Open access peer-reviewed chapter

Endometrial Receptivity in Patients with Polycystic Ovary Syndrome

Written By

Alice Albu and Dragoș Albu

Submitted: 02 March 2021 Reviewed: 28 May 2021 Published: 24 June 2021

DOI: 10.5772/intechopen.98624

From the Edited Volume

Infertility and Assisted Reproduction

Edited by Wei-Hua Wang

Chapter metrics overview

601 Chapter Downloads

View Full Metrics

Abstract

Polycystic ovary syndrome (PCOS) is a frequent disorder affecting women of reproductive age characterized by infertility. Affected endometrial receptivity seems to contribute to decreased fertility of these patients as suggested by several studies. Understanding the mechanism behind this reduced endometrial receptivity could contribute to discovery of new therapeutic targets for infertility of PCOS. The aim of the paper is to review the current data regarding endometrial receptivity in PCOS patients, the potential mechanisms involved with particular focus on recent findings as the impact of gut microbiota on endometrium, the relationship between vitamin D and endometrial receptivity and the different impact of letrozole and clomiphene citrate on endometrial receptivity in infertile PCOS women.

Keywords

  • polycystic ovary syndrome
  • endometrial receptivity
  • endometrium
  • implantation
  • pregnancy

1. Introduction

Polycystic ovary syndrome (PCOS) is the most common endocrine disorder in women of reproductive age, having a prevalence of 8 to 13% and of 21% in high-risk groups [1]. Moreover, it is a leading cause of female infertility [2, 3] and the most common cause of anovulatory infertility [4]. A systematic review from 2020 [5] found that there is significant variation in prevalence probably according to ethnic background and design of the published studies, but also to diagnosis criteria used to identify the disease. Thus, they found that the reported prevalence of PCOS vary between 2,2% and 15–20%, with the studies using the Rotterdam criteria reporting the highest prevalence [5].

PCOS has significant consequences on the women health, being associated with infertility, menstrual irregularities, metabolic abnormalities, cardiovascular risk and psychological disturbances [6] and, therefore, impairing the quality of life. The latest guidelines [7] recommend the use of Rotterdam Consensus criteria for PCOS diagnosis, which assumes that two out of the following three features are present: oligo- or anovulation, hyperandrogenism (clinical or biochemical) and polycystic ovaries [8]. The use of these criteria generates several clinical phenotypes with variate impact on reproductive potential and metabolic profile, with some of them diagnosed with difficulty due to a scarce clinical picture. Therefore, PCOS can be a challenging disorder in the reproductive medicine practice.

Hyperandrogenism is a key feature of PCOS, being the result of increased production of both ovarian and adrenal androgens. Ovarian over-secretion of androgens is the consequence of LH stimulation and also the action of high insulin levels on insulin receptors from the ovarian theca cells. Moreover, bioavailability of androgens is increased due to insulin effect to reduce the hepatic production of sex-hormone binding globulin.

Advertisement

2. Fertility in patients with polycystic ovary syndrome

PCOS is considered the most common cause of anovulation, being responsible for 70% of cases of anovulatory infertility [9]. Infertility is a significant complaint among women with PCOS, being reported in 72% of women with PCOS compared with 16% in women without PCOS. However, it seems that the number of children of women with PCOS is similar with those without, suggesting that treatment for infertility is effective [10].

It was also showed that 44% of women with unexplained infertility are probable PCOS cases, underlying that the subtle clinical phenotypes in some of the patients can be the cause of misdiagnosis and, therefore, inappropriate diagnosis and treatment [11].

It seems that the criteria used for diagnosis can also impact the prevalence of infertility which is higher in women with polycystic ovary morphology in patients diagnosed according to Androgen Excess Society criteria (21,7%), while in patients diagnosed according to Rotterdam criteria infertility is present in only 6% of them [5].

Older studies report that 78% of infertile women with PCOS respond to clomiphene citrate (CC) administration [12], with only the remaining 22% requiring alternative therapies, suggesting that anovulation is not the only cause of infertility in PCOS patients. Indeed, even after restoration of ovulation, PCOS patients still have reduced cumulative pregnancy rate and higher rates of implantation failure [13]. Even in cycles with excellent embryos selected for transfer, the success of in vitro fertilization in PCOS patients remain low [14]. In animal models the transfer of blastocysts from normal mice to DHEA-induced PCOS mice resulted in a reduced implantation rate [15]. Moreover, patients with PCOS had an increased risk of miscarriage with reported rates between 30 and 50% of all conceptions [16, 17]. In addition, PCOS seems to be responsible for more than 30% of cases of recurrent miscarriages [18].

Since oocytes and embryo quality do not seems to be the cause of low implantation and pregnancy rate in PCOS patients as demonstrated by donor oocyte models [19, 20], the decreased receptivity of the endometrium seems more probable. Indeed, accumulating data support the hypothesis that endometrium of PCOS patients is affected probably as a consequence of hormonal imbalance. Thus, unopposed estrogens, hyperinsulinemia, hyperandrogenism and the members of insulin-like growth factor family were reported as possible contributors to endometrial pathology in PCOS [21]. Factors associated with unexplained recurrent pregnancy loss like high serum levels of free testosterone and LH, decreased luteal phase progesterone and delayed endometrial development [22] are also found in PCOS patients, suggesting their involvement in the high miscarriage rate in PCOS.

Several abnormalities of the endometrium were reported in PCOS women. Thus, endometrial gene expression and sex hormone receptors, co-receptors, adhesion molecules expression and endometrial markers were reported to be abnormal [23].

Advertisement

3. Endometrial receptivity in women with polycystic ovary syndrome

Endometrial receptivity is a complex feature of the endometrium that allow the embryo to attach and invade the endometrium, and its further development into a viable fetus. For normal implantation both embryo and endometrial quality are important. In normal women the endometrium is receptive to embryo implantation for a period of 3–6 days which starts seven to ten days after ovulation known as the window of implantation. In pathological conditions this window can be shorten or shifted, resulting in infertility or pregnancy loss.

Human endometrium is a tissue whose development depends on the level of circulating hormones. During the follicular phase of the cycle, the increasing circulating estradiol levels determine the proliferation of the endometrial cells and increased endometrial sensitivity to estrogen through increasing the estrogen receptors (ER) ER-α levels [24, 25]. The ER expression is highest in the late proliferative phase, decreasing in the luteal phase [25]. Following ovulation, the progesterone production determines inhibition of cellular proliferation, mitotic activity, DNA synthesis and stimulates the differentiation of the endometrial cells [26]. The inhibition of proliferation of the epithelial endometrial cells is the consequence of the progesterone-induced regulation of genes resulting in down regulation of estrogen receptors and the induction of the enzymes that metabolize estrogens reducing its cellular effects [26]. Moreover, progesterone reduces the expression of androgen receptor in endometrial cells and stroma [26]. As a consequence of all these changes the ‘window of implantation’ occur. Progesterone is also essential for decidualization, a process that allow trophoblast invasion in case the implantation occur and establish a cytokine milieu and immunomodulatory network in the stroma. The decidualization is the consequence of endometrial stromal cells modifications of the cytoskeleton and up-regulation of prolactin, insulin-like growth factors, IGF binding proteins, insulin receptor and other factors. In the case the implantation does not occur, the decreasing estrogen and progesterone levels determines a shift from the expression of the innate immune genes to inflammatory genes expression in association with cellular apoptosis, increased production of metalloproteinases and prostaglandins, followed by endometrial desquamation and menstruation [26].

In women with PCOS, in the absence of ovulation the progesterone effects on the endometrium are lacking or severely decreased, affecting the decidualization and the window of implantation. Moreover, women with PCOS may have increased exposure to estrogen levels [27] as a consequence of aromatization of increased androgens in adipose tissue and decreased sex hormone binding globulin due to hyperinsulinemia [28, 29]. Several experimental studies support the alteration of endometrium especially in the window of implantation. Thus, Avellaira et al. [30] found that the tissue homeostasis in secretory endometrium of untreated women with PCOS is affected by an imbalance between apoptosis and cell proliferation which is increased as demonstrated by a study evaluating the expression of the proteins related with the two processes [30].

It was also suggested that some endometrial alterations in PCOS are the consequence of prenatal intrauterine exposure to androgens being considered primary endometrial abnormalities [23]. Thus, the endometrium of PCOS women has a preponderance of estrogen and androgen action and decreased progesterone action as a consequence of hormone receptors expression and function.

3.1 Estrogen receptors (ER) expression and function

In PCOS patients the level of ER seems to be increased in all the cycle phases [31, 32, 33, 34, 35]. Moreover, some coactivators of the ER-α like TIF2 and AIB1 were also found in higher levels in the proliferative endometrium of the PCOS patients compared with controls [32, 33, 34, 35]. Moreover, during the proliferative phase, the endometrium of PCOS women showed a higher Bcl-2/Bax ratio, indicating the predominance of anti-apoptotic factors in the estrogen receptor increased environment [31]. The higher levels of mRNA and protein for ER-α and coactivators compared with normal women were reported in the mid-secretory phase endometrium of PCOS as well [32]. Moreover, the coactivator ARA70 was increase and epithelial expression of beta3-integrin, a protein involved in cell adhesion and cell surphace mediated signaling, was decreased in endometrium of PCOS versus control [32].

Enhanced ER-α activation was associated with rare or absent apoptosis and increased in cell proliferation in the endometrium in the mid luteal phase [30, 36, 37, 38]. On the other hand, it is possible that high circulating estrogens as in patients performing ovarian stimulation to determine the apoptosis of the endometrial glandular cells, as showed by an experimental study by Chen et al. [39].

Other factors can contribute to increased exposure of endometrial cells to estrogens. Thus, the modified activity of the endometrial enzymes 17-β-hydroxysteroid dehydrogenase, which is downregulated and hydroxysteroid dehydrogenase type 1, which is upregulated, could contribute to increased local production of estradiol and androstenediol with increased estrogenic activity [40, 41, 42].

3.2 Progesterone receptor (PR) expression and function

In endometrium of PCOS women progesterone receptors (PR) alpha and beta mRNA is overexpressed and the stromal immunostaining of PR-b and Ki67 is higher compared with BMI-matched controls [43]. Moreover, an imbalance between PR-a expression and PR-b was found in proliferative endometrium of obese PCOS women, with the predominance of the later [43]. It was also reported that endometrial tissue of PCOS patients has decreased responsiveness to progesterone [44], also known as ‘P resistance’ [45]. This progesterone resistance might be due to reduced binding and activation of PR [46] or to an altered expression of its isoforms [46, 47]. Elevated PR isoform expression was showed to be associated with increased systemic levels of estrogen [48] and of androgen [49, 50]. Thus, hyperandrogenism modulates the expression and function of PR being associated with inactive or less active isoforms of PR [49, 50, 51], being probably responsible for progesterone resistance in PCOS women.

Mucin 1 (MUC1) is a progesterone-regulated molecule that carries selectin ligands recognized by the human blastocyst. Thus, Margarit et al. [52] showed that MUC1 expression is lower in anovulatory PCOS than in fertile patients, being a possible contributor to decreased implantation.

3.3 Androgen receptor

In the endometrium of PCOS women the androgen receptor (AR) mRNA and protein expression is increased [23] and coactivators of AR were found to be overexpressed as well [37]. Among these coactivators, Melanoma-associated antigen 11 (MAGEA11) was showed to bind to AR resulting in alteration of window of implantation [11]. Endometrial microenvironment can also contribute to over-exposure to androgen. Thus, low level of SHBG can increase the bioavailable testosteron at endometrial level [53] and increased activity of endometrial 5a-reductase generates potent androgens such as di-hydro-testosterone [37]. The uptake of intracrine precursors of testosterone [38, 54] and increased activity of hydroxi-steroid-dehydrogenase might contribute to high androgen exposure of endometrium in PCOS [55].

3.4 Hyperandrogenism

During the mid-secretory phase of the menstrual cycle and the window of implantation the level of circulating androgens reach a nadir in normal cycles [56], while in PCOS patients with hyperandrogenism the level of circulating androgens is constantly increased. There is a significant body of evidence suggesting that overexposure to androgens may affect the normal development of endometrium and, therefore the endometrial receptivity. Thus, Cermik et al. [57] studied the effect of testosterone on HOXA10 expression in endometrium, a gene well known to be involved in endometrial receptivity. They demonstrated that, in vitro testosterone exposure decreases the expression on HOXA10 and prevents the increase of this gene in response to estrogen and progesterone [57]. They also confirmed that the expression of HOXA10 is decreased in the endometrium of the hyperandrogenic PCOS patients [57]. Homebox (HOX) are genes essential for endometrial receptivity which are maximally expressed in endometrium during the window of implantation [58]. Their importance in implantation is demonstrated by experimental studies on female mice with disruption of HOXA10 which show infertility with implantation failure in spite of the presence of ovulation [59].

It was also showed that Wilms tumor suppressor (WT1) gene which is expressed in endometrium in the window of implantation, was downregulated in ovulatory women with PCOS in comparison with normal controls and that this downregulation is the consequence of androgen exposure in in vitro models [60] Since high androgen are associated with elevated PR isoforms, it was suggested that hyperandogenism could be a contributor to progesterone resistance found in PCOS women [50, 61]. High androgens can also affect the number and function of endometrial pinopodes which are associated with endometrial receptivity [62, 63]. Androgens can also influence the decidualization by their ability to modulate the oxidative stress response in decidualized endometrial cells [64] since the oxidative stress was showed to influence factors involved in embryo implantation like cytokeratin 8 (CK-8) [65]. Other androgens can also act on endometrium. Thus, DHEA seems to block glucose utilization resulting in inhibition of decidualization [66] and modulation of cell survival and apoptosis [67]. In pregnant mice treated with DHEA an impaired LIF-signal transducer and activator of transcription 3 (STAT3) pathway was observed, which was associated with implantation failure [68].

Clinical data showed that pregnant women with PCOS have lower endovascular trophoblast invasion in relation to circulating testosterone and the clinical phenotypes involving hyperandrogenism [69, 70].

3.5 Hyperinsulinemia and insulin resistance

In clinical studies, in PCOS patients undergoing in vitro maturation-in vitro fertilization embryo transfer cycle insulin resistance was associated with decreased implantation, clinical pregnancy and ongoing pregnancy rates [71]. Energy metabolism is vital for proper endometrial function taking into consideration the rapid turnover of endometrial tissue. Therefore, insulin action on endometrial tissue might be essential for endometrial receptivity. Insulin resistance and compensatory hyperinsulinemia can have detrimental effects on the implantation process as hyperinsulinemia has been shown to impair stromal cell decidualization in vitro [72, 73, 74]. In PCOS endometrium several abnormalities in insulin signaling and glucose transport have been reported. Thus, hyperinsulinemia can reduce insulin receptor substrate 1 (InRS-1) activation and glucose transport in endometrial stromal cells in PCOS women [75, 76], probably via inflammatory pathways [76, 77]. Experimental studies showed that in vitro exposure to dihydrotestosterone altered the expression of insulin receptor and insulin receptor substrates and the phosphorylation of insulin receptor in endometrial stromal cells [74]. Other studies suggests that insulin action is decreased in endometrium of hyperinsulinemic women with PCOS, by showing that pAS160T642 and SLC2A4 which are substrates of insulin receptor are decreased in comparison with non-hyperinsulinemic PCOS and controls [75].

In vitro studies showed that insulin inhibits the production of IGFBP-1 in the endometrial stoma. IGFBP-1 is considered a biomarker of decidualization [73]. It is also possible that adiponectin, an insulin regulating molecule and regulator of glucose metabolism, which is decreased in PCOS patients, to be involved in endometrial receptivity of these patients. Thus, adiponectin receptors were found to be highly expressed in the human endometrium during the window of implantation [78]. Moreover, decidualized mouse endometrium is able to secrete adiponectin and adiponectin receptors were found both in decidual cells and embryo after implantation [79]. This data supports the hypothesis that adiponectin might play a role in endometrial receptivity and implantation. Studies showing that metformin reduces the miscarriage rates in PCOS patients indirectly support the hypothesis of insulin resistance involvement in endometrial receptivity [80].

3.6 Vitamin D

Vitamin D deficiency is a frequent condition among women with infertility or PCOS. Numerous studies suggested a role of vitamin D in reproductive health at variate level of reproductive system, including endometrium. However, clinical data regarding the association between serum vitamin D level and endometrial pathology are divergent. A prospective controlled study [81] analyzed factors associated with recurrent implantation failure and found higher prevalence of chronic endometritis, a lower vitamin D level and a borderline lower progesterone level in comparison with controls [81]. However, two recent systematic reviews found no association between serum vitamin D level and miscarriage rate in women who performed in vitro fertilization [82, 83], while one of these reviews reported higher pregnancy rates in vitamin D replete patients undergoing assisted reproduction treatments [82].

Experimental studies support the role of vitamin D in endometrial receptivity. Thus, Guo et al. [84] found that circulating level of vitamin D was positively associated with vitamin D receptor and HOXA10 protein level expression in the endometrium and these were substantially elevated in pregnant women compared to non-pregnant women. During the window of implantation, higher serum vitamin D levels were associated with more mature pinopodes (84). Another study showed that in patients with recurrent implantation failure vitamin D treatment increases progesterone receptor mRNA and protein level and its phosphorylation on Ser294 residues in the endometrial cells (85). These results suggest that vitamin D may play a key role in the endometrial receptivity [85].

In patients with PCOS vitamin D deficiency is frequently found [86]. However, specific data regarding the relationship between serum vitamin D and endometrium in PCOS patients are scarce. Several studies reported that vitamin D treatment can improve the features associated with endometrial receptivity like hyperandrogenism [87], hyperinsulinism and insulin resistance [88], inflammation and oxidative stress [89].

In an experimental study in a rat model of PCOS it was found that immunohistochemical staining of caspase-3 and Ki-67 were decreased with vitamin D treatment compared non-treated group [90]. Moreover, endometrial, epithelial and stromal thickness measurements were decreased in the vitamin D treatment group compared to non-treated PCOS group [90].

Advertisement

4. Strategies to improve endometrial function in PCOS

4.1 Ovulation induction agents

First-line treatment for ovulation induction in the treatment of infertility in PCOS are oral agents, with letrozole being superior to clomiphene citrate (CC) in terms of endometrial thickness and markers of endometrial receptivity [7, 91, 92]. Gonadotropins are the second line treatment and it seems that they have a less deleterious impact on endometrium [93]. However, no data today support the ovulation induction in infertile ovulatory PCOS for the modification of endometrial receptivity.

Several studies evaluated the different impact of ovarian stimulation with letrozole versus clomiphene citrate in patients with PCOS. Thus, Wallace et al. published in 2011 a randomized controlled study which reported that letrozole positively influenced a number of markers of endometrial receptivity like mRNA expression of leukemia inhibitory factor, dickkhopf homolog 1, fibroblast growth factor 22 compared with CC [94]. Another randomized controlled study which included 160 patients diagnosed with PCOS found that indices of endometrial receptivity like the volume, vascularization index, flow index and vascularization flow index of endometrium on the day of hCG administration and 7–9 days after ovulation were significantly increased in letrozole group compared with CC [95]. Moreover, the biochemical pregnancy rate, clinical pregnancy rate and ongoing pregnancy rate in letrozole group were significantly higher compared with CC group [95]. The same authors evaluated the indices of endometrial receptivity in treated PCOS patients (Letrozole or clomiphene citrate) and non-treated [96]. They noticed that, although the successful ovulation rate did not differ between the letrozole group and CC group, endometrial thickness, endometrial volume, vascularization index, flow index, vascularization flow index, integrin αvβ3 and VEGF concentrations in uterine fluid were significantly higher in the window of implantation in the letrozole group compared with the CC group and natural cycle group [96]. Moreover, the clinical pregnancy and ongoing pregnancy rates of the letrozole group were significantly higher than in the CC group [96]. The markers of endometrial receptivity analyzed were significantly higher in pregnant patients [96]. The endometrial flow index during the implantation window had the highest predictive value for pregnancy. The integrin αvβ3 in uterine fluid had better predictive value than VEGF [96].

In an experimental study on female rats it was found that the expression of integrin αvβ3 in the clomiphene citrate group was lower than in the letrozole and saline solution groups [97]. The expression of HOXA10 was statistically significantly higher in the saline solution group than in the letrozole group, and the letrozole group showed a statistically significantly higher expression of HOXA10 compared with the clomiphene citrate group. The authors concluded that, in rats, letrozole affects the expression of HOXA10 in uterine epithelium but has no effect on the expression of integrin αvβ3, which suggests that clomiphene suppresses endometrial receptivity more than letrozole.

4.2 Antiandrogens

Antiandrogens as flutamide, finasteride and spironolactone are used in clinical practice to improve the signs of clinical hyperandrogenism like hirsutism, acne, alopecia. Animal studies demonstrated the ability of antiandrogens to improve endometrial function [98, 99]. However, their use in patients seeking infertility treatment is prohibited by their teratogenic effects, being generally recommended to be administrated only in association with oral contraception [100]. Another way to improve hyperandrogenism is by administration of combined oral contraceptives which decrease ovarian androgen production and decrease androgen bioavailability through stimulation of sex hormone binding globulin production. Indeed, oral contraceptives, administrated in a successive or unsuccessive manner before in vitro fertilization, were showed to increase the implantation and pregnancy rate and reduce the risk of pregnancy complications along with significant decrease in circulating androgens [101]. However, in a randomized controlled trial ovulation rate and live birth rate were superior in PCOS patients receiving life style modification with or without oral contraceptive in comparison with oral contraceptive alone [102].

4.3 Freeze all strategy

Elective frozen embryo transfer after a freeze-all strategy was suggested for infertile PCOS patients undergoing IVF [103]. This approach could be particularly effective in PCOS due to exposure of the endometrium to high estrogen levels during controlled ovarian stimulation. Thus, transfer of the embryo in a subsequent cycle may avoid failure of implantation due to inadequate endometrium receptivity. This strategy was showed of particular benefit in PCOS patients and only in case of more than 16 oocytes or estradiol higher than 3000 pg./ml, resulting in higher live birth rate [104]. Regarding the preparation of endometrium, it seems that the pregnancy rate is higher in natural cycles over the hormone replacement cycles in women performing frozen embryo transfer [105]. However, specific data for PCOS women are lacking.

In women with PCOS, no difference in live birth/ongoing pregnancy and clinical pregnancy rates was detected between FET cycles stimulated with hMG and cycles artificially prepared with E2 valerate [106, 107, 108].

4.4 Gut microbiota and endometrial receptivity

There is accumulating evidence that gut microbiota might play a role in PCOS pathogenesis [109, 110]. Thus, it was showed that gut microbiota of PCOS patients is different from controls [111] and that variate profiles of gut microbiota are associated with features of PCOS such as hyperandrogenism [112], hyperinsulinemia and insulin resistance [113] and obesity [114]. In experimental studies transplantation of feces from PCOS women leads to development of PCOS features in mice [109] and oral administration of variate bacteria was associated with improvement of PCOS manifestation [110]. Thus, Qi X et al. [109] showed that Bacteroides vulgatus was markedly elevated in the gut microbiota of individuals with PCOS and that transplantation of fecal microbiota from women with PCOS or B. vulgatus-colonized recipient mice resulted in increased disruption of ovarian functions, insulin resistance, altered bile acid metabolism, reduced interleukin-22 secretion and infertility [109]. It was also showed that IL-22 improved the PCOS phenotype [109]. Guo Y et al. [110] showed that PCOS rats displayed different composition of gut microbiota that in the controls. Lactobacillus, Ruminococcus and Clostridium were lower while Prevotella was higher in PCOS rats when compared with control rats. After treating PCOS rats with Lactobacillus and fecal microbiota transplantation from healthy rats estrous cycles were improved with decreasing androgen biosynthesis [110].

However, it is possible that the relationship between gut microbiota and PCOS to be bidirectional since in androgen-induced PCOS animal models the disturbances of gut microbiota was reported [115, 116]. In turn, DHEA-shaped gut microbiota transplanted to pseudo germ-free rats recipients trigged disturbances in reproductive hormone [115]. Another study showed that both androgens and high fat diet could shift the overall gut microbial composition, being associated with the development and pathology of PCOS by shaping gut microbial communities [117]. Although direct evidence of a connection between gut microbiota and endometrial receptivity was not reported yet, we can hypothesize a connection between these two due to the recognized involvement of insulin resistance, hyperandrogenemia and obesity in endometrial receptivity which, in turn, can be shaped by intestinal bacterial community.

Regarding the mechanisms connecting gut microbiota and PCOS, Tremellen and Pearce suggest that dysbiosis of the gut microbiota brought about by a high fat-sugar diet in PCOS patients leads to an increase in intestinal permeability. Lipopolysaccharide produced by Gram-negative bacteria traverse the gut wall to enter the circulation, leading to a chronic state of low-grade inflammation. Activation of the immune system interferes with insulin receptor, driving up insulin levels, which boost testosterone production in the ovary, leading to PCOS [118].

Another way by which gut microbiota might influence the endometrial receptivity is through its recently demonstrated close connection with genital tract microbiota [119, 120, 121, 122]. The crosstalk between gut and vaginal microbiota is highlighted by studies showing that oral administration of probiotics or bacteria can influence immunity in the vagina [120]. Thus, oral administration of Lactobacillus johnsonii was showed to inhibit the expression of inflammatory molecules in the vagina and to alleviate Gardnerella vaginalis induced vaginosis [122]. Kutteh et al. [121] demonstrated that intestinal tract immunization by oral and rectal route is followed by the induction of specific antibodies in human female genital tract secretions [Kutteh 2001]. Kim et al. [120] showed that in ovarectomized female mice, the oral administration of anti-inflammatory Lactobacillus plantarum NK3 and Bifidobacterium longum NK49 from kimchi and human fecal lactic acid bacteria collection was associated with alleviation of Gardnerella vaginalis induced vaginosis, inhibition of NF-κB activation and TNF-α expression in the vagina and uterus, and decreased the Gardnerella vaginalis population in the vagina [120]. In turn, Gardnerella vaginalis-induced vaginosis increased colonic myeloperoxidase activity, TNF-α expression, and fecal Proteobacteria population. NK3 and/or NK49 treatments reduced TNF-α expression and NF-κB activation in the colon and restored Gardnerella vaginalis disrupted gut microbiota composition [120].

This interplay between gut and reproductive tract microbiota may be related with endometrium function since, recently, it was showed that pathological modification of the profile of the bacterial community of the endometrial fluid might play a role in poor reproductive outcome for in vitro fertilization patients [123]. Thus, the presence of a non-Lactobacillus-dominated microbiota in a receptive endometrium was associated with significant decreases in implantation, pregnancy, ongoing pregnancy, and live birth rates [123]. Haahr et al. [124] found a decreased pregnancy rate in patients with abnormal vaginal microbiota performing IVF [124]. These studies highlight the importance of reproductive tract microbiota in fertility, possibly by modulation of endometrial receptivity.

All these data offer future therapeutic strategies to counteract decreased endometrial receptivity in PCOS women by modulating gut or reproductive tract microbiota or by administration of interleukin 22 as suggested by Qi X et al. [109].

4.5 Assessment of endometrial receptivity and therapeutical implications

The evaluation of endometrial receptivity was historically done by endometrial histology. However, recent studies find little concordance with new transcriptomic methods of endometrial receptivity assessment [125]. Endometrial receptivity array (ERA) is a test that analyzes the gene expression of the endometrium using a panel of 238 genes that have been implicated in endometrial receptivity. The indication of this test in the clinical practice is to identify the window of implantation in patients with accelerated or delayed endometrial luteal phase development, therefore allowing the transfer of the embryo in the right moment for the embryonic-endometrial synchrony. However, data regarding the benefit of this test are limited [126]. Although in PCOS patients performing IVF ERA test has hypothetical indications, studies evaluating this aspect are lacking.

Another aspect that should be clarified further is the consistency of the ERA findings from cycle to cycle since the transfer of the embryo should be done in a non-biopsy cycle. This aspect was analyzed in seven women who performed the ERA test in separate endometrial samples obtained 29–40 months apart and found high similarity across the samples [127]. However, only five samples were from the luteal phase and only four were in the receptive phase, all the samples being normal. Therefore, the possibility to extrapolate these findings to patients with endometrium receptivity abnormalities or dyssynchrony is unknown.

The ERA test was studied in several categories of infertile patients. Tan et al. [128] found that among patients with a history of at least one euploid blastocyst implantation failure personalized frozen embryo transfer (FET) according to ERA test results was superior to standard FET in terms of implantation and ongoing pregnancy rate, although the difference was not statistically different [128]. Other studies showed a similar pregnancy rate between patients with receptive and non-receptive endometrium according to the ERA test when a personalized embryo transfer was performed in patients with recurrent implantation failure [129] or with a history of implantation failure [125]. On the other hand, Eisman et al. [130] found a similar pregnancy rate in women who had a prior failed embryo transfer and personalized frozen embryo transfer following ERA test compared with women without a prior failed embryo transfer [130]. However, the pregnancy rate was lower in patients with more than three failed embryo transfer despite personalized transfer [130].

A multicenter, open-label randomized controlled trial evaluated patients at their first appointment for IVF after exclusion of patients with recurrent miscarriage and implantation failure. They found that a personalized frozen embryo transfer guided by ERA test was superior in terms of cumulative pregnancy and live birth rate to standard frozen embryo transfers and fresh embryo transfers [131]. At the first embryo transfer cycle, although the pregnancy rate and implantation rates were significantly higher compared with the two groups without ERA guidance, the live birth rates were similar [131]. Basil et al. [132] performed a single center retrospective cohort study and found that using the ERA test in patients undergoing frozen embryo transfer, there is no benefit in terms of ongoing pregnancy rate in good prognosis patients [132].

Taken all together, these studies suggest that the ERA test might be beneficial in patients with implantation failure, although future studies are necessary to clarify this aspect. In patients at their first attempt of IVF personalized frozen embryo transfer guided by ERA test may increase the cumulative pregnancy and live birth rate, although without obvious benefits at the first transfer. Whether these results are also valid for PCOS patients remains to be established.

Advertisement

5. Conclusion

An increasing body of evidence suggests that endometrial receptivity in PCOS patients is decreased, being a significant contributor to infertility in these patients. However, specific strategies to overcome this barrier in infertile PCOS women should be created in order to improve fertility treatment outcome.

References

  1. 1. Bozdag G, Mumusoglu S, Zengin D, Karabulut E, Yildiz BO. The prevalence and phenotypic features of polycystic ovary syndrome: a systematic review and meta-analysis. Hum Reprod 2016; 31 (12) 2841-2855
  2. 2. Azziz, R. et al. The prevalence and features of the polycystic ovary syndrome in an unselected population. The Journal of clinical endocrinology and metabolism 89, 2745-2749, doi: 10.1210/jc.2003-032046 (2004).
  3. 3. Norman RJ, Dewailly D, Legro RS, Hickey TE. Polycystic ovary syndrome. Lancet 370, 685-697, doi: 10.1016/S0140-6736(07)61345-2 (2007).
  4. 4. Franks S, Stark J, Hardy K. Follicle dynamics and anovulation in polycystic ovary syndrome. Human reproduction update 14, 367-378, doi: 10.1093/humupd/dmn015 (2008)
  5. 5. Deswal R, Narwal V, Dang A and Pundir CS. The Prevalence of Polycystic Ovary Syndrome: A Brief Systematic Review. J Hum Reprod Sci. 2020 Oct-Dec; 13(4): 261-271. Published online 2020 Dec 28. doi: 10.4103/jhrs.JHRS_95_18
  6. 6. Cooney LG, Lee I, Sammel MD, Dokras A.Hum Reprod. High prevalence of moderate and severe depressive and anxiety symptoms in polycystic ovary syndrome: a systematic review and meta-analysis. 2017 May 1;32(5):1075-1091. doi: 10.1093/humrep/dex044.
  7. 7. Teede HJ, Misso ML, Costello MF, Dokras A, Laven J, Moran L, Piltonen T, Norman RJ; International PCOS Network. Recommendations from the international evidence-based guideline for the assessment and management of polycystic ovary syndrome. Hum Reprod. 2018 Sep 1;33(9):1602-1618. doi: 10.1093/humrep/dey256.
  8. 8. Rotterdam ESHRE/ASRM-Sponsored PCOS Consensus Workshop Group. Revised 2003 consensus on diagnostic criteria and long-term health risks related to polycystic ovary syndrome. Fertil Steril 2004; 81 (01) 19-25
  9. 9. Brassard M, AinMelk Y, Baillargeon JP. Basic infertility including polycystic ovary syndrome. Med Clin North Am. 2008 Sep;92(5):1163-1192, xi. doi: 10.1016/j.mcna.2008.04.008.
  10. 10. Joham AE, Teede HJ, Ranasinha S, Zoungas S, Boyle J. Prevalence of infertility and use of fertility treatment in women with polycystic ovary syndrome: data from a large community-based cohort study. J Womens Health (Larchmt). 2015 Apr;24(4):299-307. doi: 10.1089/jwh.2014.5000. Epub 2015 Feb 5.
  11. 11. Younas K, Quintela M, Thomas S, Garcia-Parra J, Blake L, Whiteland H, Bunkheila A, Francis LW, Margarit L, Gonzalez D and Conlan RS. Delayed endometrial decidualisation in polycystic ovary syndrome; the role of AR-MAGEA11. J Mol Med (Berl). 2019; 97(9): 1315-1327. Published online 2019 Jun 29. doi: 10.1007/s00109-019-01809-6
  12. 12. Hull MG. Epidemiology of infertility and polycystic ovarian disease: endocrinological and demographic studies. Gynecol Endocrinol 1987 Sep;1(3):235-245. doi: 10.3109/09513598709023610.
  13. 13. Bellver J, et al. Endometrial gene expression in the window of implantation is altered in obese women especially in association with polycystic ovary syndrome. Fertil and Steril 95, 2335-2341, doi:10.1016/j.fertnstert.2011.03.021 (2011)
  14. 14. Lopes IM, et al. Endometrium in women with polycystic ovary syndrome during the window of implantation. Revista da Associacao Medica Brasileira 57,702-709 (2011)
  15. 15. Li SY, Song Z, Song MJ et al. Impaired receptivity and decidualization in DHEA-induced PCOS mice. Sci Rep 6,38134 (2016). https://doi.org/10.1038/srep38134
  16. 16. Balen AH, S.L. Tan, J. MacDougall, H.S.Jacobs. Miscarriage rates following in-vitro fertilization are increased in women with polycystic ovaries and reduced by pituitary desensitization with buserelin. Human Reproduction (Oxford, England), 8 (1993), pp. 959-964
  17. 17. Sagle M, K. Bishop, N. Ridley, et al. Recurrent early miscarriage and polycystic ovaries. BMJ (Clinical research ed. ), 297 (1988), pp. 1027-1028
  18. 18. Nestler JE, Stovall D, Akhter N, Iuorno MJ, Jakubowicz DJ. Strategies for the use of insulin-sensitizing drugs to treat infertility in women with polycystic ovary syndrome. Fertil Steril. 2002 Feb; 77(2):209-215.
  19. 19. Vaz GQ, Evangelista AV, Sartorio CAP, Cardoso MCA, Erthal MC, Gallo P, Oliveira MAP. Are patients with polycystic ovary syndrome ideal candidates for oocyte donation? Biomed Res Int 2016; 2016:5701609.
  20. 20. Ashkenazi J, Farhi J, Orvieto R. Polycystic ovary syndrome patients as oocyte donors: the effect of ovarian stimulation protocol on the implantation rate of the recipient. Fertility and Sterility, 64 (1995), pp. 564-567
  21. 21. Giudice LC. Endometrium in PCOS: Implantation and predisposition to endocrine CA. Best Practice & Research Clinical Endocrinology & Metabolism. June 2006;20(2):235-244
  22. 22. Dosiou C, L.C. Giudice. Natural killer cells in pregnancy and recurrent pregnancy loss: endocrine and immunologic perspectives. Endocrine Reviews, 26 (2005), pp. 44-62
  23. 23. Palomba S, Piltonen TT, Giudice LC. Endometrial function in women with polycystic ovary syndrome: a comprehensive review. Human Reproduction Update, Vol.27, No.3, pp. 584-618, 2021
  24. 24. Giudice LC. Elucidating endometrial function in the post-genomic era. Human Reproduction Update, 9 (2003), pp. 223-235
  25. 25. Xu XL, Deng SL, Lian ZX, Yu K. Estrogen Receptors in Polycystic Ovary Syndrome. Cells 2021 Feb; 10(2): 459. Published online 2021 Feb 21. doi: 10.3390/cells10020459
  26. 26. Giudice LC. Endometrium in PCOS: implantation and predisposition to endocrine CA. Best Pract Res Clin Endocrinol Metab 2006;20: 235-244
  27. 27. Venturoli S, E. Porcu, R. Fabbri, et al. Episodic pulsatile secretion of FSH, LH, prolactin, oestradiol, oestrone, and LH circadian variations in polycystic ovary syndrome. Clinical Endocrinology, 28 (1988), pp. 93-107
  28. 28. Nestler JE, L.P. Powers, D.W. Matt, et al. A direct effect of hyperinsulinemia on serum sex hormone-binding globulin levels in obese women with the polycystic ovary syndrome. The Journal of Clinical Endocrinology and Metabolism, 72 (1991), pp. 83-89
  29. 29. S. Robinson, D. Kiddy, S.V. Gelding, et al. The relationship of insulin insensitivity to menstrual pattern in women with hyperandrogenism and polycystic ovaries. Clinical Endocrinology, 39 (1993), pp. 351-355
  30. 30. Avellaira C, Villavicencio A, Bacallao K, Gabler F, Wells P, Romero C, Vega M. Expression of molecules associated with tissue homeostasis in secretory endometria from untreated women with polycystic ovary syndrome. Hum Reprod 2006;21:3116-3121.
  31. 31. Maliqueo M, Clementi M, Gabler F, Johnson C, Palomino A, Sir Peterman T, Vega M. Expression of steroid receptors and proteins related to apoptosis in endometria of women with polycystic ovary syndrome. Fertil Steril 2003, 812-819
  32. 32. Quezada S, Avellaira C, Johnson MC, Gabler F, Fuentes A, Vega M. Evaluation of steroid receptors, coregulators, and molecules associated with uterine receptivity in secretory endometria from untreated women with polycystic ovary syndrome. Fertil Steril 2006;85:1017-1026.
  33. 33. Gregory CW, Wilson EM, Apparao EKB, Lininger RA, Meyer WR, Kowalik A, Fritz MA, Lessey BA. Steroid receptor coactivator expression throughout the menstrual cycle in normal and abnormal endometrium. J Clin Endocrinol Metab 2002;87:2960-2966,
  34. 34. Villavicencio A, Bacallao K, Avellaira C, Gabler F, Fuentes A, Vega M. Androgen and estrogen receptors and co-regulators levels in endometria from patients with polycystic ovarian syndrome with and without endometrial hyperplasia. Gynecol Oncol 2006;103: 307-314
  35. 35. Wang A, Ji L, Shang W, Li M, Chen L, White RE, Han G. Expression of GPR30, ERalpha and ERbeta in endometrium during window of implantation in patients with polycystic ovary syndrome: a pilot study. Gynecol Endocrinol 2011;27:251-255
  36. 36. Plaza-Parrochia F, Bacallao K, Poblete C, Gabler F, Carvajal R, Romero C, Valladares L, Vega M. The role of androst-5-ene3b,17b-diol (androstenediol) in cell proliferation in endometrium of women with polycystic ovary syndrome. Steroids 2014;89: 11-19.
  37. 37. Plaza-Parrochia F, Oro´ stica L, Garcı´a P, Vera C, Romero C, Valladares L, Vega M. Molecular mechanisms of androstenediol in the regulation of the proliferative process of human endometrial cells. Reprod Sci 2017a;24:1079-1087.
  38. 38. Plaza-Parrochia F, Romero C, Valladares L, Vega M. Endometrium and steroids, a pathologic overview. Steroids 2017b;126:85-91.
  39. 39. Chen SU, Chou CH, Chen MJ, Chen TH, Yang YS, Yang JH. Apoptotic effects of high estradiol concentrations on endometrial glandular cells. J Clin Endocrinol Metab 2014;99:E971–E980.
  40. 40. Ito K, Utsunomiya H, Suzuki T, Saitou S, Akahira J, Okamura K, Yaegashi N, Sasano H. 17b-hydroxysteroid dehydrogenases in human endometrium and its disorders. Mol Cell Endocrinol 2006;248: 136-140
  41. 41. Leon L, Bacallao K, Gabler F, Romero C, Valladares L, Vega M. Activities of steroid metabolic enzymes in secretory endometria from untreated women with polycystic ovary syndrome. Steroids 2008;73:88-95.
  42. 42. Plaza F, Gabler F, Romero C, Vantman D, Valladares L, Vega M. The conversion of dehydroepiandrosterone into androst-5-ene-3b, 17b-diol (androstenediol) is increased in endometria from untreated women with polycystic ovarian syndrome. Steroids 2010; 75:810-817.
  43. 43. Paulson M, Norstedt G, Sahlin L, Hirschberg AL. Association between prolactin receptor expression and proliferation in the endometrium of obese women with polycystic ovary syndrome. Gynecol Endocrinol 2020a;36:226-232.
  44. 44. Chrousos GP, MacLusky NJ, Brandon DD, Tomita M, Renquist DM, Loriaux DL, Lipsett MB. Progesterone resistance. Adv Exp Med Biol 1986;196:317-328.45. Graham and Clarke, 1997
  45. 45. Graham JD, Clarke CL. Physiological action of progesterone in target tissues. Endocr Rev 1997;18:502-519.
  46. 46. Mote PA, Balleine RL, McGowan EM, Clarke CL. Heterogeneity of progesterone receptors A and B expression in human endometrial glands and stroma. Hum Reprod 2000:15 Suppl 3:48-56.
  47. 47. Patel B, Elguero S, Thakore S, Dahoud W, Bedaiwy M, Mesiano S. Role of nuclear progesterone receptor isoforms in uterine pathophysiology. Hum Reprod Update 2015;21:155-173.
  48. 48. Hu M, Li J, Zhang Y, Li X, Bra¨nnstro¨m M, Shao LR, Billig H. Endometrial progesterone receptor isoforms in women with polycystic ovary syndrome. Am J Transl Res 2018;10:2696-2705.
  49. 49. Babayev SN, Park CW, Keller PW, Carr BR, Word RA, Bukulmez O. Androgens upregulate endometrial epithelial progesterone receptor expression: potential implications for endometriosis. Reprod Sci 2017;24:1454-1461.
  50. 50. Young SL. Androgens and endometrium: new lessons from the corpus luteum via the adrenal cortex? Fertil Steril 2018;109:623-624.
  51. 51. Su S, Blackwelder AJ, Grossman G, Minges JT, Yuan L, Young SL, Wilson EM. Primate-specific melanoma antigen-A11 regulates isoform-specific human progesterone receptor-B transactivation. J Biol Chem 2012;287:34809-34824.
  52. 52. Margarit L, A. Taylor, M. H. Roberts, L. Hopkins, C. Davies, A. G. Brenton, R. S. Conlan, A. Bunkheila, L. Joels, J. O. White … Show more. MUC1 as a Discriminator between Endometrium from Fertile and Infertile Patients with PCOS and Endometriosis. The Journal of Clinical Endocrinology & Metabolism, Volume 95, Issue 12, 1 December 2010, Pages 5320-5329
  53. 53. Maliqueo M, Bacallao K, Quezada S, Clementi M, Gabler F, Johnson MC, Vega M. Sex hormone-binding globulin expression in the endometria of women with polycystic ovary syndrome. Fertil Steril 2007;87:321-328
  54. 54. Plaza-Parrochia F, Poblete C, Gabler F, Carvajal R, Romero C, Valladares L, Vega M. Expression of steroid sulfated transporters and 3b-HSD activity in endometrium of women having polycystic ovary syndrome. Steroids 2015;104:189-195.
  55. 55. Bacallao K, Leon L, Gabler F, Soto E, Romero C, Valladares L, Vega M. In situ estrogen metabolism in proliferative endometria from untreated women with polycystic ovarian syndrome with and without endometrial hyperplasia. J Steroid Biochem Mol Biol 2008;110: 163-169.
  56. 56. Schulte MMB, J, KH. The Effect of Metabolic Derangements on Endometrial Receptivity at the Time of Implantation. Reprod Sci. 2015 Jan; 22(1): 6-14.
  57. 57. Cermik D, Selam B, Taylor HS. Regulation of HOXA-10 expression by testosterone in vitro and in the endometrium of patients with polycystic ovary syndrome. J Clin Endocrinol Metab. 2003 Jan; 88(1):238-243. doi: 10.1210/jc.2002-021072.
  58. 58. Du H, Sarno J, Taylor HS. HOXA10 inhibits Kruppel-like factor 9 expression in the human endometrial epithelium. Biol Reprod. 2010;83 (2):205-211.
  59. 59. Satokata I, Benson G, Maas R. Sexually dimorphic sterility phenotypes in Hoxa10-deficient mice. Nature. 1995;374 (6521):460-463.
  60. 60. Gonzalez D, Thackeray H, Lewis PD, et al. Loss of WT1 expression in the endometrium of infertile PCOS patients: a hyperandrogenic effect? J Clin Endocrinol Metab. 2012;97 (3):957-966
  61. 61. Babayev SN, Park CW, Keller PW, Carr BR, Word RA, Bukulmez O. Androgens upregulate endometrial epithelial progesterone receptor expression: potential implications for endometriosis. Reprod Sci 2017;24:1454-1461.
  62. 62. Bentin-Ley U. Relevance of endometrial pinopodes for human blastocyst implantation. Hum Reprod 2000;15:67-73.
  63. 63. Mokhtar HM, Giribabu N, Muniandy S, Salleh N. Testosterone decreases the expression of endometrial pinopode and L-selectin ligand (MECA-79) in adult female rats during uterine receptivity period. Int J Clin Exp Pathol 2014;7:1967-1976
  64. 64. Kajihara T, Tochigi H, Prechapanich J, Uchino S, Itakura A, Brosens JJ, Ishihara O. Androgen signaling in decidualizing human endometrial stromal cells enhances resistance to oxidative stress. Fertil Steril 2012;97:185-191.
  65. 65. Hu M, Zhang Y, Li X, Cui P, Li J, Bra¨nnstro¨m M, Shao LR, Billig H. Alterations of endometrial epithelial-mesenchymal transition and MAPK signaling components in women with PCOS are partially modulated by metformin in vitro. Mol Hum Reprod 2020c;26: 312-326
  66. 66. Frolova AI, O'Neill K, Moley KH. Dehydroepiandrosterone inhibits glucose flux through the pentose phosphate pathway in human and mouse endometrial stromal cells, preventing decidualization and implantation. Mol Endocrinol. 2011;25 (8):1444-1455
  67. 67. Maliqueo MA, Quezada S, Clementi M, Bacallao K, Anido M, Johnson C, Vega M. Potential action of androstenedione on the proliferation and apoptosis of stromal endometrial cells. Reprod Biol Endocrinol 2004;2:81
  68. 68. Li SY, Song Z, Song MJ, Qin JW, Zhao ML, Yang ZM. Impaired receptivity and decidualization in DHEA-induced PCOS mice. Sci Rep 2016;6:6
  69. 69. Palomba S, Russo T, Falbo A, Di Cello A, Amendola G, Mazza R, Tolino A, Zullo F, Tucci L, La Sala GB. Decidual endovascular trophoblast invasion in women with polycystic ovary syndrome: an experimental case-control study. J Clin Endocrinol Metab 2012;97: 2441-2449
  70. 70. Palomba S, Falbo A, La Sala GB. Metformin and gonadotropins for ovulation induction in patients with polycystic ovary syndrome: a systematic review with meta-analysis of randomized controlled trials. Reprod Biol Endocrinol 2014b;12:3.
  71. 71. Chang EM, Han JE, Seok HH, Lee DR, Yoon TK, Lee WS. Insulin resistance does not affect early embryo development but lowers implantation rate in in vitro maturation-in vitro fertilization-embryo transfer cycle. Clin Endocrinol (Oxf). 2013;79 (1):93-99.
  72. 72. Irwin JC, de las Fuentes L, Dsupin BA, Giudice LC. Insulin-like growth factor regulation of human endometrial stromal cell function: coordinate effects on insulin-like growth factor binding protein-1, cell proliferation and prolactin secretion. Regul Pept 1993;48:165-177
  73. 73. Ujvari D, Jakson I, Babayeva S, Salamon D, Rethi B, Gidlöf S, Hirschberg AL. Dysregulation of In Vitro Decidualization of Human Endometrial Stromal Cells by Insulin via Transcriptional Inhibition of Forkhead Box Protein O1. PLoS One. 2017 Jan 30;12(1):e0171004. doi: 10.1371/journal.pone.0171004. eCollection 2017.
  74. 74. Lee MH, Yoon JA, Kim HR, Kim YS, Lyu SW, Lee BS, Song H, Choi DH. Hyperandrogenic milieu dysregulates the expression of insulin signaling factors and glucose transporters in the endometrium of patients with polycystic ovary syndrome. Reprod Sci 2020;27: 1637-1647
  75. 75. Fornes R, Ormazabal P, Rosas C, et al. Changes in the expression of insulin signaling pathway molecules in endometria from polycystic ovary syndrome women with or without hyperinsulinemia. Mol Med. 2010;16 (3-4):129-136.
  76. 76. Oro´ stica L, Poblete C, Romero C, Vega M. Pro-inflammatory markers negatively regulate IRS1 in endometrial cells and endometrium from women with obesity and PCOS. Reprod Sci 2020;27: 290-300.
  77. 77. Piltonen TT, Chen JC, Khatun M, Kangasniemi M, Liakka A, Spitzer T, Tran N, Huddleston H, Irwin JC, Giudice LC. Endometrial stromal fibroblasts from women with polycystic ovary syndrome have impaired progesterone-mediated decidualization, aberrant cytokine profiles and promote enhanced immune cell migration in vitro. Hum Reprod 2015;30:1203-1215.
  78. 78. Takemura Y, Osuga Y, Yamauchi T, et al. Expression of adiponectin receptors and its possible implication in the human endometrium. Endocrinology. 2006;147 (7):3203-3210
  79. 79. Dunaif A, Graf M. Insulin administration alters gonadal steroid metabolism independent of changes in gonadotropin secretion in insulin-resistant women with the polycystic ovary syndrome. J Clin Invest. 1989;83 (1):23-29.
  80. 80. Nestler JE. Should patients with polycystic ovarian syndrome be treated with metformin?: an enthusiastic endorsement. Hum Reprod. 2002 Aug;17(8):1950-1953. doi: 10.1093/humrep/17.8.1950.
  81. 81. Saxtorph MH, Hallager T, Persson G, Petersen KB, Eriksen JO, Larsen LG, Hviid TV, Macklon N. Assessing endometrial receptivity after recurrent implantation failure: a prospective controlled cohort study. Reprod Biomed Online. 2020 Dec;41(6):998-1006. doi: 10.1016/j.rbmo.2020.08.015.
  82. 82. Chu J, Gallos I, Tobias A, Tan B, Eapen A, Coomarasamy A. Vitamin D and assisted reproductive treatment outcome: a systematic review and meta-analysis. Hum Reprod 2018 Jan 1;33(1):65-80. doi: 10.1093/humrep/dex326
  83. 83. Cozzolino M, Busnelli A, Pellegrini L, Riviello E, Vitagliano A. How vitamin D level influences in vitro fertilization outcomes: results of a systematic review and meta-analysis. Fertil Steril 2020 Nov;114(5):1014-1025. doi: 10.1016/j.fertnstert.2020.05.040. Epub 2020 Oct 1.
  84. 84. Guo J, Liu S, Wang P, Ren H, Li Y. Characterization of VDR and CYP27B1 expression in the endometrium during the menstrual cycle before embryo transfer: implications for endometrial receptivity. Reprod Biol Endocrinol 2020 Mar 17;18(1):24. doi: 10.1186/s12958-020-00579-y.
  85. 85. Hosseinirad H, Novin MG, Hosseini S, Nazarian H, Amidi F, Paktinat S, Azizi E, Mofarahe ZS. Effect of 1,25(OH)2-vitamin D3 on expression and phosphorylation of progesterone receptor in cultured endometrial stromal cells of patients with repeated implantation failure. Acta Histochem. 2020 Feb;122(2):151489. doi: 10.1016/j.acthis.2019.151489. Epub 2019 Dec 24
  86. 86. Li HWR, Brereton RE, Anderson RA, Wallace AM, Ho CKM. Vitamin D deficiency is common and associated with metabolic risk factors in patients with polycystic ovary syndrome. Metabolism 2011 Oct;60(10):1475-1481. doi: 10.1016/j.metabol.2011.03.002. Epub 2011 May 6.
  87. 87. Al-Bayyari N, Al-Domi H, Zayed F, Hailat R, Eaton A. Androgens and hirsutism score of overweight women with polycystic ovary syndrome improved after vitamin D treatment: A randomized placebo controlled clinical trial. Clin Nutr 2021 Mar;40(3):870-878. doi: 10.1016/j.clnu.2020.09.024. Epub 2020 Sep 24.
  88. 88. Wang L, Wen X, Lv S, Tian S, Jiang Y, Yang X. Effects of vitamin D supplementation on metabolic parameters of women with polycystic ovary syndrome: a meta-analysis of randomized controlled trials. Gynecol Endocrinol 2021 May;37(5):446-455. doi: 10.1080/09513590.2020.1813272. Epub 2020 Sep 10.
  89. 89. Foroozanfard F, Jamilian M, Bahmani F, Talaee R, Talaee N, Hashemi T, Nasri K, Asemi Z, Esmaillzadeh A. Calcium plus vitamin D supplementation influences biomarkers of inflammation and oxidative stress in overweight and vitamin D-deficient women with polycystic ovary syndrome: a randomized double-blind placebo-controlled clinical trial. Clin Endocrinol (Oxf) 2015 Dec;83(6):888-894. doi: 10.1111/cen.12840. Epub 2015 Jul 23.
  90. 90. Kuyucu Y, Çelik LS, Kendirlinan O, Tap O, Mete UO. Investigation of the uterine structural changes in the experimental model with polycystic ovary syndrome and effects of vitamin D treatment: An ultrastructural and immunohistochemical study. Reprod Biol 2018 Mar;18(1):53-59. doi: 10.1016/j.repbio.2018.01.002. Epub 2018 Jan 8.
  91. 91. Gadalla MA, Huang S, Wang R, Norman RJ, Abdullah SA, El Saman AM, Ismail AM, van Wely M, Mol BW. Effect of clomiphene citrate on endometrial thickness, ovulation, pregnancy and live birth in anovulatory women: systematic review and meta-analysis. Ultrasound Obstet Gynecol 2018;51:64-76
  92. 92. Mehdinejadiani S, Amidi F, Mehdizadeh M, Barati M, Safdarian L, Aflatoonian R, Alyasin A, Aghahosseini M, Pazhohan A, Hayat P et al. The effects of letrozole and clomiphene citrate on ligands expression of Wnt3, Wnt7a, and Wnt8b in proliferative endometrium of women with polycystic ovarian syndrome. Gynecol Endocrinol 2018;34:775-780
  93. 93. Weiss NS, Kostova E, Nahuis M, Mol BWJ, van der Veen F, van Wely M. Gonadotrophins for ovulation induction in women with polycystic ovary syndrome. Cochrane Database Syst Rev 2019;1: CD010290
  94. 94. Wallace KL, Venessia Johnson, Victoria Sopelak, Randall Hines. Clomiphene citrate versus letrozole: molecular analysis of the endometrium in women with polycystic ovary syndrome. Fertil Steril. 2011 Oct;96(4):1051-1056. doi: 10.1016/j.fertnstert.2011.07.1092.
  95. 95. Wang L, Xinqiang Wen, Shulan Lv, Juan Zhao, Ting Yang, Xiaofeng Yang. Comparison of endometrial receptivity of clomiphene citrate versus letrozole in women with polycystic ovary syndrome: a randomized controlled study. Gynecol Endocrinol 2019 Oct;35(10):862-865. doi: 10.1080/09513590.2019.1612358. Epub 2019 May 12.
  96. 96. Wang L, Shulan Lv, Fen Li, E Bai, Xiaofeng Yang. Letrozole Versus Clomiphene Citrate and Natural Cycle: Endometrial Receptivity During Implantation Window in Women With Polycystic Ovary Syndrome. Front Endocrinol (Lausanne). 2021 Jan 18;11:532692. doi: 10.3389/fendo.2020.532692. eCollection 2020.
  97. 97. Bao SH, Shi Le Sheng, Yi Feng Peng, Qi De Lin. Effects of letrozole and clomiphene citrate on the expression of HOXA10 and integrin alpha v beta 3 in uterine epithelium of rats. Fertil Steril 2009 Jan;91(1):244-248. doi: 10.1016/j.fertnstert.2007.11.024. Epub 2008 Feb 4.
  98. 98. Gong H, Wu W, Xu J, Yu D, Qiao B, Liu H, Yang B, Li Y, Ling Y, Kuang H. Flutamide ameliorates uterine decidualization and angiogenesis in the mouse hyperandrogenemia model during mid-pregnancy. PLoS One 2019;14:e0217095.
  99. 99. Slayden OD, Brenner RM. Flutamide counteracts the antiproliferative effects of antiprogestins in the primate endometrium. J Clin Endocrinol Metab 2003;88:946-949.
  100. 100. ACOG. ACOG Practice Bulletin N. 194. Polycystic ovary syndrome. Obstet Gynecol 2018;131:e157–e171.
  101. 101. Pan JX, Liu Y, Ke ZH, Zhou CL, Meng Q, Ding GL, Xu GF, Sheng JZ, Huang HF. Successive and cyclic oral contraceptive pill pretreatment improves IVF/ICSI outcomes of PCOS patients and ameliorates hyperandrogenism and antral follicle excess. Gynecol Endocrinol 2015;31:332-336.
  102. 102. Legro RS, Dodson WC, Kris-Etherton PM, Kunselman AR, Stetter CM, Williams NI, Gnatuk CL, Estes SJ, Fleming J, Allison KC et al. Randomized controlled trial of preconception interventions in infertile women with polycystic ovary syndrome. J Clin Endocrinol Metab 2015;100:4048-4058
  103. 103. Wei D, Shi Y, Li J, Wang Z, Zhang L, Sun Y, Zhou H, Xu Y, Wu C, Liu L et al. Effect of pretreatment with oral contraceptives and progestins on IVF outcomes in women with polycystic ovary syndrome. Hum Reprod 2017;32:354-361.
  104. 104. Wei D, Yu Y, Sun M, Shi Y, Sun Y, Deng X, Li J, Wang Z, Zhao S, Zhang H et al. The effect of supraphysiological estradiol on pregnancy outcomes differs between women with PCOS and ovulatory women. J Clin Endocrinol Metab 2018;103:2735-2742.
  105. 105. Levi Setti PE, Cirillo F, De Cesare R, Morenghi E, Canevisio V, Ronchetti C, Baggiani A, Smeraldi A, Albani E, Patrizio P. Seven years of vitrified blastocyst transfers: comparison of 3 preparation protocols at a single ART center. Front Endocrinol (Lausanne) 2020; 11:346
  106. 106. Muasher SJ, Kruithoff C, Simonetti S, Oehninger S, Acosta AA, Jones GS. Controlled preparation of the endometrium with exogenous steroids for the transfer of frozen-thawed pre-embryos in patients with anovulatory or irregular cycles. Hum Reprod 1991;6:443-445.
  107. 107. Yu J, Ma Y, Wu Z, Li Y, Tang L, Li Y, Deng B. Endometrial preparation protocol of the frozen-thawed embryo transfer in patients with polycystic ovary syndrome. Arch Gynecol Obstet 2015;291: 201-211.
  108. 108. Kollmann M, Martins WP, Lima ML, Craciunas L, Nastri CO, Richardson A, Raine-Fenning N. Strategies for improving outcome of assisted reproduction in women with polycystic ovary syndrome: systematic review and meta-analysis. Ultrasound Obstet Gynecol 2016;48:709-718.
  109. 109. Qi X, Chuyu Yun, Lulu Sun, Jialin Xia, Qing Wu, Ying Wang, Lina Wang, Yangming Zhang, Xianyi Liang, Liying Wang, Frank J Gonzalez, Andrew D Patterson, Huiying Liu, Liangshan Mu, Zehong Zhou, Yue Zhao, Rong Li, Ping Liu, Chao Zhong, Yanli Pang, Changtao Jiang, Jie Qiao. Gut microbiota-bile acid-interleukin-22 axis orchestrates polycystic ovary syndrome. Nat Med. 2019 Aug;25(8):1225-1233. doi: 10.1038/s41591-019-0509-0. Epub 2019 Jul 22.
  110. 110. Guo Y, Yane Qi, Xuefei Yang, Lihui Zhao, Shu Wen, Yinhui Liu, Li Tang. Association between Polycystic Ovary Syndrome and Gut Microbiota. PLoS One. 2016 Apr 19;11(4):e0153196. doi: 10.1371/journal.pone.0153196. eCollection 2016.
  111. 111. Lindheim L, Bashir M, Münzker J, Trummer C, Zachhuber V, Leber B, Horvath A, Pieber TR, Gorkiewicz G, Stadlbauer V, Obermayer-Pietsch B. Alterations in Gut Microbiome Composition and Barrier Function Are Associated with Reproductive and Metabolic Defects in Women with Polycystic Ovary Syndrome (PCOS): A Pilot Study. PLoS One. 2017 Jan 3;12(1):e0168390. doi: 10.1371/journal.pone.0168390. eCollection 2017.
  112. 112. Torres PJ, Martyna Siakowska M, Banaszewska B, Pawelczyk L, Duleba AJ, Kelley ST, Thackray VG. Gut Microbial Diversity in Women With Polycystic Ovary Syndrome Correlates With Hyperandrogenism. J Clin Endocrinol Metab. 2018 Apr 1;103(4):1502-1511. doi: 10.1210/jc.2017-02153.
  113. 113. He F, Li Y. The gut microbial composition in polycystic ovary syndrome with insulin resistance: findings from a normal-weight population. J Ovarian Res. 2021 Mar 27;14(1):50. doi: 10.1186/s13048-021-00799-9.
  114. 114. Zhou L, NI Z, Yu J, Cheng W, Cai Z, You C. Correlation Between Fecal Metabolomics and Gut Microbiota in Obesity and Polycystic Ovary Syndrome. Front Endocrinol (Lausanne). 2020 Sep 8;11:628. doi: 10.3389/fendo.2020.00628. eCollection 2020.
  115. 115. Han Q, Wang J, Weiping Li, Zi-Jiang Chen, Yanzhi Du. Androgen-induced gut dysbiosis disrupts glucolipid metabolism and endocrinal functions in polycystic ovary syndrome. Microbiome. 2021 May 6;9(1):101. doi: 10.1186/s40168-021-01046-5.
  116. 116. Kelley ST, Danalea V Skarra, Alissa J Rivera , Varykina G Thackray. The Gut Microbiome Is Altered in a Letrozole-Induced Mouse Model of Polycystic Ovary Syndrome. PLoS One. 2016 Jan 5;11(1):e0146509. doi: 10.1371/journal.pone.0146509. eCollection 2016.
  117. 117. Yanhua Zheng, Jingwei Yu, Chengjie Liang, Shuna Li, Xiaohui Wen, Yanmei Li. Characterization on gut microbiome of PCOS rats and its further design by shifts in high-fat diet and dihydrotestosterone induction in PCOS rats. Bioprocess Biosyst Eng. 2021 May;44(5):953-964. doi: 10.1007/s00449-020-02320-w. Epub 2020 Mar 10.
  118. 118. Tremellen K, Pearce K. Dysbiosis of Gut Microbiota (DOGMA)--a novel theory for the development of Polycystic Ovarian Syndrome. Med Hypotheses. 2012 Jul;79(1):104-112. doi: 10.1016/j.mehy.2012.04.016. Epub 2012 Apr 27.
  119. 119. Quaranta G, Sanguinetti M, Masucci L. Fecal Microbiota Transplantation: A Potential Tool for Treatment of Human Female Reproductive Tract Diseases. Front Immunol. 2019; 10: 2653. Published online 2019 Nov 26. doi: 10.3389/fimmu.2019.02653
  120. 120. Kim DE, Kim JK, Han SK, Jang SE, Han MJ, Kim DH. Lactobacillus plantarum NK3 and Bifidobacterium longum NK49 Alleviate Bacterial Vaginosis and Osteoporosis in Mice by Suppressing NF-κB-Linked TNF-α Expression. J Med Food. (2019) 22:1022-1031. 10.1089/jmf.2019.4419
  121. 121. Kutteh WH, Kantele A, Moldoveanu Z, Crowley-Nowick PA, Mestecky J. Induction of specific immune responses in the genital tract of women after oral or rectal immunization and rectal boosting with Salmonella typhi Ty 21a vaccine. J Reprod Immunol. (2001) 52:61-75. 10.1016/S0165-0378(01)00109-7
  122. 122. Joo HM, Hyun YJ, Myoung KS, Ahn YT, Lee JH, Huh CS, et al. Lactobacillus johnsonii HY7042 ameliorates Gardnerella vaginalis-induced vaginosis by killing Gardnerella vaginalis and inhibiting NF-κB activation. Int Immunopharmacol. (2011) 11:1758-65. 10.1016/j.intimp.2011.07.002
  123. 123. Moreno I, Codoner FM, Vilella F, Valbuena D, Martinez-Blanch JF, Jimenez-Almazab J, Alonso R, Alama P, Remohi J, Pellicer A, Ramon D, Simon C. Evidence that the endometrial microbiota has an effect on implantation success or failure. Am J Obstet Gynecol. 2016 Dec;215(6):684-703. doi: 10.1016/j.ajog.2016.09.075. Epub 2016 Oct 4.
  124. 124. Haahr T, Jensen JS, Thomsen L, Duus L, Rygaard K, Humaidan P. Abnormal vaginal microbiota may be associated with poor reproductive outcomes: a prospective study in IVF patients. Hum Reprod. 2016 Apr;31(4):795-803. doi: 10.1093/humrep/dew026. Epub 2016 Feb 23.
  125. 125. Cohen AM, Ye XY, Colgan TJ, Greenblatt EM, Chan C. Comparing endometrial receptivity array to histologic dating of the endometrium in women with a history of implantation failure. Syst Biol Reprod Med. 2020 Dec;66(6):347-354. doi: 10.1080/19396368.2020.1824032.Epub 2020 Sep 30.
  126. 126. Lessey BA, Young SL. What exactly is endometrial receptivity? Fertil Steril 2019;111(4):611-617
  127. 127. Ruiz-Alonso M, Blesa D, Bosch N, Martínez-Conejero J.A, Alamá P. The accuracy and reproducibility of the endometrial receptivity array is superior to histology as a diagnostic method for endometrial receptivity. Fertil Steril. 2013; 99: 508-517
  128. 128. Tan J, Kan A, Hitkari J, Taylor B, Tallon N, Warraich G, Yuzpe A, Nakhuda G. The role of the endometrial receptivity array (ERA) in patients who have failed euploid embryo transfers. J Assist Reprod Genet. 2018 Apr;35(4):683-692. doi: 10.1007/s10815-017-1112-2.Epub 2018 Jan 11
  129. 129. Hashimoto T, Koizumi M, Doshida M, Toya M, Sagara E, Oka N, Nakajo Y, Aono N, Igarashi H, Kiono K. Efficacy of the endometrial receptivity array for repeated implantation failure in Japan: A retrospective, two-centers study. Reprod Med Biol. 2017 Jul; 16(3): 290-296. doi: 10.1002/rmb2.12041
  130. 130. Eisman LE, Pisarska MD, Wertheimer S, Chan JL, Akopians AL, Surrey MW, Danzer HC, Ghadir S, Chang WY, Alexander CJ, Wang ET. Clinical utility of the endometrial receptivity analysis in women with prior failed transfers. J Assist Reprod Genet. 2021 Mar;38(3):645-650. doi: 10.1007/s10815-020-02041-9. Epub 2021 Jan 17.
  131. 131. Simon C, Gomez C, Cabanillas S, Vladimirov I, Castillon G, Giles J, Boynukalin K, Findikli N, Bahceci M, Ortega I, Vidal C, Funabiki M, Izquierdo A, Lopez L, Portela S, Frantz N, Kulmann M, Taguchi S, Labarta E, Colucci F, Mackens S, Santamaria X, Munoz E, Barrera S, Garcia-Velasco A, Fernandez M, Ferrando M, Ruiz M, Mol BW, Valbuena D for the ERA-RCT Study Consortium Group. A 5-year multicentre randomized controlled trial comparing personalized, frozen and fresh blastocyst transfer in IVF. Reprod BioMed Online 2020;41(3):402-415
  132. 132. Bassil R, Casper R, Samara N, Hsieh T, Barzilay E, Orvieto R, Haas J. Does the endometrial receptivity array really provide personalized embryo transfer? J Assist Reprod Genet. 2018 Jul;35(7):1301-1305. doi: 10.1007/s10815-018-1190-9.Epub 2018 May 8.

Written By

Alice Albu and Dragoș Albu

Submitted: 02 March 2021 Reviewed: 28 May 2021 Published: 24 June 2021