NAFLD and NASH share the risk factor of insulin resistance, a common problem associated with PCOS [108-110]. Approximately 50% of PCOS patients exhibit insulin resistance and also have metabolic syndrome.
A study conducted by Cerda et al. determined that the prevalence of NAFLD in PCOS patients was 41%, which was significantly higher than the prevalence of the disease in the control group (19%) [106]. Additionally, the PCOS patients in the study had a much higher frequency of insulin resistance; 63 vs. 31% in the control group. The relationship between insulin resistance and NAFLD in PCOS patients was also related to their BMI: a higher BMI promoted increased levels of insulin resistance, which acted to increase the prevalence of NAFLD [111]. Another serious finding of the study by Cerda et al. is that PCOS women have high levels of aminotransferase, which is an indicator for the development of NASH [106]. Overall, this study demonstrated the importance of early detection and treatment of liver disease in PCOS patients to avoid serious complications like NASH, which is more amenable to treatment during adolescence [104, 112].
Cardiovascular disease
The development of cardiovascular disease and other vascular conditions is a serious consequence associated with PCOS. Similarly, as in the development of metabolic syndrome, the pathophysiology of PCOS is also related to cardiovascular diseases. Obesity, high blood pressure, type II diabetes, insulin resistance, dyslipidemia, abdominal fat, and decreased ability to break down blood clots – all factors that relate to PCOS – also put PCOS women at a greater risk for heart disease. High androgen levels in the body also play a critical role in the development of heart disease as calcification of the aorta is related to hyperandrogenemia independent of BMI, age, and incidence of PCOS, highlighting their significant role in the deve-lopment of the disease [113]. All of these are risk factors for heart disease. Obesity during adolescence was also correlated with carotid intimal medial thickening, which may partly explain how PCOS predisposes women with a high BMI and hypertension to vascular diseases [114, 115]. Additionally, another study found that women with PCOS who have a combination of insulin resistance, high blood pressure, and dyslipidemia also have a greater chance for developing cardiovascular disease and cerebral vascular issues [116].
In a recent study by Battaglia et al., PCOS women had higher white blood cell counts than healthy women (7,273±789 vs. 6,182±1,004, respectively). Studies have discovered that increased inflammation can lead to the onset and progression of atherosclerosis. Consequently, this finding further emphasizes that women with PCOS are prone to heart disease [117]. The study by Battaglia et al. also found that in PCOS women, the mean (SD) level of homocysteine was 11.5±0.5 vs. 9.8±1.5 µmol/l in the control population. Homocysteine levels have long been recognized as a risk factor for heart disease [118]. The elevation of homocysteine levels combined with advancing age [117] puts POCS women at risk for heart disease [119].
Furthermore, lipid and androgen levels were linked with cardiovascular disease in patients with PCOS. Previous studies have connected insulin resistance to dyslipidemia, a condition found in PCOS women, because insulin plays a role in the up-regulation of lipoprotein-lipase. However, the Battaglia study found that lipid levels were also correlated with androgen levels, triglyceride levels, and cholesterol, independent of the insulin levels in the body. This finding was confirmed by the fact that the atherogenic index, a marker that is used to determine one’s risk for heart disease [120], is correlated with higher levels of androgens, triglycerides, and cholesterol levels in PCOS women 117]. The atherogenic index is defined as the log (triglycerides/HDL-c) and has been found to be related with one’s risk for heart disease [120]. This relationship highlights the fact that high androgen levels and dyslipidemia may be related to the development of heart disease [117].
The pathophysiology of PCOS also puts women at risk for other vascular problems. Low levels of NO2–/NO3– combined with lipid dysregulation and high levels of androgens impede vasodilation in the endothelium of PCOS women, a condition which can precede other vascular diseases [117]. Measurements of ophthalmic nerve pressure were obtained from PCOS women and used to estimate cerebral pressure. Polycystic ovary syndrome women had increased pressure in their ophthalmic nerves, which may be due to increased arterial stiffness caused by calcification, lower amounts of NO2–/NO3–, and more deposits of triglycerides in the arteries [117]. In obese individuals with PCOS, this effect could be magnified because obesity can put pressure on the right side of the heart and reduce the reuptake of cerebrospinal fluid, which causes increased pressure on the brain [121, 122]. Increased cerebral pressure is also related to another disease called idiopathic intracranial hypertension or IIH [123]. Symptoms include papilledema, impaired vision, increased cerebrospinal fluid pressure, and headache [124-128].
Type II diabetes mellitus
The combination of abdominal fat, obesity, insulin resistance, and b-cell dysfunction in women with PCOS creates an environment in the body that makes them more susceptible to type II diabetes mellitus [83, 85, 88, 100, 129-131]. Type II diabetes mellitus can exacerbate the already fragile vascular state of PCOS patients and can decrease the amount of blood flow to the brain [132]. Women with PCOS are at a greater risk for developing diabetes than healthy women. Long-term studies of women with PCOS have observed that 13 to 16% developed diabetes over a three- to eight-year period [133, 134]. The prevalence of the disease in women without PCOS is only 2% [135]. Additionally, the combination of obesity and PCOS is particularly harmful [136]. Obese women with PCOS have ten times the risk of developing the disease than non-obese PCOS patients [135] and five times the risk of developing type II diabetes mellitus than non-PCOS women when obesity and age were controlled [133].
Gestational diabetes mellitus (GDM), an intolerance to carbohydrates that begins during the early stages of pregnancy, is another problem afflicting PCOS patients, especially those who are already suffering from type II diabetes mellitus [137-142]. Risk factors for GDM non-Caucasian ethnic backgrounds, advanced age, obesity, intolerance to glucose, and a previous experience with GDM during pregnancy [139-143]. The pathophysiological factors of PCOS that predispose women to GDM are obesity, glucose intolerance, and type II diabetes [20, 144-146]. In a study examining the prevalence of GDM in women with PCOS, the authors determined that women with PCOS have twice the risk of developing GDM in comparison to women without PCOS after controlling for age, race, and the number of previous pregnancies [147]. This finding highlights the necessity for clinicians to counsel their PCOS patients about the development of the disease during their pregnancies and to provide screening.
Cancer
Although no definite correlation between cancer and PCOS has yet been established, women with PCOS are predisposed to cancers, in particular endometrial carcinoma (EC). It is considered conscientious practice to be aware of the relationship between PCOS and EC [148], especially since EC is the most common form of gynecological cancer in the United States [149]. High levels of unopposed estrogen, infertility, and symptoms of metabolic syndrome like obesity, glucose intolerance, and high blood pressure are all risk factors for EC [17, 150]. Persistent estrogen stimulation of the endometrial tissue combined with a lack of negative feedback or opposition by progesterone due to dysregulation of the menstrual cycle has been found to be a risk factor for EC as well [151]. Furthermore, oligomenorrheic women were found to have cancer genes that were activated in their endometrium earlier in life than in normally cycling women [152]. Other studies reported altered expression of genes that are related to EC in women with PCOS [153]. The elevated levels of androgens in women with PCOS have also been correlated with EC; women with EC had three times the levels of androgens than controls [154]. Additionally, women with EC had higher levels of testosterone and androstenedione in their ovarian veins than with women without the disease [155]. Furthermore, androgens were discovered to have stimulatory effects on EC cells [156, 157]. Because of their reduced fertility, women with PCOS are more likely to be nulliparous than the normal women, which is another factor relating to the development of EC [158]. Another hormone that is dysregulated in both PCOS and EC is LH. LH levels are elevated in 65% of women with PCOS and women with EC have an over proliferation of LH receptors, which suggests a possible correlation between the two diseases [159].
Impact of polycystic ovary syndrome on psychological state
Studies have also been conducted to assess the psychological state of women who are suffering from PCOS. The objective in these studies was to determine the effect PCOS has on quality of life or psychological morbidity [160]. Some studies have indicated that women with PCOS have a lower health-related quality of life (HRQoL), especially in regards to sex, self, and health [161]. Women with PCOS also have been determined to have higher levels of psychological distress than healthy controls [162] and a physiological morbidity of 62.4 vs. 26.4% in the control population [160]. The common symptoms of obesity and hirsuitism seen in PCOS contributed largely to the decreased self confidence and body image demonstrated in these women [160]. Women with a higher BMI were found to have a lower quality of life [160] and similarly, women with hirsutism indicated higher levels of social anxiety [163]. These findings are particularly noteworthy for clinicians working with young women with PCOS because adolescence is a sensitive developmental period [164]. Adolescent patients possibly experience negative effects later in life associated with the stress of living with the physical manifestations of the disease at such an early age. However, one study discovered that adolescent patients who received detailed information about the disease had a greater psychological quality of life than other, less informed patients [160]. Therefore, it is critical that clinicians serve as a reassuring and supportive source of information for their adolescent patients.
Lifestyle changes
Although the long-term consequences of PCOS are varied, many stem from the same underlying pathophysiological factors. Lifestyle changes can be utilized to prevent many of the diseases associated with PCOS. Therefore, it is critical to identify PCOS in adolescent girls to initiate these changes immediately and to prevent the onset and progression of other diseases such as type II diabetes, cardiovascular disease, and cancer. These diseases can be avoided by making healthy lifestyle choices such as exercising regularly and maintaining a nutritious, low-fat diet. In addition, it is recommended that women with PCOS be screened for the most common long-term consequences in order to determine their risk for developing these complications and/or detect them early. For example, adolescents with PCOS or high levels of androgens should be screened regularly with oral glucose tolerance test [136].
Genetics of polycystic ovary syndrome
Polycystic ovary syndrome is believed to have a complex genetic trait [165] in which a variety of genes interact with the environment to produce the disease [166]. Because of its varied pathophysiology and the fact that it shares many of its symptoms with other syndromes, it is necessary to rule out other genetic disorders such as 21-hydroxylase-deficient non-classic adrenal hyperplasia (NCAH) when diagnosing PCOS [167]. There are several models that have been proposed to explain the genetic nature of the disease: the “single-gene mendelian” model, the “multifactorial” model, and the “variable expression-single gene” model [9]. The “single-gene mendelian” model is based on the assumption that the mutation that causes PCOS is unique to the disease and is the result of a single-gene mutation. This would cause the disease to be inherited in a recessive or dominant manner [9]. The “multifactorial model” assumes that the changes that take place in PCOS are not specific to the disease; they may be implicated in other syndromes and already exist in the population, but in combination, they act to cause PCOS [9]. The “variable expression-single gene” model is a combination of the other two models. It proposes that PCOS is caused by a single-gene defect but the severity of the disease is modulated by other genetic or environmental factors [9].
Most studies of the genetic factors underlying PCOS have followed the candidate gene approach, which utilizes genetic markers such as single nucleotide polymorphisms (SNPs) [167]. Geneticists have examined pathways that are related to the pathophysiology of PCOS in order to identify candidate genes for further examination. These pathways include steroid production, folliculogenesis, weight gain, hormone function, and insulin function [9, 167].
Familial studies have also provided evidence to the genetic basis underlying PCOS. Some evidence suggests that the disease is more common in families than in the general population [86]. In a study examining the prevalence of PCOS in female relatives, 35% of premenopausal mothers and 40% of sisters had the disease [168]. These statistics provide a stark contrast with the prevalence of the disease in the general population, which is only 6 to 8% [2, 16]. A study of brothers of mothers with PCOS also revealed that they have elevated androgen levels [86]. Familial studies have also distinguished a genetic basis for insulin resistance and secretion dysfunction [167]. A study performed in the Australian population discovered that in families with a member with PCOS, almost 70% of the other members were resistant to insulin [169]. It was also found that b-cell dysfunction in PCOS may in part be determined by genetic inheritance [170].
The genetic basis of insulin dysregulation was also suggested by examination of cell function. The abnormal uptake and signaling responses of the cells of PCOS women may be due to dysregulation or mutations in the insulin-signaling pathway [171-173]. The regulation of androgens in women with PCOS appears to be unaffected by other hormones and feedback systems. This observation was demonstrated by an experiment in which theca cells removed from women with PCOS continued to secrete testosterone inde-pendently in culture [174].
Recent studies of candidate genes
Recent studies have identified new candidate genes for PCOS in the areas of androgen production and regulation and insulin regulation. Goodarzi et al. studied SRD5A1 and SRD5A2, which code for the isoforms of 5a-reductase, the enzyme which converts testosterone to dihydrotestosterone (DHT). Levels of this enzyme are frequently elevated in women with PCOS [175-179]. Transcripts of both of the genes were discovered in the ovary, and these may play a role in the arrest of follicle development. The study discovered that the genes had haplotypes related to an increased susceptibility to PCOS. Specifically, for the SRD5A1 gene, the TA haplotype was found in almost 15% of the PCOS population while only in 10% of controls. The SRD5A1 gene was also found to have a variant that was related to the severity of hirsutism. In addi- tion to a haplotype, which increased the risk of developing PCOS, gene SRD5A2 also had a haplotype that decreased the risk of PCOS [167].
Another study of androgen function in PCOS examined the action of the aromatase gene. It has been documented that decreased action of the aromatase enzyme, which converts androgens to estrogens, can lead to PCOS [180]. Furthermore, loss of function mutations in the aromatase gene (CYP19) have been observed in PCOS patients [181-183]. Excess levels of androgens in the ovaries were also linked to the stimulation of precocious pubarche [184, 185], a risk factor for PCOS [186]. A recent study by Petry et al. determined that the distal promoter region of the gene that codes for aromatase has genetic variations that are related to androgen levels and hyperandrogenism in young girls. Regulation of androgens was examined in a study of the variations in genes related to the control of dehydroepiandrosterone sulfate (DHEAS) – a marker for the production of adrenal androgen – in women with PCOS. Approximately 25% of women with PCOS have extremely high levels of DHEAS and the majority of PCOS patients have levels that are moderately elevated [187]. DHEA sulfotransferase is coded for by the gene SULT2A1 and it acts to add a sulfonate group to dehydroepiandrosterone (DHEA), a metabolite of andrenal androgen. Conversely, the sulfonate group of DHEAS is removed by steroid sulfatase (STS). Therefore, these genes are useful markers for examining dysregulation of androgen levels in patients with PCOS. Additionally, DHEAS levels in the body do not seem to be regulated by insulin or hormones suggesting that PCOS is under genetic control [188-192]. One study discovered that a haplotype and an SNP within the SULT2A1 gene were both related to lower levels of DHEAS in women with PCOS [193]. However, no association was found between variation in the STS gene and androgen levels in women with PCOS [193]. Furthermore, it seems that DHEAS levels are more influenced by genetic factors in women than in men [194]. The results from this study indicate that the variation in the gene SULT2A1 may not be a risk factor for the disease but could modulate the amount of excess adrenal androgens in PCOS [193].
A recently proposed mechanism for insulin resistance in PCOS has also been proposed involves abnormal phosphorylation after the binding of insulin to its receptor which causes changes in the signal transduction pathway. It appears that in insulin resistant women with PCOS, serine (Ser) residues were phosphorylated, impeding the activity of the tyrosine kinase activity, which is critical in the normal insulin receptor pathway [17]. Dysregulation of the insulin pathways contributes to the hyperandrogemia in women with PCOS. Excess levels of insulin can bind to type 1 IGF-1 (insulin growth factor-1) receptors, which increases the response of the thecal cells to LH and produces higher levels of androgens. Additionally, excess levels of insulin decrease the binding ability of SHBG, which also contributes to increased androgen levels in PCOS. High levels of insulin in the body can also prevent the synthesis of the protein that binds with IGF-1, which leads to elevated levels of IGF-1 and hyperandrogenemia [17].
Genetic approaches have also been employed to study ovarian function. A study comparing the pattern of gene expression in the ovary in normal women and those with PCOS found that there were 119 differentially expressed genes. Many of the identified genes were involved in cellular metabolism and control of gene expression, indicating that PCOS is a complex, multifactorial syndrome [195]. One gene that was found to be downregulated in PCOS women is NR4A1, which plays an integral role in the induction of apoptosis. It is believed that dysregulation of apoptosis factors in the ovary can lead to an imbalance in the cycles of germ cell death and follicular atresia. This dysregulation could lead to the accumulation of arrested follicles in the ovary. NR4A1 is mainly expressed by the thecal cells, which suggests that its downregulation in PCOS could be associated with the excess of the theca cells in PCOS women [196]. Another interesting gene identified in this study was the SET (SET translocation) gene, which is involved in the regulation and production of androgens controlled by P450c17. Abnormal expression of this gene may contribute to the high levels of androgens present in PCOS patients [195]. A comparison of the mRNA of oocytes between women with PCOS and healthy women found that PCOS oocytes have abnormalities in gene expression that could lead to reduced fertility. The 374 genes identified were involved in several areas essential to reproductive function such as chromosome alignment, the regulation of the cell cycle, and the function of the spindle [197]. Additionally, several genes that were categorized as “maternal-effect” genes, which help regulate zygote development [198], were expressed in elevated levels in the oocytes of women with PCOS. These abnormalities in the oocyte could lead to later complications in the development of the embryo [197].
Another mechanism proposed to explain the genetic background of PCOS is the follicle stimulating hormone-granulosa cell (FSH-GC) theory. This theory states that decreased levels of FSH diminish the action of cytochrome P450 aromatase in granulosa cells, causing the accumulation of androgens in the body. This could occur due to various impediments that cause abnormal expression of P450 aromatase such as problems in signaling and insufficient levels of FSH to stimulate the gene expression of P450 aromatase [199].
Limitations
It is important to note that many of the studies discussed in this paper were limited by small control populations and an insufficient number of genetic variants. In addition, the phenotype of PCOS is difficult to determine in all the groups that it affects: young girls, women, and men. Diagnosis is challenging because the phenotypes of the disease are compounded by other syndromes that have similar phenotypes and ethnic factors and the criteria for PCOS has not yet been standardized [167].
Conclusions
Polycystic ovarian syndrome is the most common endocrine disorder in women. Potential complications such as metabolic syndrome (MBS) manifest due to the hyperandrogenemia and insulin resistance associated with the syndrome. Adolescent PCOS may be characterized by earlier onset of hyperinsulinemia and insulin resistance. In adolescents, PCOS classically manifests post menarche, but it is not uncommon for patients to exhibit symptoms of the disease premenarche or in addition to precocious puberty. Review of the existing literature indicates that MS or, alternatively, individual metabolic risk factors may be present in PCOS patients and most importantly that MS may arise at a significantly younger age among PCOS women. The fact that a reproductive disorder like PCOS could have a significant metabolic impact on affected adolescents has generated medical interest on the mechanisms underlying the multiplicative sequelae of PCOS. Although obesity indisputably compounds the clinical course of women with PCOS, this appears to be just the tip of the iceberg. Insulin resistance and hyperinsulinemia have also been implicated as critical components due to their contribution to the pathophysiology and clinical presentation of both PCOS and MS. Hyper-androgenemia, the predominant endocrine hallmark of PCOS, has also been implicated as a contributing factor to the suggested interrelationship. Polycystic ovary syndrome is believed to have a complex genetic trait in which a variety of genes interact with the environment to produce the disease.
References

1. Xita N, Tsatsoulis A. Review: fetal programming of polycystic ovary syndrome by androgen excess: evidence from experimental, clinical, and genetic association studies. J Clin Endocrinol Metab 2006; 91: 1660-6.
2. Gambineri A, Pelusi C, Vicennati V, Pagotto U, Pasquali R. Obesity and the polycystic ovary syndrome. Int J Obes Relat Metab Disord 2002; 26: 883-96.
3. Jensterle M, Weber M, Pfeifer M, Prezelj J, Pfutzner A, Janez A. Assessment of insulin resistance in young women with polycystic ovary syndrome. Int J Gynaecol Obstet 2008; 102: 137-40.
4. Hoeger KM. Role of lifestyle modification in the management of polycystic ovary syndrome. Best Pract Res Clin Endocrinol Metab 2006; 20: 293-310.
5. Hashemipour M, Faghihimani S, Zolfaghary B, Hovsepian S, Ahmadi F, Haghighi S. Prevalence of polycystic ovary syndrome in girls aged 14-18 years in Isfahan, Iran. Horm Res 2004; 62: 278-82.
6. Diamanti-Kandarakis E, Christakou C, Palioura E, Kandaraki E, Livadas S. Does polycystic ovary syndrome start in childhood? Pediatr Endocrinol Rev 2008; 5: 904-11.
7. de Zegher F, Ibanez L. Prenatal growth restraint followed by catch-up of weight: a hyperinsulinemic pathway to polycystic ovary syndrome. Fertil Steril 2006; 86 Suppl 1: S4-5.
8. De Leo V, Musacchio MC, Morgante G, Piomboni P, Petraglia F. Metformin treatment is effective in obese teenage girls with PCOS. Hum Reprod 2006; 21: 2252-6.
9. Dasgupta S, Reddy BM. Present status of understanding on the genetic etiology of polycystic ovary syndrome. J Postgrad Med 2008; 54: 115-25.
10. Broekmans FJ, Knauff EA, Valkenburg O, Laven JS, Eijkemans MJ, Fauser BC. PCOS according to the Rotterdam consensus criteria: Change in prevalence among WHO-II anovulation and association with metabolic factors. BJOG 2006; 113: 1210-7.
11. Diamanti-Kandarakis E, Christakou C, Kandarakis H. Polycystic ovarian syndrome: the commonest cause of hyperandrogenemia in women as a risk factor for metabolic syndrome. Minerva Endocrinol 2007; 32: 35-47.
12. Khan U. Polycystic ovary syndrome in adolescents. J Pediatr Adolesc Gynecol 2007; 20: 101-4.
13. Wiksten-Almstromer M, Hirschberg AL, Hagenfeldt K. Menstrual disorders and associated factors among adolescent girls visiting a youth clinic. Acta Obstet Gynecol Scand 2007; 86: 65-72.
14. Stanley T, Misra M. Polycystic ovary syndrome in obese adolescents. Curr Opin Endocrinol Diabetes Obes 2008; 15: 30-6.
15. Creatsas G, Deligeoroglou E. Polycystic ovarian syndrome in adolescents. Curr Opin Obstet Gynecol 2007; 19: 420-6.
16. Strauss JF III, Barbieri RL. Reproductive Endocrinology: Physiology, Pathophysiology and Clinical Management. New York: W.B. Saunders Co.; 2004.
17. Johnson A, Brannian J, Hansen K. Current diagnosis and treatment of polycystic ovary syndrome. S D Med 2008; 61: 129-31.
18. Sawathiparnich P, Weerakulwattana L, Santiprabhob J, Likitmaskul S. Obese adolescent girls with polycystic ovary syndrome (PCOS) have more severe insulin resistance measured by HOMA-IR score than obese girls without PCOS. J Med Assoc Thai 2005; 88 Suppl 8: S33-7.
19. Gambineri A, Pelusi C, Genghini S, et al. Effect of flutamide and metformin administered alone or in combination in dieting obese women with polycystic ovary syndrome. Clin Endocrinol 2004; 60: 241-9.
20. Palmert MR, Gordon CM, Kartashov AI, Legro RS, Emans SJ, Dunaif A. Screening for abnormal glucose tolerance in adolescents with polycystic ovary syndrome. J Clin Endocrinol Metab 2002; 87: 1017-23.
21. Kirchengast S, Huber J. Body composition characteristics and body fat distribution in lean women with polycystic ovary syndrome. Hum Reprod 2001; 16: 1255-60.
22. Pasquali R, Casimirri F, Venturoli S, et al. Body fat distribution has weight-independent effects on clinical, hormonal, and metabolic features of women with polycystic ovary syndrome. Metabolism 1994; 43: 706-13.
23. Diamanti-Kandarakis E, Christakou CD, Kandaraki E, Alexandraki KI. Early onset adiposity: a pathway to polycystic ovary syndrome in adolescents? Hormones (Athens, Greece) 2007; 6: 210-7.
24. Silfen ME, Denburg MR, Manibo AM, et al. Early endocrine, metabolic, and sonographic characteristics of polycystic ovary syndrome (PCOS): comparison between nonobese and obese adolescents. J Clin Endocrinol Metab 2003; 88: 4682-8.
25. Weiss R, Dziura J, Burgert TS, et al. Obesity and the metabolic syndrome in children and adolescents. N Engl J Med 2004; 350: 2362-74.
26. Carmina E, Bucchieri S, Esposito A, et al. Abdominal fat quantity and distribution in women with polycystic ovary syndrome and extent of its relation to insulin resistance. J Clin Endocrinol Metab 2007; 92: 2500-5.
27. Marshall JC. Obesity in adolescent girls: is excess androgen the real bad actor? J Clin Endocrinol Metab 2006; 91: 393-5.
28. Pasquali R. Obesity and androgens: facts and perspectives. Fertil Steril 2006; 85: 1319-40.
29. Lewy VD, Danadian K, Witchel SF, Arslanian S. Early metabolic abnormalities in adolescent girls with polycystic ovarian syndrome. J Pediatr 2001; 138: 38-44.
30. Ehrmann DA. Polycystic ovary syndrome. N Engl J Med 2005; 352: 1223-36.
31. McCartney CR, Prendergast KA, Chhabra S, et al. The association of obesity and hyperandrogenemia during the pubertal transition in girls: obesity as a potential factor in the genesis of postpubertal hyperandrogenism. J Clin Endocrinol Metab 2006; 91: 1714-22.
32. Lukanova A, Lundin E, Micheli A, et al. Circulating levels of sex steroid hormones and risk of endometrial cancer in postmenopausal women. Int J Cancer 2004; 108: 425-32.
33. Morales AJ, Laughlin GA, Butzow T, Maheshwari H, Baumann G, Yen SS. Insulin, somatotropic, and luteinizing hormone axes in lean and obese women with polycystic ovary syndrome: common and distinct features. J Clin Endocrinol Metab 1996; 81: 2854-64.
34. Zidenberg N, Wright S. Care of the overweight adolescent including polycystic ovarian syndrome. Clin Obstet Gynecol 2008; 51: 249-56.
35. Nestler JE, Jakubowicz DJ. Decreases in ovarian cytochrome P450c17 alpha activity and serum free testosterone after reduction of insulin secretion in polycystic ovary syndrome. N Engl J Med 1996; 335: 617-23.
36. Rosenfield RL. Ovarian and adrenal function in polycystic ovary syndrome. Endocrinol Metab Clin North Am 1999; 28: 265-93.
37. Pasquali R, Gambineri A. Insulin-sensitizing agents in polycystic ovary syndrome. Eur J Endocrinol 2006; 154: 763-75.
38. Catteau-Jonard S, Pigny P, Reyss AC, Decanter C, Poncelet E, Dewailly D. Changes in serum anti-mullerian hormone level during low-dose recombinant follicular-stimulating hormone therapy for anovulation in polycystic ovary syndrome. J Clin Endocrinol Metab 2007; 92: 4138-43.
39. Grossman MP, Nakajima ST, Fallat ME, Siow Y. Mullerian- inhibiting substance inhibits cytochrome P450 aromatase activity in human granulosa lutein cell culture. Fertil Steril 2008; 89 (5 Suppl): 1364-70.
40. Wachs DS, Coffler MS, Malcom PJ, Chang RJ. Serum anti-mullerian hormone concentrations are not altered by acute administration of follicle stimulating hormone in polycystic ovary syndrome and normal women. J Clin Endocrinol Metab 2007; 92: 1871-4.
41. Leibel RL. The molecular genetics of the melanocortin pathway and energy homeostasis. Cell Metab 2006; 3: 79-81.
42. West C, Foster DL, Evans NP, Robinson J, Padmanabhan V. Intra-follicular activin availability is altered in prenatally-androgenized lambs. Mol Cell Endocrinol 2001; 185: 51-9.
43. Robinson JE, Forsdike RA, Taylor JA. In utero exposure of female lambs to testosterone reduces the sensitivity of the gonadotropin-releasing hormone neuronal network to inhibition by progesterone. Endocrinology 1999; 140: 5797-805.
44. Abbott DH, Barnett DK, Bruns CM, Dumesic DA. Androgen excess fetal programming of female reproduction: a developmental aetiology for polycystic ovary syndrome? Hum Reprod Update 2005; 11: 357-74.
45. Dumesic DA, Abbott DH, Eisner JR, Goy RW. Prenatal exposure of female rhesus monkeys to testosterone propionate increases serum luteinizing hormone levels in adulthood. Fertil Steril 1997; 67: 155-63.
46. Barnes RB, Rosenfield RL, Ehrmann DA, et al. Ovarian hyperandrogynism as a result of congenital adrenal virilizing disorders: evidence for perinatal masculinization of neuroendocrine function in women. J Clin Endocrinol Metabol 1994; 79: 1328-33.
47. Hague WM, Adams J, Rodda C, et al. The prevalence of polycystic ovaries in patients with congenital adrenal hyperplasia and their close relatives. Clin Endocrinol 1990; 33: 501-10.
48. Hogeveen KN, Cousin P, Pugeat M, Dewailly D, Soudan B, Hammond GL. Human sex hormone-binding globulin variants associated with hyperandrogenism and ovarian dysfunction. J Clin Investig 2002; 109: 973-81.
49. Morishima A, Grumbach MM, Simpson ER, Fisher C, Qin K. Aromatase deficiency in male and female siblings caused by a novel mutation and the physiological role of estrogens. J Clin Endocrinol Metabol 1995; 80: 3689-98.
50. Sir-Petermann T, Maliqueo M, Angel B, Lara HE, Perez-Bravo F, Recabarren SE. Maternal serum androgens in pregnant women with polycystic ovarian syndrome: possible implications in prenatal androgenization. Hum Reprod (Oxford, England) 2002; 17: 2573-9.
51. Nestler JE. Modulation of aromatase and P450 cholesterol side-chain cleavage enzyme activities of human placental cytotrophoblasts by insulin and insulin-like growth factor I. Endocrinology 1987; 121: 1845-52.
52. Nestler JE. Insulin and insulin-like growth factor-I stimulate the 3 beta-hydroxysteroid dehydrogenase activity of human placental cytotrophoblasts. Endocrinology 1989; 125: 2127-33.
53. Nestler JE. Insulin-like growth factor II is a potent inhibitor of the aromatase activity of human placental cytotrophoblasts. Endocrinology 1990; 127: 2064-70.
54. Blondeau B, Lesage J, Czernichow P, Dupouy JP, Breant B. Glucocorticoids impair fetal beta-cell development in rats. Am J Physiol 2001; 281: E592-9.
55. Mackenzie HS, Brenner BM. Fewer nephrons at birth: a missing link in the etiology of essential hypertension? Am J Kidney Dis 1995; 26: 91-8.
56. Tanaka Y, Kikuchi T, Nagasaki K, Hiura M, Ogawa Y, Uchiyama M. Lower birth weight and visceral fat accumulation are related to hyperinsulinemia and insulin resistance in obese Japanese children. Hypertens Res 2005; 28: 529-36.
57. Ibanez L, Lopez-Bermejo A, Callejo J, et al. Polycystic ovaries in nonobese adolescents and young women with ovarian androgen excess: relation to prenatal growth. J Clin Endocrinol Metabol 2008; 93: 196-9.
58. Ibanez L, Jaramillo A, Enriquez G, et al. Polycystic ovaries after precocious pubarche: relation to prenatal growth. Human reproduction (Oxford, England) 2007; 22: 395-400.
59. Ibanez L, Potau N, Francois I, de Zegher F. Precocious pubarche, hyperinsulinism, and ovarian hyperandrogenism in girls: relation to reduced fetal growth. J Clin Endocrinol Metabol 1998; 83: 3558-62.
60. Ibanez L, Valls C, Potau N, Marcos MV, de Zegher F. Polycystic ovary syndrome after precocious pubarche: ontogeny of the low-birthweight effect. Clin Endocrinol 2001; 55: 667-72.
61. Reinehr T, de Sousa G, Roth CL, Andler W. Androgens before and after weight loss in obese children. J Clin Endocrinol Metabol 2005; 90: 5588-95.
62. Strauss RS. Childhood obesity. Pediatr Clin North Am 2002; 49: 175-201.
63. Genazzani AR, Pintor C, Corda R. Plasma levels of gonadotropins, prolactin, thyroxine, and adrenal and gonadal steroids in obese prepubertal girls. J Clin Endocrinol Metabol 1978; 47: 974-9.
64. Leibel NI, Baumann EE, Kocherginsky M, Rosenfield RL. Relationship of adolescent polycystic ovary syndrome to parental metabolic syndrome. J Clin Endocrinol Metabol 2006; 91: 1275-83.
65. Nader S. Adrenarche and polycystic ovary syndrome: a tale of two hypotheses. J Pediatr Adolescent Gynecol 2007; 20: 353-60.
66. Blank SK, McCartney CR, Marshall JC. The origins and sequelae of abnormal neuroendocrine function in polycystic ovary syndrome. Hum Reprod Update 2006; 12: 351-61.
67. Gross KM, Matsumoto AM, Bremner WJ. Differential control of luteinizing hormone and follicle-stimulating hormone secretion by luteinizing hormone-releasing hormone pulse frequency in man. J Clin Endocrinol Metabol 1987; 64: 675-80.
68. Spratt DI, Finkelstein JS, Butler JP, Badger TM, Crowley WF Jr. Effects of increasing the frequency of low doses of gonadotropin-releasing hormone (GnRH) on gonadotropin secretion in GnRH-deficient men. J Clin Endocrinol Metabol 1987; 64: 1179-86.

69. Wildt L, Hausler A, Marshall G, et al. Frequency and amplitude of gonadotropin-releasing hormone stimulation and gonadotropin secretion in the rhesus monkey. Endocrinology 1981; 109: 376-85.
70. Kirk SE, Dalkin AC, Yasin M, Haisenleder DJ, Marshall JC. Gonadotropin-releasing hormone pulse frequency regulates expression of pituitary follistatin messenger ribonucleic acid: a mechanism for differential gonadotrope function. Endocrinology 1994; 135: 876-80.
71. Adams JM, Taylor AE, Schoenfeld DA, Crowley WF Jr, Hall JE. The midcycle gonadotropin surge in normal women occurs in the face of an unchanging gonadotropin- releasing hormone pulse frequency. J Clin Endocrinol Metabol 1994; 79: 858-64.
72. Filicori M, Santoro N, Merriam GR, Crowley WF Jr. Characterization of the physiological pattern of episodic gonadotropin secretion throughout the human menstrual cycle. J Clin Endocrinol Metabol 1986; 62: 1136-44.
73. Karsch FJ, Dierschke DK, Weick RF, Yamaji T, Hotchkiss J, Knobil E. Positive and negative feedback control by estrogen of luteinizing hormone secretion in the rhesus monkey. Endocrinology 1973; 92: 799-804.
74. McCartney CR, Gingrich MB, Hu Y, Evans WS, Marshall JC. Hypothalamic regulation of cyclic ovulation: evidence that the increase in gonadotropin-releasing hormone pulse frequency during the follicular phase reflects the gradual loss of the restraining effects of progesterone. J Clin Endocrinol Metabol 2002; 87: 2194-200.
75. Messinis IE. Ovarian feedback, mechanism of action and possible clinical implications. Hum Reprod Update 2006; 12: 557-71.
76. Waldstreicher J, Santoro NF, Hall JE, Filicori M, Crowley WF Jr. Hyperfunction of the hypothalamic-pituitary axis in women with polycystic ovarian disease: indirect evidence for partial gonadotroph desensitization. J Clin Endocrinol Metabol 1988; 66: 165-72.
77. Taylor AE, McCourt B, Martin KA, et al. Determinants of abnormal gonadotropin secretion in clinically defined women with polycystic ovary syndrome. J Clin Endocrinol Metabol 1997; 82: 2248-56.
78. Kawadzki J, Dunaif A, Givens JR, Haseltine F, Merriam GR. Diagnostic Criteria for Polycystic Ovary Syndrome: a rational approach. In: Anonymous, editor. Polycystic Ovary Syndrome. Cambridge, Mass: Blackwell Scientific 1992; 377.
79. Velazquez EM, Mendoza S, Hamer T, Sosa F, Glueck CJ. Metformin therapy in polycystic ovary syndrome reduces hyperinsulinemia, insulin resistance, hyperandrogenemia, and systolic blood pressure, while facilitating normal menses and pregnancy. Metabol Clin Experiment 1994; 43: 647-54.
80. Velazquez EM, Mendoza SG, Wang P, Glueck CJ. Metformin therapy is associated with a decrease in plasma plasminogen activator inhibitor-1, lipoprotein (a), and immunoreactive insulin levels in patients with the polycystic ovary syndrome. Metabol Clin Experiment 1997; 46: 454-7.
81. Acien P, Quereda F, Matallin P, et al. Insulin, androgens, and obesity in women with and without polycystic ovary syndrome: a heterogeneous group of disorders. Fertil Steril 1999; 72: 32-40.
82. Glueck CJ, Wang P, Fontaine R, Tracy T, Sieve-Smith L. Metformin-induced resumption of normal menses in 39 of 43 (91%) previously amenorrheic women with the polycystic ovary syndrome. Metabol Clin Experiment 1999; 48: 511-9.
83. Glueck CJ, Wang P, Fontaine R, Tracy T, Sieve-Smith L. Metformin to restore normal menses in oligo-amenorrheic teenage girls with polycystic ovary syndrome (PCOS). J Adolesc Health 2001; 29: 160-9.
84. Glueck CJ, Wang P, Goldenberg N, Sieve-Smith L. Pregnancy outcomes among women with polycystic ovary syndrome treated with metformin. Hum Reprod (Oxford, England) 2002; 17: 2858-64.
85. Glueck CJ, Streicher P, Wang P. Treatment of polycystic ovary syndrome with insulin-lowering agents. Exp Opin Pharmacother 2002; 3: 1177-89.
86. Legro RS. Detection of insulin resistance and its treatment in adolescents with polycystic ovary syndrome. Journal of pediatric endocrinology and metabolism. JPEM 2002; 15 Suppl 5: 1367-78.
87. Morin-Papunen L, Rautio K, Ruokonen A, Hedberg P, Puukka M, Tapanainen JS. Metformin reduces serum C-reactive protein levels in women with polycystic ovary syndrome. J Clin Endocrinol Metabol 2003; 88: 4649-54.
88. Glueck CJ, Dharashivkar S, Wang P, et al. Obesity and extreme obesity, manifest by ages 20-24 years, continuing through 32-41 years in women, should alert physicians to the diagnostic likelihood of polycystic ovary syndrome as a reversible underlying endocrinopathy. Eur J Obstet Gynecol Reprod Biol 2005; 122: 206-12.
89. Glueck CJ, Goldenberg N, Pranikoff J, Loftspring M, Sieve L, Wang P. Height, weight, and motor-social development during the first 18 months of life in 126 infants born to 109 mothers with polycystic ovary syndrome who conceived on and continued metformin through pregnancy. Hum Reprod (Oxford, England) 2004; 19: 1323-30.
90. Glueck CJ, Goldenberg N, Streicher P, Wang P. Metformin and gestational diabetes. Cur Diabet Reports 2003; 3: 303-12.
91. Legro RS. Diagnostic criteria in polycystic ovary syndrome. Semin Reproduct Med 2003; 21: 267-75.
92. Moran LJ, Noakes M, Clifton PM, Tomlinson L, Galletly C, Norman RJ. Dietary composition in restoring reproductive and metabolic physiology in overweight women with polycystic ovary syndrome. J Clin Endocrinol Metabol 2003; 88: 812-9.
93. Seli E, Duleba AJ. Treatment of PCOS with metformin and other insulin-sensitizing agents. Cur Diabet Reports 2004; 4: 69-75.
94. Carr DB, Utzschneider KM, Hull RL, et al. Intra abdominal fat is a major determinant of the National Cholesterol Education Program Adult Treatment Panel III criteria for the metabolic syndrome. Diabetes 2004; 53: 2087-94.
95. Coviello AD, Legro RS, Dunaif A. Adolescent girls with polycystic ovary syndrome have an increased risk of the metabolic syndrome associated with increasing androgen levels independent of obesity and insulin resistance. J Clin Endocrinol Metabol 2006; 91: 492-7.
96. Katzmarzyk PT, Srinivasan SR, Chen W, Malina RM, Bouchard C, Berenson GS. Body mass index, waist circumference, and clustering of cardiovascular disease risk factors in a biracial sample of children and adolescents. Pediatrics 2004; 114: e198-205.
97. Berenson GS, Srnivasan SR; Bogalusa Heart Study G. Cardiovascular risk factors in youth with implications for aging: the Bogalusa Heart Study. Neurobiol Aging 2005; 26: 303-7.
98. Apridonidze T, Essah PA, Iuorno MJ, Nestler JE. Prevalence and characteristics of the metabolic syndrome in women with polycystic ovary syndrome. J Clin Endocrinol Metabol 2005; 90: 1929-35.
99. Dieudonne MN, Pecquery R, Boumediene A, Leneveu MC, Giudicelli Y. Androgen receptors in human preadipocytes and adipocytes: regional specificities and regulation by sex steroids. Am J Physiol 1998; 274: C1645-52.
100. Ibanez L, Ferrer A, Ong K, Amin R, Dunger D, de Zegher F. Insulin sensitization early after menarche prevents progression from precocious pubarche to polycystic ovary syndrome. J Pediatr 2004; 144: 23-9.
101. Cook S, Weitzman M, Auinger P, Nguyen M, Dietz WH. Prevalence of a metabolic syndrome phenotype in adolescents: findings from the third National Health and Nutrition Examination Survey, 1988-1994. Arch Pediatr Adolescent Med 2003; 57: 821-7.
102. de Ferranti SD, Gauvreau K, Ludwig DS, Neufeld EJ, Newburger JW, Rifai N. Prevalence of the metabolic syndrome in American adolescents: findings from the Third National Health and Nutrition Examination Survey. Circulation 2004; 110: 2494-7.
103. Neuschwander-Tetri BA. Fatty liver and the metabolic syndrome. Cur Opin Gastroenterol 2007; 23: 193-8.
104. Farrell GC, Larter CZ. Nonalcoholic fatty liver disease: from steatosis to cirrhosis. Hepatology (Baltimore, Md) 2006; 43 (2 Suppl 1): S99-112.
105. Adams LA, Angulo P, Lindor KD. Nonalcoholic fatty liver disease. Can Med Ass J 2005; 172: 899-905.
106. Cerda C, Perez-Ayuso RM, Riquelme A, et al. Nonalcoholic fatty liver disease in women with polycystic ovary syndrome. J Hepatol 2007; 47: 412-7.
107. McCullough AJ. The clinical features, diagnosis and natural history of nonalcoholic fatty liver disease. Clin Liver Dis 2004; 8: 521-33.
108. Mendez-Sanchez N, Arrese M, Zamora-Valdes D, Uribe M. Current concepts in the pathogenesis of nonalcoholic fatty liver disease. Liver Int 2007; 27: 423-33.
109. Ahima RS. Insulin resistance: cause or consequence of nonalcoholic steatohepatitis? Gastroenterology 2007; 132: 444-6.
110. Agarwal N, Sharma BC. Insulin resistance and clinical aspects of non-alcoholic steatohepatitis (NASH). Hepatol Res 2005; 33: 92-6.
111. Diamanti-Kandarakis E, Papavassiliou AG. Molecular mechanisms of insulin resistance in polycystic ovary syndrome. Trends Molecul Med 2006; 12: 324-32.
112. Neuschwander-Tetri BA, Caldwell SH. Nonalcoholic steatohepatitis: summary of an AASLD Single Topic Conference. Hepatology (Baltimore, Md) 2003; 37: 1202-19.
113. Talbott EO, Zborowski JV, Rager JR, Boudreaux MY, Edmundowicz DA, Guzick DS. Evidence for an association between metabolic cardiovascular syndrome and coronary and aortic calcification among women with polycystic ovary syndrome. J Clin Endocrinol Metabol 2004; 89: 5454-61.
114. Raitakari OT, Juonala M, Kahonen M, et al. Cardiovascular risk factors in childhood and carotid artery intima-media thickness in adulthood: the Cardiovascular Risk in Young Finns Study. JAMA 2003; 290: 2277-83.
115. Oren A, Vos LE, Uiterwaal CS, Gorissen WH, Grobbee DE, Bots ML. Change in body mass index from adolescence to young adulthood and increased carotid intima-media thickness at 28 years of age: the Atherosclerosis Risk in Young Adults study. Int J Obes Relat Metab Disord 2003; 27: 1383-90.
116. Meyer C, McGrath BP, Teede HJ. Overweight women with polycystic ovary syndrome have evidence of subclinical cardiovascular disease. J Clin Endocrinol Metabol 2005; 90: 5711-6.
117. Battaglia C, Mancini F, Cianciosi A, et al. Vascular risk in young women with polycystic ovary and polycystic ovary syndrome. Obstetrics Gynecol 2008; 111: 385-95.
118. Boushey CJ, Beresford SA, Omenn GS, Motulsky AG. A quantitative assessment of plasma homocysteine as a risk factor for vascular disease. Probable benefits of increasing folic acid intakes. JAMA 1995; 274: 1049-57.
119. Yarali H, Yildirir A, Aybar F, et al. Diastolic dysfunction and increased serum homocysteine concentrations may contribute to increased cardiovascular risk in patients with polycystic ovary syndrome. Fertil Steril 2001; 76: 511-6.
120. Tan MH, Johns D, Glazer NB. Pioglitazone reduces atherogenic index of plasma in patients with type 2 diabetes. Clin Chem 2004; 50: 1184-8.
121. Karahalios DG, Rekate HL, Khayata MH, Apostolides PJ. Elevated intracranial venous pressure as a universal mechanism in pseudotumor cerebri of varying etiologies. Neurology 1996; 46: 198-202.
122. Sugerman HJ, DeMaria EJ, Felton WL 3rd, Nakatsuka M, Sismanis A. Increased intra-abdominal pressure and cardiac filling pressures in obesity-associated pseudotumor cerebri. Neurology 1997; 49: 507-11.
123. Glueck CJ, Golnik KC, Aregawi D, Goldenberg N, Sieve L, Wang P. Changes in weight, papilledema, headache, visual field, and life status in response to diet and metformin in women with idiopathic intracranial hypertension with and without concurrent polycystic ovary syndrome or hyperinsulinemia. Translat Res 2006; 148: 215-22.
124. Friedman DI, Jacobson DM. Idiopathic intracranial hypertension. J Neuroophthalmol 2004; 24: 138-45.
125. Friedman DI. Pseudotumor cerebri. Neurol Clin 2004; 22: 99-131, vi.
126. Friedman DI, Jacobson DM. Diagnostic criteria for idiopathic intracranial hypertension. Neurology 2002; 59: 1492-5.
127. Tang RA, Dorotheo EU, Schiffman JS, Bahrani HM. Medical and surgical management of idiopathic intracranial hypertension in pregnancy. Cur Neurol Neurosci Reports 2004; 4: 398-409.
128. Binder DK, Horton JC, Lawton MT, McDermott MW. Idiopathic intracranial hypertension. Neurosurgery 2004; 54: 538-51; discussion 51-2.
129. Franks S, Gilling-Smith C, Watson H, Willis D. Insulin action in the normal and polycystic ovary. Endocrinol Metabol Clin North Am 1999; 28: 361-78.
130. Dunaif A. Insulin resistance and the polycystic ovary syndrome: mechanism and implications for pathogenesis. Endocrine Rev 1997; 18: 774-800.
131. Dunaif A, Graf M, Mandeli J, Laumas V, Dobrjansky A. Characterization of groups of hyperandrogenic women with acanthosis nigricans, impaired glucose tolerance, and/or hyperinsulinemia. J Clin Endocrinol Metabol 1987; 65: 499-507.
132. Wakisaka M, Nagamachi S, Inoue K, Morotomi Y, Nunoi K, Fujishima M. Reduced regional cerebral blood flow in aged noninsulin-dependent diabetic patients with no history of cerebrovascular disease: evaluation by N-isopropyl-123I- p-iodoamphetamine with single-photon emission computed tomography. J Diabetic Complicat 1990; 4: 170-4.
133. Boudreaux MY, Talbott EO, Kip KE, Brooks MM, Witchel SF. Risk of T2DM and impaired fasting glucose among PCOS subjects: results of an 8-year follow-up. Cur Diabetes Reports 2006; 6: 77-83.
134. Legro RS, Gnatuk CL, Kunselman AR, Dunaif A. Changes in glucose tolerance over time in women with polycystic ovary syndrome: a controlled study. J Clin Endocrinol Metabol 2005; 90: 3236-42.
135. Norman RJ, Masters L, Milner CR, Wang JX, Davies MJ. Relative risk of conversion from normoglycaemia to impaired glucose tolerance or non-insulin dependent diabetes mellitus in polycystic ovarian syndrome. Hum Reprod (Oxford, England) 2001; 16: 1995-8.
136. Talbott EO, Zborowski JV, Rager JR, Kip KE, Xu X, Orchard TJ. Polycystic ovarian syndrome (PCOS): a significant contributor to the overall burden of type 2 diabetes in women. J Wom Health 2007; 16: 191-7.
137. Ferrara A, Kahn HS, Quesenberry CP, Riley C, Hedderson MM. An increase in the incidence of gestational diabetes mellitus: Northern California, 1991-2000. Obstet Gynecol 2004; 103: 526-33.
138. Dabelea D, Snell-Bergeon JK, Hartsfield CL, Bischoff KJ, Hamman RF, McDuffie RS. Increasing prevalence of gestational diabetes mellitus (GDM) over time and by birth cohort: Kaiser Permanente of Colorado GDM Screening Program. Diabetes Care 2005; 28: 579-84.
139. Gestational diabetes mellitus. Diabetes Care 2000; 23 Suppl 1: S77-9.
140. Dooley SL, Metzger BE, Cho NH. Gestational diabetes mellitus. Influence of race on disease prevalence and perinatal outcome in a U.S. population. Diabetes 1991; 40 Suppl 2: 25-9.
141. King H. Epidemiology of glucose intolerance and gestational diabetes in women of childbearing age. Diabetes Care 1998; 21 Suppl 2: B9-13.
142. Kjos SL, Buchanan TA. Gestational diabetes mellitus. N Engl J Med 1999; 341: 1749-56.
143. Solomon CG, Willett WC, Carey VJ, et al. A prospective study of pregravid determinants of gestational diabetes mellitus. JAMA 1997; 278: 1078-83.
144. Legro RS, Gnatuk CL, Kunselman AR, Dunaif A. Changes in glucose tolerance over time in women with polycystic ovary syndrome: a controlled study. J Clin Endocrinol Metab 2005; 90: 3236-42.
145. Legro RS, Kunselman AR, Dodson WC, Dunaif A. Prevalence and predictors of risk for type 2 diabetes mellitus and impaired glucose tolerance in polycystic ovary syndrome: a prospective, controlled study in 254 affected women. J Clin Endocrinol Metab 1999; 84: 165-9.
146. Solomon CG, Hu FB, Dunaif A, et al. Long or highly irregular menstrual cycles as a marker for risk of type 2 diabetes mellitus. JAMA 2001; 286: 2421-6.
147. Lo JC, Feigenbaum SL, Escobar GJ, Yang J, Crites YM, Ferrara A. Increased prevalence of gestational diabetes mellitus among women with diagnosed polycystic ovary syndrome: a population-based study. Diabetes Care 2006; 29: 1915-7.
148. Hardiman P, Pillay OC, Atiomo W. Polycystic ovary syndrome and endometrial carcinoma. Lancet 2003; 361: 1810-2.
149. Jemal A, Thomas A, Murray T, Thun M. Cancer statistics, 2002. CA Cancer J Clin 2002; 52: 23-47.
150. Navaratnarajah R, Pillay OC, Hardiman P. Polycystic ovary syndrome and endometrial cancer. Semin Reprod Med 2008; 26: 62-71.
151. Giangrande PH, McDonnell DP. The A and B isoforms of the human progesterone receptor: two functionally different transcription factors encoded by a single gene. Recent Prog Horm Res 1999; 54: 291-313.
152. Pillay OC, Te Fong LF, Crow JC, et al. The association between polycystic ovaries and endometrial cancer. Hum Reprod 2006; 21: 924-9.
153. MacLaughlan SD, Palomino WA, Mo B, Lewis TD, Lininger RA, Lessey BA. Endometrial expression of Cyr61: a marker of estrogenic activity in normal and abnormal endometrium. Obstet Gynecol 2007; 110: 146-54.
154. Lukanova A, Lundin E, Micheli A, et al. Circulating levels of sex steroid hormones and risk of endometrial cancer in postmenopausal women. Int J Cancer 2004; 108: 425-32.
155. Nagamani M, Hannigan EV, Dillard EA Jr, Van Dinh T. Ovarian steroid secretion in postmenopausal women with and without endometrial cancer. J Clin Endocrinol Metab 1986; 62: 508-12.
156. Tada A, Sasaki H, Nakamura J, Yoshihama M, Terashima Y. Aromatase activity and the effect of estradiol and testosterone on DNA synthesis in endometrial carcinoma cell lines. J Steroid Biochem Mol Biol 1993; 44: 661-6.
157. Apparao KB, Lovely LP, Gui Y, Lininger RA, Lessey BA. Elevated endometrial androgen receptor expression in women with polycystic ovarian syndrome. Biol Reprod 2002; 66: 297-304.
158. Schmeler KM, Soliman PT, Sun CC, Slomovitz BM, Gershenson DM, Lu KH. Endometrial cancer in young, normal-weight women. Gynecol Oncol 2005; 99: 388-92.
159. Konishi I, Koshiyama M, Mandai M, et al. Increased expression of LH/hCG receptors in endometrial hyperplasia and carcinoma in anovulatory women. Gynecol Oncol 1997; 65: 273-80.
160. Ching HL, Burke V, Stuckey BG. Quality of life and psychological morbidity in women with polycystic ovary syndrome: body mass index, age and the provision of patient information are significant modifiers. Clin Endocrinol 2007; 66: 373-9.
161. Weiner CL, Primeau M, Ehrmann DA. Androgens and mood dysfunction in women: comparison of women with polycystic ovarian syndrome to healthy controls. Psychosom Med 2004; 66: 356-62.
162. Esler DM, Travers CA, Guttikonda K, Dixon A, Lewis PR. The psychosocial experience of women with PCOS – a case control study. Australian Fam Physician 2007; 36: 965-7.
163. Sonino N, Fava GA, Mani E, Belluardo P, Boscaro M. Quality of life of hirsute women. Postgraduate Med J 1993; 69: 186-9.
164. McKelvey RS, Pfaff JJ, Acres JG. The relationship between chief complaints, psychological distress, and suicidal ideation in 15-24-year-old patients presenting to general practitioners. Med J Australia 2001; 175: 550-2.
165. Legro RS. The genetics of obesity. Lessons for polycystic ovary syndrome. Ann N Y Acad Sci 2000; 900: 193-202.
166. King RA, Rotter JI, Motulsky AG. The Genetic Basis of Common Diseases. Anonymous, editor. New York: Oxford University Press; 2002.
167. Goodarzi MO, Azziz R. Diagnosis, epidemiology, and genetics of the polycystic ovary syndrome. Best Pract Res Clin Endocrinol Metab 2006; 20: 193-205.
168. Kahsar-Miller MD, Nixon C, Boots LR, Go RC, Azziz R. Prevalence of polycystic ovary syndrome (PCOS) in first-degree relatives of patients with PCOS. Fertil Steril 2001; 75: 53-8.
169. Norman RJ, Masters S, Hague W. Hyperinsulinemia is common in family members of women with polycystic ovary syndrome. Fertil Steril 1996; 66: 942-7.
170. Colilla S, Cox NJ, Ehrmann DA. Heritability of insulin secretion and insulin action in women with polycystic ovary syndrome and their first degree relatives. J Clin Endocrinol Metab 2001; 86: 2027-31.
171. Dunaif A, Wu X, Lee A, Diamanti-Kandarakis E. Defects in insulin receptor signaling in vivo in the polycystic ovary syndrome (PCOS). Am J Physiol Endocrinol Metabol 2001; 281: E392-9.
172. Rosenbaum D, Haber RS, Dunaif A. Insulin resistance in polycystic ovary syndrome: decreased expression of GLUT-4 glucose transporters in adipocytes. Am J Physiol 1993; 264: E197-202.
173. Ciaraldi TP, el-Roeiy A, Madar Z, Reichart D, Olefsky JM, Yen SS. Cellular mechanisms of insulin resistance in polycystic ovarian syndrome. J Clin Endocrinol Metab 1992; 75: 577-83.
174. Nelson VL, Legro RS, Strauss JF 3rd, McAllister JM. Augmented androgen production is a stable steroidogenic phenotype of propagated theca cells from polycystic ovaries. Molecul Endocrinol (Baltimore, Md) 1999; 13: 946-57.
175. Jakimiuk AJ, Weitsman SR, Magoffin DA. 5alpha-reductase activity in women with polycystic ovary syndrome. J Clin Endocrinol Metab 1999; 84: 2414-8.
176. Stewart PM, Shackleton CH, Beastall GH, Edwards CR. 5 alpha-reductase activity in polycystic ovary syndrome. Lancet 1990; 335: 431-3.
177. Tsilchorozidou T, Honour JW, Conway GS. Altered cortisol metabolism in polycystic ovary syndrome: insulin enhances 5alpha-reduction but not the elevated adrenal steroid production rates. J Clin Endocrinol Metabol 2003; 88: 5907-13.
178. Fassnacht M, Schlenz N, Schneider SB, Wudy SA, Allolio B, Arlt W. Beyond adrenal and ovarian androgen generation: Increased peripheral 5 alpha-reductase activity in women with polycystic ovary syndrome. J Clin Endocrinol Metab 2003; 88: 2760-6.
179. Chin D, Shackleton C, Prasad VK, et al. Increased 5alpha-reductase and normal 11betahydroxysteroid dehydrogenase metabolism of C19 and C21 steroids in a young population with polycystic ovarian syndrome. J Pediatr Endocrinol Metab 2000; 13: 253-9.
180. Gabrilove JL. The pathogenesis of the polycystic ovary syndrome: a hypothesis. Endocr Pract 2002; 8: 127-32.
181. Harada N, Ogawa H, Shozu M, Yamada K. Genetic studies to characterize the origin of the mutation in placental aromatase deficiency. Am J Hum Genet 1992; 51: 666-72.
182. Conte FA, Grumbach MM, Ito Y, Fisher CR, Simpson ER. A syndrome of female pseudohermaphrodism, hypergonadotropic hypogonadism, and multicystic ovaries associated with missense mutations in the gene encoding aromatase (P450arom). J Clin Endocrinol Metab 1994; 78: 1287-92.
183. Belgorosky A, Pepe C, Marino R, et al. Hypothalamic pituitary-ovarian axis during infancy, early and late prepuberty in an aromatase-deficient girl who is a compound heterocygote for two new point mutations of the CYP19 gene. J Clin Endocrinol Metabol 2003; 88: 5127-31.
184. Ibanez L, Potau N, Virdis R, et al. Postpubertal outcome in girls diagnosed of premature pubarche during childhood: increased frequency of functional ovarian hyperandrogenism. J Clin Endocrinol Metab 1993; 76: 1599-603.
185. Ibanez L, Dimartino-Nardi J, Potau N, Saenger P. Premature adrenarche – normal variant or forerunner of adult disease? Endocr Rev 2000; 21: 671-96.
186. Ibanez L, Potau N, Francois I, de Zegher F. Precocious pubarche, hyperinsulinism, and ovarian hyperandrogenism in girls: relation to reduced fetal growth. J Clin Endocrinol Metabol 1998; 83: 3558-62.
187. Kumar A, Woods KS, Bartolucci AA, Azziz R. Prevalence of adrenal androgen excess in patients with the polycystic ovary syndrome (PCOS). Clin Endocrinol 2005; 62: 644-9.
188. Azziz R, Gay FL, Potter SR, Bradley E Jr, Boots LR. The effects of prolonged hypertestosteronemia on adrenocortical biosynthesis in oophorectomized women. J Clin Endocrinol Metab 1991; 72: 1025-30. 189. Azziz R, Rittmaster RS, Fox LM, Bradley EL Jr, Potter HD, Boots LR. Role of the ovary in the adrenal androgen excess of hyperandrogenic women. Fertil Steril 1998; 69: 851-9.
190. Slayden SM, Crabbe L, Bae S, Potter HD, Azziz R, Parker CR Jr. The effect of 17 beta-estradiol on adrenocortical sensitivity, responsiveness, and steroidogenesis in postmenopausal women. J Clin Endocrinol Metab 1998; 83: 519-24.
191. Farah-Eways L, Reyna R, Knochenhauer ES, Bartolucci AA, Azziz R. Glucose action and adrenocortical biosynthesis in women with polycystic ovary syndrome. Fertil Steril 2004; 81: 120-5.
192. Hines GA, Smith ER, Azziz R. Influence of insulin and testosterone on adrenocortical steroidogenesis in vitro: preliminary studies. Fertil Steril 2001; 76: 730-5.
193. Goodarzi MO, Antoine HJ, Azziz R. Genes for enzymes regulating dehydroepiandrosterone sulfonation are associated with levels of dehydroepiandrosterone sulfate in polycystic ovary syndrome. J Clin Endocrinol Metab 2007; 92: 2659-64.
194. Rice T, Sprecher DL, Borecki IB, Mitchell LE, Laskarzewski PM, Rao DC. The Cincinnati Myocardial Infarction and Hormone Family Study: family resemblance for dehydroepiandrosterone sulfate in control and myocardial infarction families. Metabol Clin Experimental 1993; 42: 1284-90.
195. Diao FY, Xu M, Hu Y, et al. The molecular characteristics of polycystic ovary syndrome (PCOS) ovary defined by human ovary cDNA microarray. J Molecul Endocrinol 2004; 33: 59-72.
196. Park JI, Park HJ, Choi HS, Lee K, Lee WK, Chun SY. Gonadotropin regulation of NGFI-B messenger ribonucleic acid expression during ovarian follicle development in the rat. Endocrinology 2001; 142: 3051-9.
197. Wood JR, Dumesic DA, Abbott DH, Strauss JF 3rd. Molecular abnormalities in oocytes from women with polycystic ovary syndrome revealed by microarray analysis. J Clin Endocrinol Metab 2007; 92: 705-13.
198. Amleh A, Dean J. Mouse genetics provides insight into folliculogenesis, fertilization and early embryonic development. Hum Reprod Update 2002; 8: 395-403.
199. Odunsi K, Kidd KK. A paradigm for finding genes for a complex human trait: polycystic ovary syndrome and follistatin. Proc Natl Acad Sci U S A 1999; 96: 8315-7.
Copyright: © 2009 Termedia & Banach. This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International (CC BY-NC-SA 4.0) License (http://creativecommons.org/licenses/by-nc-sa/4.0/), allowing third parties to copy and redistribute the material in any medium or format and to remix, transform, and build upon the material, provided the original work is properly cited and states its license.
Quick links
© 2024 Termedia Sp. z o.o.
Developed by Bentus.