ArticlePDF AvailableLiterature Review

The Disorders of Endometrial Receptivity in PCOS and Its Mechanisms

Authors:

Abstract

Polycystic ovary syndrome (PCOS) is a mysterious and complicated endocrine disease with the combination of metabolic, reproductive, psychological dysfunctions. Impaired endometrial receptivity and ovulation disorders/anovulation are both important causes of PCOS-related infertility. However, change in endometrium has never received the same attention as ovulatory dysfunction. Besides, putting emphasis on endometrial function may be more realistic for PCOS-related infertility, given the wide use of assisted reproductive technology. The present review focuses on the disorders of endometrial receptivity of patients with PCOS, summarizes the changes of the indicators of endometrial receptivity including leukemia inhibitory factor, homeobox genes A, pinopodes, αvβ3-integrin, and intercellular junctions and also analyzes the possible mechanisms of decreased endometrial receptivity and its relationship with the main endocrine and metabolic disorders of PCOS such as hyperandrogenism, inflammation, insulin resistance, and obesity. Despite several biomarkers have been found to be associated with decreased endometrial receptivity in PCOS, the clinical relevance of these findings still awaits future clarification.
REVIEW
The Disorders of Endometrial Receptivity in PCOS and Its Mechanisms
Nan-Xing Jiang
1,2
&Xue-Lian Li
1,2
Received: 22 February 2021 /Accepted: 19 May 2021
#Society for Reproductive Investigation 2021
Abstract
Polycystic ovary syndrome (PCOS) is a mysterious and complicated endocrine disease with the combination of metabolic,
reproductive, psychological dysfunctions. Impaired endometrial receptivity and ovulation disorders/anovulation are both impor-
tant causes of PCOS-related infertility. However, change in endometrium has never received the same attention as ovulatory
dysfunction. Besides, putting emphasis on endometrial function may be more realistic for PCOS-related infertility, given the
wide use of assisted reproductive technology. The present review focuses on the disorders of endometrial receptivity of patients
with PCOS, summarizes the changes of the indicators of endometrial receptivity including leukemia inhibitory factor, homeobox
genes A, pinopodes, αvβ3-integrin, and intercellular junctions and also analyzes the possible mechanisms of decreased endo-
metrial receptivity and its relationship with the main endocrine and metabolic disorders of PCOS such as hyperandrogenism,
inflammation, insulin resistance, and obesity. Despite several biomarkers have been found to be associated with decreased
endometrial receptivity in PCOS, the clinical relevance of these findings still awaits future clarification.
Keywords Polycystic ovary syndrome (PCOS) .Endometrial receptivity .Insulin resistance .Infertility
Introduction
Polycystic ovary syndrome (PCOS), one of the most common
endocrine disorders in women, affects 813% of women of
reproductive age [1]. As one of the main features, infertility
affects 40% of women with PCOS [2].
Ovulation disorders/anovulation is an important cause of
PCOS-related infertility. With the development of technology,
nowadays we are able to obtain high-quality oocytes by con-
trolled ovarian hyperstimulation (COH), furthermore, the
quality of fertilized eggs are guaranteed by in vitro fertilization
(IVF). However, literatures have shown that when an ideal
blastocyst is transplanted in vitro, the success rate of trans-
plantation and live birth are lower among the women with
PCOS comparing to normal controls [3,4]. At the same time,
during assisted reproductive technology (ART), using the oo-
cytes donated by PCOS women will not lower the overall
success rate [5]. These make us consider the endometrial re-
ceptivity may be another important factor of PCOS-related
infertility. This review aims to illustrate the situation of endo-
metrial receptivity in women with PCOS and discuss the pos-
sible mechanisms.
Endometrial receptivitymeans the ability of endometri-
um to accept the implantation of embryo. Zygote is a seed and
endometrium is soil. The entrance of blastocysts into the en-
dometrium is called implantation. There are three processes
involved in the period of implantation: location,adhesion
and invasion. In four seasons of a year, only spring is suit-
able for sowing. Similarly, for women of childbearing age,
endometrium receptivity is present only for a very short time
in one menstrual cycle, typically occurring in the 22th to 24th
days of the cycle in the midsecretory phase [6]. This fleeting
moment is called windows of implantation (WOI).
Endometrial receptivity mainly depends on the precise control
of estrogen (E2) and progesterone (P4) [7,8].
For infertile patients, there are no specific test to detect the
establishment of endometrial receptivity [9]. Methods like
hormone determination, ultrasonographic endometrial thick-
ness (EMT), spiral artery blood flow parameters, and uterine
artery blood flow parameters are proved to have some refer-
ence value [10], but without high accuracy [11]. Endometrial
receptivity array (ERA) is a unique innovative and objective
procedure for clinical endometrial evaluation [12]. It contains
*Xue-Lian Li
xllifc@fudan.edu.cn
1
Obstetrics and Gynecology Hospital, Fudan University,
Shanghai 200011, Peoples Republic of China
2
Shanghai Key Laboratory of Female Reproductive Endocrine
Related Diseases, Obstetrics and Gynecology Hospital, Fudan
University, Shanghai 200011, PeoplesRepublicofChina
Reproductive Sciences
https://doi.org/10.1007/s43032-021-00629-9
a finite number of genes involved in endometrial receptivity,
thus avoiding the use of whole genome microarrays, which
cuts down on costs and simplifies the data analysis [12].
Further, ERA is more accurate than histologic dating and
is a reproducible method for the diagnosis of endometrial
dating and receptivity status [13]. However, as a new geno-
mic tool, ERA test has its limitations, it could not identify
the most appropriate time for embryo transfer or detect
uterine diseases affecting implantation. The invasive nature
of ERA is also its shortcoming that cannot be ignored [14].
New technologies may be necessary to assess the endome-
trial aspect of implantation. Nevertheless, the discovery of
some modern molecules has brought a glimmer of hope to
the evaluation of endometrial receptivity [15]. Nowadays,
some indicators have been found to be closely related to
endometrial receptivity, such as leukemia inhibitory factor
(LIF), homeobox genes A (HOXA), pinopodes, αvβ3-
integrin, and intercellular junctions [16,17]. The detailed
explanations are as follows.
I. LIF is a kind of cytokine secreted by endometrial glands
and belongs to IL-6 family. LIF plays an amazingly important
role in embryo implantation during WOI [18,19].
II. HOXA-gene is expressed in the epithelial and stromal
cells of uterus. Rats deficient in HOXA-10 and HOXA-11
showed embryo implantation disorders [20]. Similarly, re-
search found the downregulation of HOXA-10 mRNA and
protein expression levels in part of infertility patients [21].
III. Pinopodes are mushroom-resembled protrusions lo-
cated on luminal epithelium observed under electron micro-
scope. During WOI, the quantity and the density of
pinopodes increased rapidly, indicating the preparation for
embryo implantation [22].
IV. Integrin, a kind of adhesion molecule commonly found
on cell membrane, is a heterodimer composed of αand β
subunits. Integrins are present in both epithelial cells and stro-
mal cells in utero [23]. Integrin-ligand interactions enhance
the migration, adhesion, and invasion of trophoblasts [24].
Among them, the relationship between αvβ3-integrin and en-
dometrial receptivity is the most prominent.
V. Intercellular junctions between endometrial epithelial
cells not only constitute a complete epithelial barrier to main-
tain the integrity of the endometrium and prevent invasion of
external pathogenic factors, but also play an important role in
establishment of endometrial receptivity. The abnormal local-
ization or expression of intercellular junctions can lead to the
failure of endometrial decidualization and abnormal embryo
implantation [25,26].
These factors mentioned above are closely related. High
expression ofHOXA-10 was associated with the development
and increase of pinopodes. HOXA-genes also regulate β3-
integrin subunit in endometrium [27]. αvβ3-integrin, located
on pinopodes, participates in the crosstalk between embryo
and endometrium. Pinopodes can also release LIF-
containing secretory vesicles into uterine cavity [28], which
can increase the adhesion of trophoblast to endometrium by
upregulating the expression of αvβ3-integrin [29]. And these
indicators also have complicated changes in patients with
PCOS.
The Whole Picture of Endometrial Receptivity
in PCOS
More than 10 years ago, there was a controversy about wheth-
er PCOS had a negative effect on endometrial function [30].
Nowadays, with the gradual deepening of research, sufficient
evidence has proved that hormonal disturbances, as well as
metabolic changes in PCOS patients, can both influence the
endometrial receptivity [31,32].
At gene level, there is a differential gene expression in
endometrium of PCOS detected by microarray evaluation
[30,33]. It is noteworthy that the downregulated genes in
endometrium of PCOS are closely related to cell membrane,
extracellular matrix, cell adhesion and invasion, cytoskeleton,
and so on. Whats more, most of these genes are involved in
steroid hormone synthesis, inflammation, and oxidative stress,
which are indispensable for the establishment of endometrial
receptivity [33]. The proteome pattern of endometrium during
WOI in PCOS patients is significantly different with that in
normal female [34,35]. These differences in transcription,
post-transcriptional modification and translation are often re-
lated to metabolism, cell cycle, DNA repair, apoptosis, signal
transduction [33] and may consequently cause endometrial
hyperplasia, cancer, and impaired endometrial receptivity in
PCOS patients. However, specific mechanism is still un-
known. If we can pick out some proteins strongly associated
with endometrial receptivity from the proteome, itspossible
to use them as drug targets for regulating suboptimal implan-
tation in PCOS. For instance, the protein complex consists of
osteopontin (OPN) and CD44 is an important adhesion mole-
cule in embryoimplantationthrough STAT1 and NF-κBpath-
ways [36]. Detection of CD44-OPN might be used to assess
the endometrial function of infertile patients in the future.
Recently, an article reviewed the importance of MicroRNAs
(miRNA) for female reproductive function [37]. At present,
human follicular fluids microarray profiling has proved the
significant difference of miRNA between PCOS patients and
normal female. Therefore, detection of miRNAs in peripheral
blood of PCOS patients may be used as one of the noninva-
sive methods to evaluate endometrial receptivity in the fu-
ture. NOTCH signaling pathway is widely present in verte-
brates and nonvertebrates and closely related to the differ-
entiation, as well as development of cells, tissues, and or-
gans. Whats more, NOTCH signaling pathway plays a
crucial role in embryo implantation. A study showed that
the expression of NOTCH3 was significantly increased in
Reprod. Sci.
PCOS women [38]. Changes in NOTCH signaling pathway
may be associated with decreased endometrial receptivity
in PCOS, but few studies have been done in this area up to
now [38,39].
The expression of LIF gene is obviously declined in en-
dometrial glandular epithelial cells in PCOS patients [40],
which is consistent with endometrial dysfunction and im-
plantation failure. LIF is regulated by estrogen and shows a
negative correlation, which is generally accepted at present
[41,42]. The precise regulation of sex hormones runs
through the whole pregnancy process, and the establish-
ment of endometrial receptivity is no exception. The regu-
lation of endometrial function by sex hormones may be
achieved through Wnt signaling pathway [43]. Generally
speaking, PCOS patients have upregulations of androgen
and estrogen, but a lower level of progesterone [44], which
may be a reason for the decrease of LIF in endometrium
with PCOS. However, there are few studies on how PCOS
affects endometrial receptivity through LIF-related path-
ways. The decrease of endometrial LIF in PCOS patients
may be result from inhibition of gene expression [40]. LIF
regulates endometrial receptivity via LIF/gp130-JAK/
STAT3 signaling pathway [45], but there is still a lack of
further researches on the upstream or downstream mole-
cules of this pathway. While the downstream of LIF-
STAT3 pathway may be through the transcription factor
named early growth response 1 (Egr1) [46]orkeyproteins
on endometrial lumen epithelial [45] to regulate endometri-
al receptivity. There is a research which holds the view that
its the adequate expression of LIFR, but not LIF, plays the
key role during the establishment of endometrial receptivity
[47]. But studies on LIFR and endometrial receptivity in
PCOS are still fruitless.
Endometrial biopsies obtained from PCOS women dem-
onstrated decreased HOXA10 mRNA [48].
Downregulation of HOXA-10 may affect endometrial re-
ceptivity by inhibiting the development of pinopodes [49].
Another article suggested that HOXA-10 is multifunctional
by regulating the proliferation, morphology and identity of
endometrial stromal cells and epithelium[50]. Laparoscopic
ovarian drilling (LOD), one of the treatments for PCOS, can
improve the levels of HOXA-10 and HOXA-11 mRNA in
endometrium of PCOS patients [51]. A previous study has
also found a decrease in αvβ3-integrin in endometrium of
PCOS patients [52],whichprobablycausedbythepersis-
tence of progesterone receptors (PR) in PCOS [53].
Recently, it has been proposed that epithelial β3-integrin
(EB3) can be used to evaluate endometrial receptivity [54].
Moreover, uterine fluid αvβ3-integrin has been proved as a
superior biomarker for embryo implantation [55].
Therefore, αvβ3-integrin may also be a potential target
molecule for evaluating the endometrial receptivity in
PCOS patients in the future [56].
The Effect of the Endocrine and Metabolic
Disorders of PCOS on Endometrial Receptivity
Hyperandrogenism (HA) and Endometrial Receptivity
in PCOS
Androgen regulates the endometrial receptivity by binding to
androgen receptors (AR), which is widely distributed in
glands, luminal epithelium, and stromal of uterus. The mech-
anisms of PCOS-related HA are as follows: PCOS ovary theca
cells expressed elevated 17α-hydroxylase/C17,20 lyase/17-
hydroxylase and 3β-HSD enzyme under pathologically stim-
ulated by hyperinsulinemia and overmuch LH, driving in-
creased androgen[57,58]. IR, pancreatic B cell dysfunction
and lipotoxicity [59,60] will reduce sex hormone-binding
globulin (SHBG), resulting in the soar of free testosterone
[61]. Meanwhile, androgen cannot convert to estrogen due
to the suppression of aromatase (CYP19), which is a key
enzyme for estrogen synthesis [62,63]. In turn, HA inhibits
the function of CYP19 as well, reinforcing a vicious cycle.
The latest research shows that the suppressing aromatase ex-
pression may connected to HUPCOS, a newfound lncRNA
[64]. PCOS-related HA also affects gene expression profiles,
including gene sets associated with autophagy, insulin signal-
ing, cell cycle, and glucose metabolism both in human endo-
metrium and during in vitro decidualization[65,66].
As a transcription factor, Wilms tumor suppressor gene
(WT1) expressed on endometrial stromal cells during WOI
[67,68]. Increased androgen receptors (AR) on endometrium
followed by the decline of WT1. Elevated Bcl2, which is one
of the targets of WT1, thereby blocks the cell cycle via p27
and impairs endometrial function. On the other hand, the sup-
pression of WT1 restrains epidermal growth factor receptor
(EGFR). The combination of EGFR and its ligand EGF is
bound up with the implantation of eggs during WOI. To
sum up, impaired endometrial receptivity of PCOS-related
infertility may closely relate to HA-WT1 pathway. Also, re-
covery of WT1 expression may be one of the reasons for the
effectiveness of antiandrogen therapy in PCOS [6870].
Interestingly, contrast with endometrium, WT1 is
overexpressed in PCOS womens ovaries, implicating the ef-
fects of the WT1 pathway are multifarious [71].
Both clinical and animal studies have shown that PCOS is
associated with the downregulation of HOXA-10 and HOXA-
11[40,48]. HOXA genes go hand in hand with steroid hor-
mones. In human endometrium, HOXA-10 increased in re-
sponse to elevated estrogen or progesterone. Androgen, how-
ever, has an opposite effect [48]. A dose-responsive decrease
of HOXA-10 mRNA level caused by testosterone was con-
firmed in vitro [48]. Similar phenomenon was observed in
extrauterine tissue. Testosterone at 10
6
M can significantly
decrease HOXA10 protein in granulosa cells (GCs) [72].
LOD is helpful to improve the levels of HOXA-10 and
Reprod. Sci.
HOXA-11 mRNA in PCOS endometrium, which could be
explained by the remission of HA after surgery [51].
However, not all patients of PCOS have HA and therefore
there may exists more than one reasons responsible for the
altered expression of HOXA-gene.
In addition, the expression of various proteins in endome-
trial cells is androgen-dependent [73]. Ongoing HA may alter
the expression of proteins associated with endometrial devel-
opment and embryo implantation, which may be one of the
causes of decreased endometrial receptivity and miscarriage in
PCOS [73,74]. Elevated androgen may lead to a decrease the
level of CDKN2A in endometrium, thereby affecting the ac-
tivity of cyclin-dependent kinase (CDK). In vitro experiments
showed that the reduction of CDKN2a in Ishikawa cells re-
sulted in decreased migration, proliferation, invasion, and the
rate of Jar spheroid attachment to Ishikawa cell monolayer
[73]. L-selectin Ligand (MECA-79), an implantation protein
specifically expressed in WOI, reduced as well by the treat-
ment of testosterone [75]. HA causes the delayed
decidualization of endometrium, which is related to a
coregulator protein of AR named MAGEA-11. This may also
be a reason for PCOS-related infertility, but the specific mech-
anism is still unknown [70].
Testosterone can affect the expression of αvβ3-integrin, E-
Cadherin and Mucin-1[23]. It was reported that persistence of
PR in PCOS affects the expression of αvβ3-integrin [53]. The
downregulation of integrin after high-dose testosterone treat-
ment may be due to the counteraction of progesterone, which
causes the failure of maternalfetal recognition, and then im-
pairs the establishment of pregnancy [23].
HA also affects intercellular junctions, thereby impairing
endometrial receptivity. Recently, an in-vivo study investigat-
ed the correlation between HA and intercellular junctions in
endometrium. The composition and proportion of claudin-4
and occludin determine the permeability and selectivity of
endometrial tight junction. HA can decrease the expression
of them, thus, impair endometrial tight junction [76].
Furthermore, in animal tests, the expression of connexin
26 improved, while connexin 43 were opposite after testos-
terone treatment [77]. Connexin 26 is acknowledged as a
marker of endometrial rejection. As for connexin 43, we
don't know yet if we can consider it as a marker of endo-
metrial receptivity from the evidence we have now. It has
been proved that HA can affect the intercellular junctions of
endometrium, thus affecting endometrial function and em-
bryo adhesion, which may be one of the reasons for the
decreased endometrial receptivity in PCOS patients.
However, these experiments have limitations. PCOS, a syn-
drome with multiple pathological features, is known for its
complexity and mystery. However, these experiments took
androgen as a single variable. No researches related to en-
dometrial intercellular junctions under complete PCOS
conditions have been published so far.
What's more, HA and IR can cause endometrial disorder of
oxidation and antioxidation by changing mitochondrial func-
tion [78]. The link between excessive ROS and PCOS has
been proved[79].
All in all, due to its complexity, follow-up studies are still
needed to figure out the relevance of HA and endometrial
receptivity of PCOS patients.
Inflammation and Endometrial Receptivity in PCOS
The whole process of pregnancy requires the precise regula-
tion of immune cells and cytokines, including embryo implan-
tation. Immune cells, especially uterine NK cells, regulatory T
cells (Treg), macrophages and dendritic cells (DC), play a key
role in inducing immune tolerance of blastocysts [8082].
Excessive inflammation exists in certain diseases like PCOS,
endometriosis and therefore resulting in adverse pregnancy
outcomes. Some of the cytokines involved in the inflammato-
ry response are secreted by endometrial cells, while others are
secreted by immune cells recruited into uterus through blood
vessels. The whole process is regulated by E2 and P4 under
physiological conditions. The concentration gradient of cyto-
kines on endometrium guides the fertilized egg to a suitable
site for implantation [80,81,83]. Disorder of inflammatory
mediators can cause implantation failure and abnormal pla-
centa formation in mice and human [84].
Recent studies have found that miRNAs have a certain
effect on the occurrence and development of PCOS-related
inflammation. miR-4651 may participate in the inflammatory
response through leukocyte transendothelial migration by reg-
ulating target genes [85]. The expression of MiR-146a and
MiR-223 were significantly decreased in infertile women with
metabolic syndrome (MS). MiR-146a inhibits IRAK1-
TRAF6-NF-κB pathway and the downregulation of MiR-
146a is responsible for increased proinflammatory cytokine
and inflammatory response. The decreased expression of
miR-223 was related to the production of IL-1βand IL-
6[86]. Microarray profiling has shown that miRNA expres-
sion in PCOS patients is different from that in normal women
[37]. Detecting inflammation state in endometrium via
miRNA level to evaluate endometrial receptivity in PCOS
may be one of the future directions.
Chronic inflammation of PCOS is systemic. Higher levels
of serum inflammatory markers including IL-6, IL-16, IL-18,
TNF-α, and CRP are existing in PCOS [8790]. Similarly,
low-grade chronic inflammation can also be detected in endo-
metrium [87]. Besides, it should be noted that whatever way
weight goes, inflammation shows in PCOS. Obesity is just an
aggravating factor.
The changes of implantation environment caused by in-
flammatory mediators may be a reason for impaired endome-
trial receptivity in PCOS patients. Piltonen et al. [84]pointed
out that the levels of IL-6, IL-8, monocyte chemoattractant
Reprod. Sci.
protein-1 (MCP-1), granulocyte-macrophage colony-stimulat-
ing factor (GM-CSF), and RANTES are markedly elevated in
undecidualized endometrial stromal fibroblasts (eSF) of
PCOS women. The disordered chronic inflammatory state of
PCOS women is also reflected in the downregulation of some
key cytokines. In an animal test [91], the expression of IL-1 of
PCOS group was significantly downregulated, accompanied
with declined thickness of endometrium. Stanniocalcin-1
(STC-1), a protective factor that reduce inflammatory stress,
is coexpressed with endometrial receptivity markers. The ex-
pression of STC-1 altered in PCOS patients, leading to a lack
of rescue mechanism in endometrium when faced with in-
flammatory stress. In addition, lifestyle changes cannot recov-
er STC-1 expression. Since STC-1 knockout mice has been
shown to be fertile, so it probably not a necessary factor for
implantation, but its dysfunction may, to some extent, worsen
the endometrial implantation microenvironment [92]. A study
analyzed the relationship between inflammatory chemokine
named fractalkine (FKN) and PCOS. PCOS patients have
higher circulating FKN levels than controls, and FKN level
is positively correlated with BMI, IR, inflammatory marker
CRP and total testosterone [93]. That is to say, FKN may
plays an important role in the intricated pathophysiology of
PCOS. Some inflammatory factors have been shown to be
associated with the expression of HOXA-gene, but the mech-
anism remains to be further investigated [94,95].
In recent years, researches on advanced glycation end
Products (AGEs) have received increasing attention. AGEs
are the end product of a nonenzymatic glycosylation reaction
(Maillard reaction) [96,97]. The accumulation of AGEs has
been confirmed to be closely related to many diseases, such as
diabetes and cardiovascular disease [98100]. In this section,
we mainly describe the potential link among AGEs, inflam-
mation and decreased endometrial receptivity in PCOS.
AGEs act by binding to the membrane receptor (RAGEs).
AGE-RAGE combination activates a complex set of signaling
pathways so as to promoting downstream proinflammatory
signals including chemokines and cytokines [101]. There is
another soluble receptor called sRAGE, the product of both
splicing of RAGE-gene and cleavage of membrane-bound
RAGE by proteases belonging to the zinc-dependent
metzincin family of metalloproteases [102]. sRAGE compet-
itively binds to AGEs, thereby inhibiting the downstream pro-
inflammatory signals aroused by AGEs-RAGE combination.
Hence sRAGE is generally considered to be a protective
antiinflammatory receptor [100,103]. AGE-RAGE binding
leads to oxidative stress and inflammation in cells. As a pro-
tective factor, sRAGE can reduce inflammation level in PCOS
patients, which has been confirmed in human follicular cells
[100,103]. However, no article targeting to endometrium was
found. The significant difference in pregnancy outcome ob-
served in obese women probably due in part to elevated AGEs
in endometrium and the resulting inflammation [104]. The
influence of AGEs on VEGF has been detected in retinal
pigment epithelium [105]. Elevated AGEs have also been
demonstrated to cause damage to endothelial cell in arterio-
sclerosis of PCOS. And AGEs are related to reproductive
dysfunction in PCOS patients [100]. VEGF is an important
marker of endometrial receptivity, which is closely bond to
endometrial function and involved in placenta formation [91].
Endothelial injury impairs the physiological function of endo-
metrium as well. Unfortunately, there are few studies concen-
trating to the relationship between AGEs and endometrial re-
ceptivity of PCOS by far.
Insulin Resistance (IR) and Endometrial Receptivity in
PCOS
IR refers to the complicated pathological state of inadequate
response to insulin. Insulin -insulin receptor (INSR) binding
regulates glucose and lipid metabolism mainly through PI3-K
and Akt/PKB pathway. GLUT4 is a kind of insulin-
responsive glucose transporter. GLUT4 is transferred from
intracellular vesicles to plasma membrane so as to realize the
uptake of glucose. The activation of PI3-K and AKT/PKB
pathway also leads to serine phosphorylation of GSK3 to pro-
motes glycogen synthesis. Dysfunction of any link in insulin
metabolic pathway will lead to IR. [106108]
Appropriate glucose metabolism is a key factor for embryo
implantation. Studies have shown that the oocytes from PCOS
patients with IR, neither its development or quality was affect-
ed after invitro culture. Unfortunately, the pregnancy rate was
significantly declined, which suggests that the foundation of
declined pregnancy rate caused by IR is the damage of endo-
metrium function and the obstacle of embryo implantation.
[108,109]
About 5070% of PCOS patients have IR and concomitant
hyperinsulinemia [110]. There are multiple reasons for PCOS-
related IR, including HA, obesity, and inflammation.
Furthermore, these factors do not work alone but jointly form
an intricate network. PCOS women are prone to obesity, over-
much adipose tissue can affect insulinsensitivity.The death of
hypertrophic adipocytes activates IR and HA through inflam-
mation [111]. In human endometrial stromal cells, overload
TNF-αhas a negative effect on insulin sensitivity by reducing
adiponectin signal and blocking the transport of GLUT-4 to
membrane, resulting in abnormal energy metabolism [112,
113], which may be one of the reasons for impaired endome-
trial receptivity in PCOS. Meanwhile, excessive IL-6 reduces
the activation of IRS1 and AKT. When obesity and PCOS are
present, the destruction of insulin signaling is intensified
[114]. Besides, a research found that decreased oxidation of
cortisol might contribute to endometrial IR by inhibiting insu-
lin signaling pathway via induction of PTEN expression in
endometrial epithelial cells of PCOS women [108]. Besides,
HA aggravates IR as well [110]. In addition, sympathetic
Reprod. Sci.
nervous system (SNS) dysfunction is involved in the occur-
rence of IR. It was observed that the activation of SNS pro-
motes IR through increased lipolysis and inflammatory cas-
cades of P53 activation in adipose tissue in animal models of
heart failure [115]. Increased SNS activity can also be detected
in patients with PCOS because of inflammation. Which sug-
gests that low-grade inflammation in PCOS may contribute to
IR by mediating SNS dysfunction [110].
AGEs cause IR by affecting the insulin-mediated PI3K/
AKT signaling pathway [97]. AGEs inhibit insulin production
in pancreatic βcells and prevent membrane translocation of
GLUT4. However, these researches are mostly based on he-
patocytes and granulosa cells [98,116], studies on endometri-
um are still lacking.
It was known that PCOS affects glucose and energy me-
tabolism in endometrium by reducing GLUT4 expression,
so as to generate impaired endometrial receptivity [108].
However, a recent paper cast doubt on that point, the study
found that GLUT4 mRNA was not detected in the endome-
trium of PCOS or control group at all [66]. Therefore, the
link between PCOS-related IR and impaired endometrial
receptivity is still controversial and needs further studies
[66,108].
Obesity and Endometrial Receptivity in PCOS
Obesity is related to the above-mentioned pathophysiological
phenomena, such as HA, IR, low-grade chronic inflammation,
and excessive accumulation of AGEs [117]. Moreover, previ-
ous studies have shown that though some metabolic changes
caused by PCOS are independent of obesity, obesity undoubt-
edly an aggravating factor in metabolic disorders [117,118].
The influence of obesity, especially central obesity, on endo-
metrial receptivity of PCOS women cannot be ignored.
During WOI, obese women present significantly different en-
dometrial gene expression from the control group, which is
more obvious when PCOS or infertility is associated [119,
120].
In PCOS patients, there is a vicious circle between obesity
and other metabolic disturbances, thereby weakening the
function of endometrium (Fig. 1). Obesity exacerbates low-
grade chronic inflammation [121] and causes a drop in serum
SHBG level [122], thus aggravating HA. In addition, obesity
promote IR and overaccumulation of AGEs [121,123,124].
At the same time, these metabolic changes make an impact on
the occurrence and aggravation of obesity among PCOS pa-
tients. For instance, HA and IR can inhibit lipolysis and pro-
mote fat formation [125]. Overload inflammatory adipokine
can increase fat production in turn [126]. Lifestyle changes
such as weight loss or exercise can help PCOS patients alle-
viate symptoms and restore endometrial function [121,122,
127,128].
The Current Therapy Methods
and Endometrial Receptivity in PCOS
The specific treatment of implantation disorder in PCOS is
still no consensus, but some therapy methods have been
proved to be effective for the recovery of endometrial function
in PCOS.
The effect of metformin on PCOS endometrium has
gradually attracted more attention. Metformin improves
the systemic and intrauterine environment of PCOS pa-
tients by reducing HA, IR, and LH [129]. And studies have
confirmed that metformin may downregulate the expres-
sion of miR-491-3p and miR-1910-3p, thereby increasing
the levels of HOXA-10 and β3-integrin [130]. Moreover,
metformin is conducive to reverse the abnormal transcrip-
tion of endometrial autophagy genes caused by HA in
PCOS patients [65]. Nonetheless, the role of metformin in
endometrial receptivity with PCOS is not entirely benefi-
cial. Metformin seems to have a tendency to delay histolog-
ical glandular maturation [131]. Moreover, it is dose-
dependent on the proliferation of endometrial stromal cells
[132]. Inappropriate use of metformin may be counterpro-
ductive, while the optimal effective concentration is still
wrapped in the veil of mystery.
HA can inhibit the proliferation of endometrial stromal
cells. Restoring androgen level to normal range is benefi-
cial to the recovery of endometrial function in PCOS pa-
tients [132]. After LOD surgery, the increased expression
of HOXA-gene in endometrium of PCOS patients may be
relatedtotheremissionofHA[51]. The antiandrogen drug
named flutamide is effective in improving decidualization
and angiogenesis in uterine [133]. LIF is a marker of endo-
metrial receptivity, but studies have shown that treatment
with LIF cannot improve the success rate of embryo im-
plantation in infertility patients [134]. Myo-inositol
(MYO), an insulin sensitizer, is helpful to improve IR in
PCOS.MYOmayimproveendometrialfunctioninPCOS
patients through the increase of AMPK activation.
Furthermore, compared with metformin, MYO has a better
tolerance by women [135].
AGE/sRAGE may be a potential biomarker and a
promising therapeutic target for PCOS. In a 1-year ran-
domized controlled trial [136], scholars found that diet
low in AGEs (L-AGE) do good for IR in obese people
with MS, which suggests that AGEs might have a nega-
tive effect on insulin metabolism. AGE level affected
decidualization and endoplasmic reticulum stress of endo-
metrial stromal cells, and significantly inhibited the adhe-
sionandinvasionofembryo[104].RegardingtoL-AGE
diet, it gives us a hint, whether it is MS, type-2 diabetes,
or PCOS, L-AGE diet may be an adjuvant treatment
worth trying. As for PCOS-related infertility, if purely
from the perspective of endometrial receptivity. L-AGE
Reprod. Sci.
diet is possible to increase insulin sensitivity, reduce in-
flammation level and androgen, thereby alleviating resis-
tance to embryo implantation [136]. Last but not least,
appropriate exercise and normal range of BMI is essential
to improve the endometrial receptivity for PCOS patients
[121,122,127,128].
Vicious circle
Vicious circle
HA
HA
Inflammation
Inflammation
IR
IR
•HA/IR/inflammation promotes fat
formation
Ref. [125, 126]
•Obesity exacerbates inflamma-
tion and IR
•Obesity aggravates HA by
reducing SHBG
Ref. [121-124]
f.
f.
INSR and SNS
dysfunction
Ref. [114]
e.
e.
Elevated TNF-α and
macrophages
Ref. [89, 112, 143]
a.
a.
•Reduction in SHBG
•Increase in LH secretion
•Affects P450c17 enzyme
Ref. [125, 139-141]
b.
b.
Hyperandrogenic women have
a higher risk of developing IR
Ref. [142]
d.
d.
HA causes fat cell
hypertrophy and
inflammation
Ref. [110]
c.
c.
Cytokines cause HA
by theca cells
Ref. [89]
Fig. 1 The interaction and
vicious cycle among metabolic
disorders of PCOS
Table 1 The key findings of PCOS-metabolic/hormonal disturbances and their association with endometrial receptivity
PCOS-metabolic/hormonal
disturbances
The influence on endometrial receptivity Ref.
Hyperandrogenism Decreases the expression of LIF gene through Wnt signaling pathway.
Affects the expression of HOXA-gene and αvβ3-integrin.
Inhibits the development of pinopodes by the down-regulation of HOXA-10.
Decreases endometrial tight junction and proportion of claudin-4 and occludin and improves the expression
of connexin 26.
Alters the expression of receptivity-related proteins, e.g., CDK signal pathway, MECA-79, and
MAGEA-11.
Impairs endometrial receptivity through HA-WT1 pathway.
[40,43]
[23,48,
72]
[49,75]
[76,77]
[70,73,
75]
[68]
Inflammation Some inflammatory factors are associated with the expression of HOXA-gene.
AGE-RAGE binding leads to oxidative stress and inflammation in cells, and impairs endometrial function by
VEGF.
Disorder of inflammatory mediators leads to impaired endometrial receptivity in PCOS.
[94,95]
[91,100]
[84,91]
Insulin resistance Causes endometrial disorder of oxidation and antioxidation by changing mitochondrial function.
PCOS affect glucose metabolism in endometrium by reducing GLUT4 expression.
[78]
[108]
Obesity During WOI, obese women present different endometrial gene expression from the control group, which is
more obvious when PCOS or infertility is associated.
There is a vicious circle between obesity and other PCOS-metabolic/hormonal disturbances. (Fig. 1)
[119,120]
[104,117,
118]
Reprod. Sci.
Conclusions and Prospects
In the present review, we discussed the factors related to
endometrial receptivity in PCOS. These factors also have
potential to be used as indicators for evaluating endometrial
receptivity in the future [1517]. The decrease of endome-
trial receptivity is one of the most important causes of
PCOS-related infertility. Yet it has never received the same
attention as ovulatory dysfunction. Part of relevant patho-
physiological changes in PCOS has been confirmed to be
associated with decreased endometrial receptivity. The key
findings of PCOS-metabolic/hormonal disturbances and
their association with endometrial receptivity are shown
in Table 1. To date, however, little progress has been
achieved for clinical integration in terms of prognostic tests
and treatments for suboptimal endometrial receptivity. We
are still unable to evaluate endometrial receptivity for
PCOS patients in an effective and noninvasive manner
[911,14]. Regarding PCOS-related infertility, the specific
mechanism of endometrial receptivity changes still needs
further researches.
However, the veil of mist hiding the truth from view will
gradually fade away. Modern molecules have shown great
potential as biomarkers in diagnosis, prognosis, and also as
possible therapeutic targets in PCOS-related infertility.
Currently, endometrial dysfunction is the biggest obstacle to
the treatment of embryo implantation difficulties [137,138].
In contrast to endometrial biopsy, as a noninvasive, accurate
marker, systematic profiling of indicators of endometrial re-
ceptivity including LIF, HOXA, pinopodes, αvβ3-integrin,
and intercellular junctions is needed both to identify implan-
tation defects in PCOS women and to signal the optimal
timing for embryo transfer. In the future, further exploration
regarding endometrial receptivity in PCOS is bound to be
difficult but full of infinite hopes and expectations.
Acknowledgements The authors would like to thank the support from
Natural Science Foundation from Science and Technology Commission
of Shanghai Municipality.
Code availability Not applicable.
Funding This study was funded by Natural Science Foundation from
Science and Technology Commission of Shanghai Municipality (grant
no. 17ZR1403100 to Xue-Lian Li).
Data availability All data generated or analyzed during this study are
included in this published article and its supplementary information files.
Declarations
Ethics approval Not applicable.
Consent to participate Not applicable.
Consent for publication Not applicable.
Competing interests The authors declare that they have no competing
interests.
References
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):284155.
2. Eden JA. The polycystic-ovary-syndrome presenting as resistant
acne successfully treated with cyproterone-acetate. Med J Aust.
1991;155(10):67780.
3. Adam H, S.L.T B, MacDougall J, Jacobs HS. Miscarriage rates
following in-vitro fertilization are increased in women with poly-
cystic ovaries and reduced by pituitary desensitization with
buserelin. Hum Reprod. 1993;8(6):95964.
4. Steiner N, et al. A comparison of IVF outcomes transferring a
single ideal blastocyst in women with polycystic ovary syndrome
and normal ovulatory control. Arch Gynecol Obstet. 2020;8.
5. Ashkenazi J, Farhi J, Orvieto R, Homburg R, Dekel A, Feldberg
D, et al. Polycystic-ovary-syndrome patients as oocyte donors
the effect of ovarian stimulation protocol on the implantation rate
of the recipient. Fertil Steril. 1995;64(3):5647.
6. Wilcox AJ, Baird DD, Wenberg CR. Time of implantation of the
conceptus and loss of pregnancy. N Engl J Med. 1999;340(23):
17969.
7. Griffiths RM, Pru CA, Behura SK, Cronrath AR, McCallum M,
Kelp NC, et al. AMPK is required for uterine receptivity and
normal responses to steroid hormones. Reproduction.
2020;159(6):70717.
8. Paulson RJ. Introduction: Endometrial receptivity: evaluation, in-
duction and inhibition. Fertil Steril. 2019;111(4):60910.
9. Cohen AM, et al. Comparing endometrial receptivity array to
histologic dating of the endometrium in women with a history of
implantation failure. Syst Biol Reprod Med. 2020:18.
10. Tong R, Zhou Y, He Q, Zhuang Y, Zhou W, Xia F. Analysis of
the guidance value of 3D ultrasound in evaluating endometrial
receptivity for frozen-thawed embryo transfer in patients with re-
peated implantation failure. Ann Transl Med. 2020;8(15):944.
11. Griesinger G, Trevisan S, Cometti B. Endometrial thickness on the
day of embryo transfer is a poor predictor of IVF treatment out-
come. Human reproduction open. 2018;2018(1):hox031.
12. Díaz-Gimeno P, et al. A genomic diagnostic tool for human en-
dometrial receptivity based on the transcriptomic signature. Fertil
Steril. 2011;95(1):50-6060.e1-15.
13. Díaz-Gimeno P, Ruiz-Alonso M, Blesa D, Bosch N, Martínez-
Conejero JA, Alamá P, et al. The accuracy and reproducibility
of the endometrial receptivity array is superior to histology as a
diagnostic method for endometrial receptivity. Fertil Steril.
2013;99(2):50817.
14. Cozzolino M, Diaz-Gimeno P, Pellicer A, Garrido N. Evaluation
of the endometrial receptivity assay and the preimplantation ge-
netic test for aneuploidy in overcoming recurrent implantation
failure. J Assist Reprod Genet. 2020;37:298997.
15. Craciunas L, Gallos I, Chu J, Bourne T, Quenby S, Brosens JJ,
et al. Conventional and modern markers of endometrial receptiv-
ity: a systematic review and meta-analysis. Hum Reprod Update.
2019;25(2):20223.
16. Lopes IM, et al. Endometrium in women with polycystic ovary
syndrome during the window of implantation. Revista da
Associacao Medica Brasileira (1992). 2011;57(6):7029.
Reprod. Sci.
17. Ribeiro Soares Lopes IM, et al. Histomorphometric analysis and
markers of endometrial receptivity embryonic implantation in
women with polycystic ovary syndrome during the treatment with
progesterone. Reprod Sci. 2014;21(7):9308.
18. Bhatt H, Brunet LJ, Stewart CL. Uterine expression of leukemia
inhibitory factor coincides with the onset of blastocyst implanta-
tion. Proc Natl Acad Sci U S A. 1991;88(24):1140812.
19. Chen JR, Cheng JG, Shatzer T, Sewell L, Hernandez L, Stewart
CL. Leukemia inhibitory factor can substitute for nidatory estro-
gen and is essential to inducing a receptive uterus for implantation
but is not essential for subsequentembryogenesis. Endocrinology.
2000;141(12):436572.
20. Akbas GE, Taylor HS. HOXC and HOXD gene expression in
human endometrium: Lack of redundancy with HOXA paralogs.
Biol Reprod. 2004;70(1):3945.
21. Chen H, et al. Expression of Hoxa-10 mRNA in the endometrium
of women with unexplained infertile. Journal of China Medical
University. 2003;32(3):2523.
22. Panzan MQ, Junior JMS, da Motta ELA, Haapalainen EF, de
Jesus Simoes M, Baptista HA, et al. Metoclopramide-induced
hyperprolactinaemia caused marked decline in pinopodes and
pregnancy rates in mice. Hum Reprod. 2006;21(10):251420.
23. Mokhtar HM, Giribabu N, Salleh N. Testosterone down-regulates
expression of alpha v beta 3-Integrin, E-Cadherin and Mucin-1
during uterine receptivity period in rats. Sains Malaysiana.
2018;47(10):250917.
24. Adu-Gyamfi, E.A., et al.. The involvement of cell adhesion mol-
ecules, tight junctions, and gap junctions in human placentation.
Reproductive Sciences., 2020.
25. Poon CE, Madawala RJ, Dowland SN, Murphy CR. Nectin-3 is
increased in the cell junctions of the uterine epithelium at implan-
tation. Reprod Sci. 2016;23(11):158092.
26. Wu JJ, Taylor RN, Sidell N. Retinoic acid regulates gap junction
intercellular communication in human endometrial stromal cells
through modulation of the phosphorylation status of connexin 43.
J Cell Physiol. 2013;228(4):90310.
27. Daftary GS, Troy PJ, Bagot CN, Young SL, Taylor HS. Direct
regulation of beta(3)-integrin subunit gene expression by
HOXA10 in endometrial cells. Mol Endocrinol. 2002;16(3):
5719.
28. Rarani FZ, Borhani F, Rashidi B. Endometrial pinopode bio-
markers: molecules and microRNAs. J Cell Physiol.
2018;233(12):914558.
29. Chung TW, Park MJ, Kim HS, Choi HJ, Ha KT. Integrin alpha V
beta 3 and alpha V beta 5 are required for leukemia inhibitory
factor-mediated the adhesion of trophoblast cells to the endome-
trial cells. Biochem Biophys Res Commun. 2016;469(4):93640.
30. Qiao J, Wang L, Li R, Zhang X. Microarray evaluation of endo-
metrial receptivity in Chinese women with polycystic ovary syn-
drome. Reprod BioMed Online. 2008;17(3):42535.
31. Ejzenberg D, Gomes TJO, Monteleone PAA, Serafini PC, Soares-
Jr JM, Baracat EC. Prognostic factors for pregnancy after intra-
uterine insemination. Int J Gynecol Obstet. 2019;147(1):6572.
32. Giordano MV, Giordano LA, Gomes RCT, Simões RS, Nader
HB, Giordano MG, et al. The evaluation of endometrial sulfate
glycosaminoglycans in women with polycystic ovary syndrome.
Gynecol Endocrinol. 2015;31(4):27881.
33. Rashid N, Nigam A, Jain SK, Naqvi SH, Wajid S. Proteomic sift
through serum and endometrium profiles unraveled signature pro-
teins associated with subdued fertility and dampened endometrial
receptivity in women with polycystic ovary syndrome. Cell Tissue
Res. 2020;380(3):593614.
34. Alikhani, M., et al.. Proteome analysis of endometrial tissue from
patients with PCOS reveals proteins predicted to impact the dis-
ease. Molecular Biology Reports.
35. Amjadi F, Mehdizadeh M, Ashrafi M, Nasrabadi D, Taleahmad S,
Mirzaei M, et al. Distinct changes in the proteome profile of en-
dometrial tissues in polycystic ovary syndrome compared with
healthy fertile women. Reprod BioMed Online. 2018;37(2):184
200.
36. Paravati R, de Mello N, Onyido EK, Francis LW, Brüsehafer K,
Younas K, et al. Differential regulation of osteopontin and CD44
correlates with infertility status in PCOS patients. Journal of
Molecular Medicine-Jmm. 2020;98(12):171325.
37. Kamalidehghan B, Habibi M, Afjeh SS, Shoai M, Alidoost S,
Almasi Ghale R, et al. The importance of small non-coding
RNAs in human reproduction: a review article. Appl Clin Genet.
2020;13:111.
38. Amjadi F, Salehi E, Zandieh Z, Rashidi M, Taleahmad S, Javedani
Masrour M, et al. Comparative evaluation of NOTCH signaling
molecules in the endometrium of women with various gynecolog-
ical diseases during the window of implantation. Iranian Journal of
Basic Medical Sciences. 2019;22(4):42631.
39. Xie Q, Cheng Z, Chen X, Lobe CG, Liu J. The role of Notch
signalling in ovarian angiogenesis. Journal of ovarian research.
2017;10(1):13.
40. Kara M, Ozcan SS, Aran T, Kara O, Yilmaz N. Evaluation of
endometrial receptivity by measuring HOXA-10, HOXA-11,
and leukemia inhibitory factor expression in patients with poly-
cystic ovary syndrome. Gynecology and Minimally Invasive
Therapy-Gmit. 2019;8(3):11822.
41. Li Z, Zhu Y, Li H, Jiang W, Liu H, Yan J, et al. Leukaemia
inhibitory factor in serum and follicular fluid of women with poly-
cystic ovary syndrome and its correlation with IVF outcome.
Reprod BioMed Online. 2018;36(4):4839.
42. Li S-Y, et al. Impaired receptivity and decidualization in DHEA-
induced PCOS mice. Sci Rep. 2016;6.
43. Mehdinejadiani S, Amidi F, Mehdizadeh M, Barati M, Pazhohan
A, Alyasin A, et al. Effects of letrozole and clomiphene citrate on
Wnt signaling pathway in endometrium of polycystic ovarian syn-
drome and healthy women. Biol Reprod. 2019;100(3):6418.
44. Hu C, et al. Immunophenotypic profiles in polycystic ovary syn-
drome. Mediat Inflamm. 2020;2020 (no pagination:5894768.
45. Pawar S, Starosvetsky E, Orvis GD, Behringer RR, Bagchi IC,
Bagchi MK. STAT3 regulates uterine epithelial remodeling and
epithelial-stromal crosstalk during implantation. Mol Endocrinol.
2013;27(12):19962012.
46. Liang X-H, Deng WB, Li M, Zhao ZA, Wang TS, Feng XH, et al.
Egr1 Protein Acts Downstream of estrogen-leukemia inhibitory
factor (LIF)-STAT3 pathway and plays a role during implantation
through targeting Wnt4. J Biol Chem. 2014;289(34):2353445.
47. Margioula-Siarkou C, et al. LIF and LIF-R expression in the en-
dometrium of fertile and infertile women: a prospective observa-
tional case-control study. Mol Med Rep. 2016;13(6):47218.
48. Cermik D, Selam B, Taylor HS. Regulation of HOXA-10 expres-
sion by testosterone in vitro and in the endometrium of patients
with polycystic ovary syndrome. J Clin Endocrinol Metab.
2003;88(1):23843.
49. Jana SK, Banerjee P, Mukherjee R, Chakravarty B, Chaudhury K.
HOXA-11 mediated dysregulation of matrix remodeling during
implantation window in women with endometriosis. J Assist
Reprod Genet. 2013;30(11):150512.
50. Bagot CN, Kliman HJ, Taylor HS. Maternal Hoxa10 is required
for pinopod formation in the development of mouse uterine recep-
tivity to embryo implantation. Dev Dyn. 2001;222(3):53844.
51. Senturk S, Celik O, Dalkilic S, Hatirnaz S, Celik N, Unlu C, et al.
Laparoscopic ovarian drilling improves endometrial homeobox
gene expression in PCOS. Reprod Sci. 2020;27(2):67580.
52. Apparao KBC, Lovely LP, Gui Y, Lininger RA, Lessey BA.
Elevated endometrial androgen receptor expression in women
Reprod. Sci.
with polycystic ovarian syndrome. Biol Reprod. 2002;66(2):297
304.
53. 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(4):101726.
54. Marron K, Harrity C, Dunne H, Shkrobot L, Kennedy J.
Cytometric assessment of uterine receptivity via epithelial beta 3
integrin expression. Reprod BioMed Online. 2019;39(2):294
303.
55. Wang L, Lv S, Mao W, Pei M, Yang X. Assessment of endome-
trial receptivity during implantation window in women with un-
explained infertility. Gynecol Endocrinol. 2020;36(10):91721.
56. Lopes IM, et al. Histomorphometric analysis and markers of en-
dometrial receptivity embryonic implantation in women with
polycystic ovary syndrome during the treatment with progester-
one. Reprod Sci. 2014;21(7):9308.
57. Nelson VL, Qin KN, Rosenfield RL, Wood JR, Penning TM,
Legro RS, et al. The biochemical basis for increased testosterone
production in theca cells propagated from patients with polycystic
ovary syndrome. J Clin Endocrinol Metab. 2001;86(12):592533.
58. CAROLE GILLING-SMITH*, D.S.W, RICHARD W. BEARD,
and A.S. Franks, Hypersecretion of androstendione by isolated
theca cells from polycystic ovaries. Clinical Endocrinology and
Metabolism, 1994.
59. Moran C, Arriaga M, Arechavaleta-Velasco F, Moran S. Adrenal
androgen excess and body mass index in polycystic ovary syn-
drome. J Clin Endocrinol Metab. 2015;100(3):94250.
60. Bellanger S, Battista MC, Fink GD, Baillargeon JP. Saturated fatty
acid exposure induces androgen overproduction in bovine adrenal
cells. Steroids. 2012;77(4):34753.
61. Rosner W. The functions of corticosteroid-binding globulin and
sex hormone-binding globulinrecent advances. Endocr Rev.
1990;11(1):8091.
62. Yang F, Ruan YC, Yang YJ, Wang K, Liang SS, Han YB, et al.
Follicular hyperandrogenism downregulates aromatase in lutein-
ized granulosa cells in polycystic ovary syndrome women.
Reproduction. 2015;150(4):28996.
63. Chen J, Shen S, Tan Y, Xia D, Xia Y, Cao Y, et al. The correlation
of aromatase activity and obesity in women with or without poly-
cystic ovary syndrome. Journal of Ovarian Research. 2015;8:11.
64. Che Q, et al. Long noncoding RNA HUPCOS promotes follicular
fluid androgen excess in PCOS patients via aromatase inhibition. J
Clin Endocrinol Metab. 2020;105(4):12.
65. Sumarac-Dumanovic M, Apostolovic M, Janjetovic K, Jeremic D,
Popadic D, Ljubic A, et al. Downregulation of autophagy gene
expression in endometria from women with polycystic ovary syn-
drome. Mol Cell Endocrinol. 2017;440:11624.
66. Lee MH, Yoon JA, Kim HR, Kim YS, Lyu SW, Lee BS, et al.
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(8):
163747.
67. Barcena C, Oliva E. WT1 Expression in the female genital tract.
Adv Anat Pathol. 2011;18(6):45465.
68. Gonzalez D, Thackeray H, Lewis PD, Mantani A, Brook N, Ahuja
K, et al. Loss of WT1 Expression in the endometrium of infertile
PCOS patients: a hyperandrogenic effect? J Clin Endocrinol
Metab. 2012;97(3):95766.
69. Taketani Y, et al. Roles of epidermal growth factor (EGF) in the
growth and differentiation of human endometrium. Hum Cell.
1989;2(3):2604.
70. Younas K, Quintela M, Thomas S, Garcia-Parra J, Blake L,
Whiteland H, et al. Delayed endometrial decidualisation in
polycystic ovary syndrome; the role of AR-MAGEA11. Journal
of Molecular Medicine-Jmm. 2019;97(9):131527.
71. Wang Q, Huang T, Shu X, Zhao SG, Liang Y, Muhammad T,
et al. Wilms' Tumor 1 overexpression in granulosa cells is associ-
ated with polycystic ovaries in polycystic ovary syndrome pa-
tients. Gynecol Obstet Investig. 2018;83(3):2416.
72. He H, Li T, Yin D, Liu R, Chen Q, Wang J, et al. HOXA10
expression is decreased by testosterone in luteinized granulosa
cells in vitro. Mol Med Rep. 2012;6(1):516.
73. Rahman TU, Ullah K, Guo MX, Pan HT, Liu J, Ren J, et al.
Androgen-induced alterations in endometrial proteins crucial in
recurrent miscarriages. Oncotarget. 2018;9(37):2462741.
74. Su Y, Shi H. High androgen level causes recurrent miscarriage and
impairs endometrial receptivity. Trop J Pharm Res. 2019;18(7):
154752.
75. Mokhtar MH, Giribabu N, Salleh N. Testosterone decreases the
number of implanting embryos, expression of pinopode and L-
selectin ligand (MECA-79) in the endometrium of early pregnant
rats. Int J Environ Res Public Health. 2020;17(7).
76. MOHD HELMY MOKHTAR1,N.G.a.N.S. Testosterone reduces
tight junction complexity and down-regulates expression of
Claudin-4 and Occludin in the endometrium in ovariectomized,
sex-steroid replacement rats. Vivo. 2020;34(1):22531.
77. Kamal DAM, Ibrahim SF, Mokhtar MH. Effects of testosterone
on the expression of Connexin 26 and Connexin 43 in the uterus of
rats during early pregnancy. In Vivo. 2020;34(4):186370.
78. Hu M, Zhang Y, Guo X, Jia W, Liu G, Zhang J, et al.
Hyperandrogenism and insulin resistance induce gravid uterine
defects in association with mitochondrial dysfunction and aber-
rant reactive oxygen species production. American Journal of
Physiology-Endocrinology and Metabolism. 2019;316(5):E794
809.
79. Ullah A, Jahan S, Razak S, Pirzada M, Ullah H, Almajwal A, et al.
Protective effects of GABA against metabolic and reproductive
disturbances in letrozole induced polycystic ovarian syndrome in
rats. Journal of Ovarian Research. 2017;10:62.
80. van Mourik MSM, Macklon NS, Heijnen CJ. Embryonic implan-
tation: cytokines, adhesion molecules, and immune cells in estab-
lishing an implantation environment. J Leukoc Biol. 2009;85(1):
419.
81. Granot I, Gnainsky Y, Dekel N. Endometrial inflammation and
effect on implantation improvement and pregnancy outcome.
Reproduction. 2012;144(6):6618.
82. Dekel N, Gnainsky Y, Granot I, Mor G. Inflammation and im-
plantation. Am J Reprod Immunol. 2010;63(1):1721.
83. Yu J, Berga SL, Zou W, Yook DG, Pan JC, Andrade AA, et al. IL-
1 beta inhibits Connexin 43 and disrupts decidualization of human
endometrial stromal cells through ERK1/2 and p38 MAP kinase.
Endocrinology. 2017;158(12):427085.
84. Piltonen TT, Chen JC, Khatun M, Kangasniemi M, Liakka A,
Spitzer T, et al. 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(5):1203
15.
85. Wang W, Ji J, Li J, Ren Q, Gu J, Zhao Y, et al. Several critical
genes and microRNAs associated with the development of poly-
cystic ovary syndrome. Annales D Endocrinologie. 2020;81(1):
1827.
86. Sheikhansari G, Soltani-Zangbar MS, Pourmoghadam Z, Kamrani
A, Azizi R, Aghebati-Maleki L, et al. Oxidative stress, inflamma-
tory settings, and microRNA regulation in the recurrent implanta-
tion failure patients with metabolic syndrome. Am J Reprod
Immunol. 2019;82(4):e13170.
Reprod. Sci.
87. Long X, Li R, Yang Y, Qiao J. Overexpression of IL-18 in the
proliferative phase endometrium of patients with polycystic ovary
syndrome. Reprod Sci. 2017;24(2):2527.
88. Samy N, Hashim M, Sayed M, Said M. Clinical significance of
inflammatory markers in polycystic ovary syndrome: their rela-
tionship to insulin resistance and Body Mass Index. Dis Markers.
2009;26(4):16370.
89. Ebejer K, Calleja-Agius J. The role of cytokines in polycystic
ovarian syndrome. Gynecol Endocrinol. 2013;29(6):53640.
90. Blumenfeld Z. The possible practical implication of high CRP
levels in PCOS. Clinical Medicine Insights-Reproductive Health.
2019;13:117955811986193.
91. Zhao DM, Shan YH, Li FH, Jiang L, Qu QL. Correlation between
endometrial receptivity with expressions of IL-1 and VEGF in rats
with polycystic ovary syndrome. Eur Rev Med Pharmacol Sci.
2019;23(13):557580.
92. Khatun M, Arffman RK, Lavogina D, Kangasniemi M, Laru J,
Ahtikoski A, et al. Women with polycystic ovary syndrome pres-
ent with altered endometrial expression of stanniocalcin-1. Biol
Reprod. 2020;102(2):30615.
93. Demi RI, et al. Fractalkine: an inflammatory chemokine elevated
in subjects with polycystic ovary syndrome. Endocrine.
2019;65(1):17583.
94. Wang J, Huang C, Jiang R, du Y, Zhou J, Jiang Y, et al. Decreased
endometrial IL-10 impairs endometrial receptivity by downregu-
lating HOXA10 expression in women with adenomyosis. Biomed
Res Int. 2018;2018:19.
95. Sarno J, Schatz F, Huang SJ, Lockwood C, Taylor HS. Thrombin
and interleukin-1 beta decrease HOX gene expression in human
first trimester decidual cells: implications for pregnancy loss. Mol
Hum Reprod. 2009;15(7):4517.
96. Kellow NJ, Savige GS. Dietary advanced glycation end-product
restriction for the attenuation of insulin resistance, oxidative stress
and endothelial dysfunction: a systematic review. Eur J Clin Nutr.
2013;67(3):23948.
97. Zhu J-L, Cai YQ, Long SL, Chen Z, Mo ZC. The role ofadvanced
glycation end products in human infertility. Life Sci. 2020;255:
117830.
98. Wang X, Liu J, Yang Y, Zhang X. An update on the potential role
of advanced glycationend products in glycolipid metabolism. Life
Sci. 2020;245:117344.
99. Liang C, Ren Y, Tan H, He Z, Jiang Q, Wu J, et al. Rosiglitazone
via upregulation of Akt/eNOS pathways attenuates dysfunction of
endothelial progenitor cells, induced by advanced glycation end
products. Br J Pharmacol. 2009;158(8):186573.
100. Pertynska-Marczewska M, Diamanti-Kandarakis E, Zhang J,
Merhi Z. Advanced glycation end products: a link between meta-
bolic and endothelial dysfunction in polycystic ovary syndrome?
Metabolism. 2015;64(11):156473.
101. Chen JH, Lin X, Bu C, Zhang X. Role of advanced glycation end
products in mobility and considerations in possible dietary and
nutritional intervention strategies. Nutrition & Metabolism.
2018;15:72.
102. Raucci A, Cugusi S, Antonelli A, Barabino SM, Monti L,
Bierhaus A, et al. A soluble form of the receptor for advanced
glycation endproducts (RAGE) is produced by proteolytic cleav-
age of the membrane-bound form by the sheddase a disintegrin
and metalloprotease 10 (ADAM10). FASEB J. 2008;22(10):
371627.
103. Wang B-J, Qian L, Li J, Wang F, Yang QL, Li G, et al. sRAGE
plays a role as a protective factor in the development of PCOS by
inhibiting inflammation. Gynecol Endocrinol. 2019;36(2):148
51.
104. Antoniotti GS, Coughlan M, Salamonsen LA, Evans J. Obesity
associated advanced glycation end products within the human
uterine cavity adversely impact endometrial function and embryo
implantation competence. Hum Reprod. 2018;33(4):65465.
105. Sun L, et al. Advanced glycation end products promote VEGF
expression and thus choroidal neovascularization via Cyr61-
PI3K/AKT signaling pathway. Sci Rep. 2017;7(1).
106. Diamanti-Kandarakis E, Dunaif A. Insulin resistance and the poly-
cystic ovary syndrome revisited: an update on mechanisms and
implications. Endocr Rev. 2012;33(6):9811030.
107. Saltiel AR, Kahn CR. Insulin signalling and the regulation of
glucose and lipid metabolism. Nature. 2001;414(6865):799806.
108. Qi J, Wang W, Zhu Q, He Y, Lu Y, Wang Y, et al. Local cortisol
elevation contributes to endometrial insulin resistance in polycys-
tic ovary syndrome. J Clin Endocrinol Metab. 2018;103(7):2457
67.
109. 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. 2013;79(1):
939.
110. Shorakae S, Ranasinha S, Abell S, Lambert G, Lambert E, de
Courten B, et al. Inter-related effects of insulin resistance,
hyperandrogenism, sympathetic dysfunction and chronic inflam-
mation in PCOS. Clin Endocrinol. 2018;89(5):62833.
111. Kumar D, Shankar K, Patel S, Gupta A, Varshney S, Gupta S,
et al. Chronic hyperinsulinemia promotes meta-inflammation and
extracellular matrix deposition in adipose tissue: implications of
nitric oxide. Mol Cell Endocrinol. 2018;477:1528.
112. Orostica L, et al. Effect of TNF-alpha on molecules related to the
insulin action in endometrial cells exposed to hyperandrogenic
and hyperinsulinic conditions characteristics of polycystic ovary
syndrome. Reprod Sci. 2018;25(7):10009.
113. Gonzalez F. Inflammation in polycystic ovary syndrome: under-
pinning of insulin resistance and ovarian dysfunction. Steroids.
2012;77(4):3005.
114. Orostica L, et al. Pro-inflammatory markers negatively regulate
IRS1 in endometrial cells and endometrium from women with
obesity and PCOS. Reprod Sci. 2020;27(1):290300.
115. Shimizu, I., Yoshida Y., Katsuno T., Tateno K., Okada S., Moriya
J., Yokoyama M., Nojima A., Ito T., Zechner R., Komuro I.,
Kobayashi Y., Minamino T., p53-induced adipose tissue inflam-
mation is critically involved in the development of insulin resis-
tance in heart failure (vol 15, pg 51, 2012). Cell Metab, 2012.
15(5). 787-787.
116. Diamanti-Kandarakis E, Chatzigeorgiou A, Papageorgiou E,
Koundouras D, Koutsilieris M. Advanced glycation end-
products and insulin signaling in granulosa cells. Exp Biol Med
(Maywood). 2016;241(13):143845.
117. Shaaban Z, Khoradmehr A, Amiri-Yekta A, Jafarzadeh Shirazi
MR, Tamadon A. Pathophysiologic mechanisms of obesity- and
chronic inflammation-related genes in etiology of polycystic ova-
ry syndrome. Iranian Journal of Basic Medical Sciences.
2019;22(12):137886.
118. Blagojevic IP, et al. Evaluation of a summary score for dyslipid-
emia, oxidative stress and inflammation (the DOI score) in women
with polycystic ovary syndrome and its relationship with obesity.
Journal of Medical Biochemistry. 2018;37(4):47685.
119. Bellver J, Martínez-Conejero JA, Labarta E, Alamá P, Melo
MAB, Remohí J, et al. Endometrial gene expression in the win-
dow of implantation is altered in obese women especially in asso-
ciation with polycystic ovary syndrome. Fertil Steril. 2011;95(7):
2335U254.
120. Comstock IA, et al. Does an increased body mass index affect
endometrial gene expression patterns in infertile patients? A func-
tional genomics analysis Fertility and Sterility. 2017;107(3):740.
121. Riley JK, Jungheim ES. Is there a role for diet in ameliorating the
reproductive sequelae associated with chronic low-grade
Reprod. Sci.
inflammation in polycystic ovary syndrome and obesity? Fertil
Steril. 2016;106(3):5207.
122. Beatriz Motta A. The role of obesity in the development of poly-
cystic ovary syndrome. Curr Pharm Des. 2012;18(17):248291.
123. Diamanti-Kandarakis E, Lambrinoudaki I, Economou F, Christou
M, Piperi C, Papavassiliou AG, et al. Androgens associated with
advanced glycation end-products in postmenopausal women.
Menopause. 2010;17(6):11827.
124. Liao Y, Huang R, Sun Y, Yue J, Zheng J, Wang L, et al. An
inverse association between serum soluble receptor of advanced
glycation end products and hyperandrogenism and potential im-
plication in polycystic ovary syndrome patients. Reprod Biol
Endocrinol. 2017;15(1):9.
125. Rosenfield RL, Ehrmann DA. The pathogenesis of polycystic
ovary syndrome (PCOS): the hypothesis of PCOS as functional
ovarian hyperandrogenism revisited. Endocr Rev. 2016;37(5):
467520.
126. Glueck CJ, Goldenberg N. Characteristics of obesity in polycystic
ovary syndrome: etiology, treatment, and genetics. Metabolism-
Clinical and Experimental. 2019;92:10820.
127. Ujvari D, Hulchiy M, Calaby A, Nybacka A, Bystrom B,
Hirschberg AL. Lifestyle intervention up-regulates gene and pro-
tein levels of molecules involved in insulin signaling in the endo-
metrium of overweight/obese women with polycystic ovary syn-
drome. Hum Reprod. 2014;29(7):152635.
128. Dantas WS, Neves W, Gil S, Barcellos CRG, Rocha MP, de Sá-
Pinto AL, et al. Exercise-induced anti-inflammatory effects in
overweight/obese women with polycystic ovary syndrome.
Cytokine. 2019;120:6670.
129. Ito-Yamaguchi A, Suganuma R, Kumagami A, Hashimoto S,
Yoshida-Komiya H, Fujimori K. Effects of metformin on endo-
crine, metabolic milieus and endometrial expression of androgen
receptor in patients with polycystic ovary syndrome. Gynecol
Endocrinol. 2015;31(1):447.
130. Zhai J, Yao GD, Wang JY, Yang QL, Wu L, Chang ZY, et al.
Metformin regulates key microRNAs to improve endometrial re-
ceptivity through increasing implantation marker gene expression
in patients with PCOS undergoing IVF/ICSI. Reprod Sci.
2019;26(11):143948.
131. Dragamestianos C, Messini CI, Antonakis PT, Zacharouli K,
Kostopoulou E, Makrigiannakis A, et al. The effect of metformin
on the endometrium of women with polycystic ovary syndrome.
Gynecol Obstet Investig. 2019;84(1):3544.
132. Freis A, Renke T, Kämmerer U, Jauckus J, Strowitzki T,
Germeyer A. Effects of a hyperandrogenaemic state on the pro-
liferation and decidualization potential in human endometrial stro-
mal cells. Arch Gynecol Obstet. 2017;295(4):100513.
133. Gong H, et al. Flutamide ameliorates uterine decidualization and
angiogenesis in the mouse hyperandrogenemia modelduring mid-
pregnancy. PLoS ONE [Electronic Resource]. 2019;14(5):
e0217095.
134. Salamonsen LA. My WOMBan's life: understanding human en-
dometrial function. Reproduction. 2019;158(6):F5567.
135. Cabrera-Cruz H, Oróstica L, Plaza-Parrochia F, Torres-Pinto I,
Romero C, Vega M. The insulin-sensitizing mechanism of myo-
inositol is associated with AMPK activation and GLUT-4 expres-
sion in human endometrial cells exposed to a PCOS environment.
Am J Physiol Endocrinol Metab. 2020;318(2):E23748.
136. Vlassara H, Cai W, Tripp E, Pyzik R, Yee K, Goldberg L, et al.
Oral AGE restriction ameliorates insulin resistance in obese indi-
viduals with the metabolic syndrome: a randomised controlled
trial. Diabetologia. 2016;59(10):218192.
137. Ashary N, Tiwari A, Modi D. Embryo implantation: war in times
of love. Endocrinology. 2018;159(2):118898.
138. Wilcox AJ, Weinberg CR, O'ConnorJF, Baird DD, Schlatterer JP,
Canfield RE, et al. Incidence of early loss of pregnancy. N Engl J
Med. 1988;319(4):18994.
139. Dunaif A, Segal KR, Futterweit W, Dobrjansky A. Profound pe-
ripheral insulin resistance, independent of obesity, in polycystic
ovary syndrome. Diabetes. 1989;38(9):116574.
140. Shaaban Z, Khoradmehr A, Jafarzadeh Shirazi MR, Tamadon A.
Pathophysiological mechanisms of gonadotropins- and steroid
hormones-related genes in etiology of polycystic ovary syndrome.
Iranian Journal of Basic Medical Sciences. 2019;22(1):316.
141. Bannigida DM, Nayak BS, Vijayaraghavan R. Insulin resistance
and oxidative marker in women with PCOS. Arch Physiol
Biochem. 2020;126(2):1836.
142. Borzan V, et al. Risk of insulin resistance and metabolic syndrome
in women with hyperandrogenemia: a comparison between PCOS
phenotypes and beyond. J Clin Med. 2021;10(4).
143. Orostica L, et al. Proinflammatory environment and role of TNF-
alpha in endometrial function of obese women having polycystic
ovarian syndrome. Int J Obes. 2016;40(11):171522.
PublishersNote Springer Nature remains neutral with regard to jurisdic-
tional claims in published maps and institutional affiliations.
Reprod. Sci.
... This period is called the implantation window, which is a short-time period during the reproductive cycle (Haouzi et al., 2021). Disturbance in endometrial receptivity may lead to problems in placental development as well as implantation failures and the capacity of the endometrium to support fetal growth (Fowden et al., 2008;Robertson et al., 2011;Jiang and Li, 2021). ...
... The morphological aspects involved evaluation of the uterine epithelium thinning, replacement of microvilli projections by pinopodes on its apical surface and endometrial glands transformation. All these markers are involved and influence and in pinopodes formation, embryo development and its attachment to the uterine epithelium and maternal immune response during pregnancy (Achache and Revel, 2006;Jiang and Li, 2021). ...
... Furthermore, PCOS is highly associated with metabolic syndrome and insulin resistance [4] . Therefore, effective management of metabolic complications associated with PCOS is paramount, emphasizing timely detection and preventive measures [5,6] . A recommended approach for screening dyslipidemia in women with PCOS is an oral glucose tolerance test, irrespective of a family history of diabetes mellitus (DM) [7] . ...
Article
Full-text available
Polycystic ovary syndrome (PCOS) is a common endocrine disorder characterized by a hormonal imbalance that affects females of reproductive age. The association between advanced glycation end products (AGEs) and PCOS has attracted considerable attention in recent years, highlighting the potential of AGEs as biomarkers for this disorder. In the present systematic review and meta-analysis, we aimed to examine the association between AGEs and PCOS, evaluate their potential as biomarkers, and improve our understanding of the pathophysiology of PCOS and its associated metabolic complications. A literature search was performed using various databases from January 2000 to March 2023 to identify relevant studies investigating the association between AGEs and PCOS. Pooled effect estimates were calculated using standardized mean differences (SMD) with 95% confidence intervals (CIs). Subgroup and meta-regression analyses were performed to examine potential sources of heterogeneity. The meta-analysis included six studies with a total of 623 participants. Our results revealed a significant increase in circulating AGE levels in females with PCOS compared to healthy females (SMD = 2.35; 95% CI: 1.10-3.60; P <0.001). Significant heterogeneity was observed between the studies (I 2 = 96.37%; P <0.001), indicating the presence of several factors influencing the association. Subgroup analyses based on body mass index, age, and homeostatic model assessment for insulin resistance indicated differential effects of AGEs on specific subgroups. This systematic review and meta-analysis support the association between elevated AGE levels and PCOS, thereby suggesting the potential role of AGEs as biomarkers in PCOS.
... Furthermore, PCOS is highly associated with metabolic syndrome and insulin resistance [4] . Therefore, effective management of metabolic complications associated with PCOS is paramount, emphasizing timely detection and preventive measures [5,6] . A recommended approach for screening dyslipidemia in women with PCOS is an oral glucose tolerance test, irrespective of a family history of diabetes mellitus (DM) [7] . ...
Article
Full-text available
Polycystic ovary syndrome (PCOS) is a common endocrine disorder characterized by a hormonal imbalance that affects females of reproductive age. The association between advanced glycation end products (AGEs) and PCOS has attracted considerable attention in recent years, highlighting the potential of AGEs as biomarkers for this disorder. In the present systematic review and meta-analysis, we aimed to examine the association between AGEs and PCOS, evaluate their potential as biomarkers, and improve our understanding of the pathophysiology of PCOS and its associated metabolic complications. A literature search was performed using various databases from January 2000 to March 2023 to identify relevant studies investigating the association between AGEs and PCOS. Pooled effect estimates were calculated using standardized mean differences (SMD) with 95% confidence intervals (CIs). Subgroup and meta-regression analyses were performed to examine potential sources of heterogeneity. The meta-analysis included six studies with a total of 623 participants. Our results revealed a significant increase in circulating AGE levels in females with PCOS compared to healthy females (SMD = 2.35; 95% CI: 1.10-3.60; P <0.001). Significant heterogeneity was observed between the studies (I 2 = 96.37%; P <0.001), indicating the presence of several factors influencing the association. Subgroup analyses based on body mass index, age, and homeostatic model assessment for insulin resistance indicated differential effects of AGEs on specific subgroups. This systematic review and meta-analysis support the association between elevated AGE levels and PCOS, thereby suggesting the potential role of AGEs as biomarkers in PCOS.
... It is affecting women of reproductive age, around the age group between 18 and 44. PCOS is also linked with irregularity in menstrual cycles, hyperandrogenism, alopecia, hirsutism, obesity, insulin (INS) resistance, anovulation, oligomenorrhea, and pre-diabetes [3,4]. It is characterized by elevated androgen levels (hyperandrogenemia) leading to anovulation, microcysts in ovaries (polycystic ovaries) and can cause inhibition of follicular development. ...
Article
Full-text available
Objectives: In this study, phytocompounds of spearmint that is known to have anti-androgenic activity are docked against a protein CYP21A2. This protein is also known as progesterone complex, one of the member cytochrome P450 enzymes; mutations in the genes encoding these proteins are causative factors of polycystic ovarian syndrome (PCOS). Methods: The study was based on computations using different phytochemicals of spearmint docking to a target protein CYP21A2 which causes hormonal imbalance leading to PCOS and hirsutism. Molecular docking was conducted using PyRx-virtual screening tool and Biovia discovery studio 2.0 to determine binding affinities of different phytochemicals to target protein. Results: The docking result revealed that bicyclogermacrene, cubebol, (-)-beta-bourbonene, alpha-bourbonene, and spathulenol showed highest binding affinities between –8.1 and –8.5 kcal/mol. Further, absorption, distribution, metabolism, excretion, and toxicity properties of these compounds are explored mainly to understand the possibility of developing potential drugs for PCOS. Conclusion: These bioactive compounds can be considered as potential agents that can be used with polyherbal plant extract to reduce the androgen levels in women suffering from PCOS.
Article
In this study, we utilized magnetic nanobeads for the extraction of nucleic acids from tissues to investigate the expression levels and correlation between lncRNA H19, miR-612, and their target gene HOXA10 in peri-implantation endometrium of mice. Furthermore, we conducted overexpression or gene knockout experiments on lncRNA H19 to observe its impact on the expression of miR-612 and HOXA10. The targeted binding relationship between lncRNA H19, miR-612, and HOXA10 was detected by dual luciferase reporter assay. The regulatory relationship between lncRNA H19, miR-612, and HOXA10 was verified through silencing or overexpression of these genes. Intrauterine transfection was used to modulate the expression of lncRNA H19 in endometria during pregnancy, followed by the detection of the expression levels of miR-612 and HOXA10 as well as ITGB3 and IGFBP-1 proteins. Compared with non-pregnant mice, we observed a significant upregulation of both lncRNA H19 and HOXA10 in the endometria of pregnant mice, while miR-612 was found to be downregulated ( P < 0.05). Further analysis revealed that the expression levels of lncRNA H19 and HOXA10 increased progressively with gestational days, peaking on Day 4 ( P < 0.05). Moreover, Through database analysis, we identified binding sites for lncRNA H19-miR-612 as well as HOXA10-miR-612 interactions. The dual-luciferase reporter assay further supported our conjecture that lncRNA H19 could specifically bind the miR-612, which in turn targets HOXA10 to regulate its expression ( P < 0.05). In conclusion, regulations of lncRNA H19 and HOXA10 expression contribute to enhancing endometrial receptivity and facilitating decidualization of endometrial stromal cells, ultimately promoting successful embryo implantation.
Article
Objective To investigate the effect of GnRH agonist (GnRH-a) down-regulation prior to hormone replacement treatment (HRT) to prepare the endometrium in frozen embryo transfer (FET) cycles in women of different ages. Methods This was a retrospective study, and after excluding patients with adenomyosis, endometriosis, severe endometrial adhesions, polycystic ovary syndrome (PCOS), and repeated embryo implantation failures, a total of 4,091 HRT cycles were collected. Patients were divided into group A (<35 years old) and group B (≥35 years old), and each group was further divided into HRT and GnRHa-HRT groups. The clinical outcomes were compared between groups. Results There was no statistically significant difference in clinical outcomes between the HRT and GnRHa-HRT groups among women aged <35 years. In women of advanced age, higher rates of clinical pregnancy and live birth were seen in the GnRHa-HRT group. Logistic regression analysis showed that female age and number of embryos transferred influenced the live birth rate in FET cycles, and in women aged ≥ 35 years, the use of GnRH-a down-regulation prior to HRT improved pregnancy outcomes. Conclusions In elderly woman without adenomyosis, endometriosis, PCOS, severe uterine adhesions, and RIF, hormone replacement treatment with GnRH agonist for pituitary suppression can improve the live birth rate of FET cycles.
Article
Background Endometrial CD138+ plasma cells serve as a diagnostic biomarker for endometrial inflammation, and their elevated occurrence correlates positively with adverse pregnancy outcomes. Infertility-related conditions like polycystic ovary syndrome (PCOS) and recurrent implantation failure (RIF) are closely associated with systemic and local chronic inflammatory status, wherein endometrial CD138+ plasma cell accumulation could also contribute to endometrial pathology. Current methods for quantifying CD138+ cells typically involve laborious and time-consuming microscopic assessments of only a few random areas from a slide. These methods have limitations in accurately representing the entire slide and are susceptible to significant biases arising from intra- and interobserver variations. Implementing artificial intelligence (AI) for CD138+ cell identification could enhance the accuracy, reproducibility, and reliability of analysis. Methods Here, an AI algorithm was developed to identify CD138+ plasma cells within endometrial tissue. The AI model comprised two layers of convolutional neural networks (CNNs). CNN1 was trained to segment epithelium and stroma across 28,363 mm² (2.56 mm² of epithelium and 24.87 mm² of stroma), while CNN2 was trained to distinguish stromal cells based on CD138 staining, encompassing 7345 cells in the object layers (6942 CD138− cells and 403 CD138+ cells). The training and performance of the AI model were validated by three experienced pathologists. We collected 193 endometrial tissues from healthy controls (n = 73), women with PCOS (n = 91), and RIF patients (n = 29) and compared the CD138+ cell percentages based on cycle phases, ovulation status, and endometrial receptivity utilizing the AI model. Results The AI algorithm consistently and reliably distinguished CD138− and CD138+ cells, with total error rates of 6.32% and 3.23%, respectively. During the training validation, there was a complete agreement between the decisions made by the pathologists and the AI algorithm, while the performance validation demonstrated excellent accuracy between the AI and human evaluation methods (intraclass correlation; 0.76, 95% confidence intervals; 0.36–0.93, p = 0.002) and a positive correlation (Spearman's rank correlation coefficient: 0.79, p < 0.01). In the AI analysis, the AI model revealed higher CD138+ cell percentages in the proliferative phase (PE) endometrium compared to the secretory phase or anovulatory PCOS endometrium, irrespective of PCOS diagnosis. Interestingly, CD138+ percentages differed according to PCOS phenotype in the PE (p = 0.03). On the other hand, the receptivity status had no impact on the cell percentages in RIF samples. Conclusion Our findings emphasize the potential and accuracy of the AI algorithm in detecting endometrial CD138+ plasma cells, offering distinct advantages over manual inspection, such as rapid analysis of whole slide images, reduction of intra- and interobserver variations, sparing the valuable time of trained specialists, and consistent productivity. This supports the application of AI technology to help clinical decision-making, for example, in understanding endometrial cycle phase-related dynamics, as well as different reproductive disorders.
Article
Background: This study investigated the therapeutic effects of Rosa canina L. seed oil (RCSO) on PCOS using an animal model, focusing on evaluating biochemical factors and the Bax/Bcl-2/p53 and Caspase-3 (Cas-3) pathways. Materials and Methods: The PCOS model was created by injecting estradiol valerate into 60 adult Wistar rats, which were assigned to six groups of 10 animals: normal, PCOS, metformin, RCSO, and co-treatment with MET and RCSO groups. After 50 days, the serum levels of testosterone, progesterone, LH, and FSH hormones were measured, along with the activity of catalase, superoxide dismutase, glutathione peroxidase, and nitric oxide. Additionally, ovarian tissues were taken for gene expression analysis of Bax/Bcl-2/p53 and Cas-3 using real-time PCR, as well as protein expression analysis with western blot. Immunohistochemistry was performed to assess the levels of p53 and Ki-67 proteins, indicating the apoptosis of ovarian granulosa cells. Results: RCSO, in synergy with MET, demonstrated reductions in the LH/FSH ratio along with a decrease in progesterone and increased activity of antioxidant enzymes. Additionally, RCSO, along with MET, upregulated the Bax/Bcl-2/p53 and Cas-3 pathways, resulting in increased p53 expression and decreased Ki-67-positive cells, indicative of enhanced apoptosis. Conclusions: These findings suggest the potential use of RCSO in women with ovulation disorders, particularly those with PCOS.
Article
Full-text available
Polycystic ovary syndrome (PCOS) is the most common endocrine disorder in premenopausal women, with a wide spectrum of possible phenotypes, symptoms and sequelae according to the current clinical definition. However, there are women who do not fulfill at least two out of the three commonly used “Rotterdam criteria” and their risk of developing type 2 diabetes or obesity later in life is not defined. Therefore, we addressed this important gap by conducting a retrospective analysis based on 750 women with and without PCOS. We compared four different PCOS phenotypes according to the Rotterdam criteria with women who exhibit only one Rotterdam criterion and with healthy controls. Hormone and metabolic differences were assessed by analysis of variance (ANOVA) as well as logistic regression analysis. We found that hyperandrogenic women have per se a higher risk of developing insulin resistance compared to phenotypes without hyperandrogenism and healthy controls. In addition, hyperandrogenemia is associated with developing insulin resistance also in women with no other Rotterdam criterion. Our study encourages further diagnostic and therapeutic approaches for PCOS phenotypes in order to account for varying risks of developing metabolic diseases. Finally, women with hyperandrogenism as the only symptom should also be screened for insulin resistance to avoid later metabolic risks.
Article
Full-text available
Polycystic ovary syndrome (PCOS) is a complex disease that causes an ovulatory infertility in approximately 10% of reproductive-age women. We searched for candidate proteins that might contribute to endometrial receptivity defects in PCOS patients, and result in adverse reproductive outcomes. Shotgun proteomics approach was used to investigate the proteome profile of the endometrium at the luteal phase in PCOS patients compared to healthy fertile individuals. Biological process and pathway analyses were conducted to categorize the proteins with differential expressions. Confirmation was performed for a number of proteins via immunoblotting in new samples. 150 proteins with higher abundance, and 46 proteins with lower abundance were identified in the endometrial tissue from PCOS patients compared to healthy fertile individuals. The proteins with higher abundance were enriched in protein degradation, cell cycle, and signaling cascades. Proteins with lower abundance in PCOS patients were enriched in extracellular matrix (ECM) composition and function, as well as the salvage pathway of purine biosynthesis. Metabolism was the most affected biological process with over 100 up-regulated, and approximately 30 down-regulated proteins. Our results indicate significant imbalances in metabolism, proteasome, cell cycle, ECM related proteins, and signaling cascades in endometrial tissue of PCOS, which may contribute to poor reproductive outcomes in these patients. We postulate that the endometria in PCOS patients may not be well-differentiated and synchronized for implantation. Possible roles of the above-mentioned pathways that underlie implantation failure in PCOS will be discussed. Our findings need to be confirmed in larger populations.
Article
Full-text available
Placentation is a major determinant of the success of pregnancy. It is regulated by several factors such as cell adhesion molecules, tight junctions, and gap junctions. The cell adhesion molecules are integrins, cadherins, immunoglobulins, nectins, and selectins. The tight junctions are composed of claudins, occludin, and junction adhesion molecule proteins while the gap junctions are composed of connexins of varying molecular weights. During placentation, some of these molecules regulate trophoblast proliferation, trophoblast fusion, trophoblast migration, trophoblast invasion, trophoblast-endothelium adhesion, glandular remodeling, and spiral artery remodeling. There is a dysregulated placental expression of some of these molecules during obstetric complications. We have, hereby, indicated the expression patterns of the subunits of each of these molecules in the various trophoblast subtypes and in the decidua, and have highlighted their involvement in physiological and pathological placentation. The available evidence points to the relevance of these molecules as distinguishing markers of the various trophoblast lineages and as potential therapeutic targets in the management of malplacentation-mediated diseases.
Article
Full-text available
Endometrial receptivity is mediated by adhesion molecules at the endometrium-trophoblast interface where osteopontin (OPN) and CD44 form a protein complex that plays an important role in embryo recognition. Here, we undertook a prospective study investigating the expression and regulation of OPN and CD44 in 50 fertile and 31 infertile ovulatory polycystic ovarian syndrome (PCOS) patients in the proliferative and secretory phases of the natural menstrual cycle and in 12 infertile anovulatory PCOS patients. Endometrial biopsies and blood samples were evaluated for expression of OPN and CD44 using RT-PCR, immunohistochemistry and ELISA analysis to determine circulating levels of OPN, CD44, TNF-α, IFN-γ and OPN and CD44 levels in biopsy media. Our findings highlighted an increased level of circulating OPN and CD44 in serum from infertile patients that inversely correlated with expression levels in endometrial tissue and positively correlated with levels secreted into biopsy media. OPN and CD44 levels positively correlated to each other in serum and media from fertile and PCOS patients, as well as to circulating TNF-α and IFN-γ. In vitro analysis revealed that hormone treatment induced recruitment of ERα to the OPN and CD44 promoters with a concomitant increase in the expression of these genes. In infertile patients, inflammatory cytokines led to recruitment of NF-κB and STAT1 proteins to the OPN and CD44 promoters, resulting in their overexpression. These observations suggest that the endometrial epithelial OPN-CD44 adhesion complex is deficient in ovulatory PCOS patients and displays an altered stoichiometry in anovulatory patients, which in both cases may perturb apposition. This, together with elevated circulating and local secreted levels of these proteins, may hinder endometrium-trophoblast interactions by saturating OPN and CD44 receptors on the surface of the blastocyst, thereby contributing to the infertility associated with ovulating PCOS patients. Key messages • Endometrial epithelial OPN-CD44 adhesion complex levels are deficient in ovulatory PCOS patients contributing to the endometrial infertility associated with ovulating PCOS patients. • Circulating levels of OPN, CD44 and inflammatory cytokines TNF-α and IFN-γ are altered in infertile PCOS patients. • Increased levels of both OPN and CD44 in biopsy media and serum inversely correlate with endometrial expression of these markers in endometrial tissue. • In infertile PCOS patients, high levels of oestrogens and inflammatory cytokines stimulate the recruitment of transcription factors to the OPN and CD44 promoters to enhance gene transcription. • Our study identifies a novel crosstalk between the CD44-OPN adhesion complex, ERα, STAT1 and NF-κB pathways modulating endometrial receptivity.
Article
Full-text available
Purpose: To evaluate the clinical usefulness of the endometrial receptivity array (ERA) and the preimplantation genetic test for aneuploidy (PGT-A) in patients with severe and moderate recurrent implantation failure (RIF). Design: A retrospective multicenter cohort study was conducted in patients who failed to achieve implantation following transfer of 3 or more or 5 or more embryos in at least three single embryo transfers; patients were classified as moderate or severe RIF, respectively. Patients with previous RIF were compared based on the testing they received: PGT-A, ERA, or PGT-A+ERA versus a control group with no testing. Mean implantation rate and ongoing pregnancy rates per embryo transfer were considered primary outcomes. Multiple logistic regression analysis was performed and adjusted ORs were calculated to control possible bias. Results: Of the 2110 patients belonging to the moderate RIF group, those who underwent transfer of euploid embryos after PGT-A had a higher implantation rate than those who did not. Additionally, the PGT-A group had a significantly higher rate of ongoing pregnancy. The same outcomes measured for the 488 patients in the severe RIF group did not reveal any statistically significant improvements. The use of the ERA test did not appear to significantly improve outcomes in either group. Conclusions: PGT-A may be beneficial for patients with moderate recurrent implantation failure but not for severe cases. At its current level of development, ERA does not appear to be clinically useful for patients with RIF.
Article
Full-text available
Background: Repeated implantation failure (RIF) is currently believed by some scholars to be mainly related to the poor endometrial receptivity. Three-dimensional (3D) ultrasound, as a noninvasive examination, has attracted the most attention. This paper further discusses whether 3D ultrasound of the endometrial receptivity index has guidance value in the evaluation of pregnancy outcomes in patients with RIF following frozen-thawed embryo transfer. Methods: A total of 79 patients with RIF were retrospectively analyzed. These 79 patients which were confirmed by the transvaginal ultrasonography detection comprised 36 cases of intrauterine pregnancy, which were included in the pregnancy group, and 43 cases with biochemical pregnancy (the HCG in blood or urine is only transient and can be detected by clinical biochemical methods. The pregnancy sac cannot be seen under ultrasound) or negative results of human chorionic gonadotropin (HCG) examination, which were included in the non-pregnancy group. The endometrial thickness, uterine volume, sub-endometrial blood flow type, pulsatility index (PI) and resistance index (RI) of the spiral artery; the RI and PI of the uterine artery; and the peak systolic velocity/end diastolic velocity (S/D) of the two groups were measured and analyzed. Results: (I) There were no significant differences in the age, infertility years, body mass index (BMI), anti-Müllerian hormone (AMH), endometrial thickness on the hCG injection day, estradiol (E2), and progesterone (P) between the pregnant and non-pregnant groups (P>0.05). (II) There were no significant differences in the endometrial thickness, uterine volume, and RI and PI of the uterine artery on the day before the implantation between the two groups (P>0.05). (III) There was statistical difference in the sum of the S/D of the two uterine arteries between the two groups (P<0.05) with a cutoff value of 14.47 (P<0.05). (IV) The RI and PI of the spiral artery in the non-pregnancy group were lower than those in the pregnancy group, and the difference was statistically significant (P<0.05). (V) The differences in the endometrial classification and subendometrial blood flow classification between the two groups were statistically significant (P<0.05). Conclusions: Ultrasonographic endometrial blood flow classification, spiral artery blood flow parameters, and uterine artery blood flow parameters can be effective indexes for evaluating endometrial receptivity, and they have a certain clinical significance in evaluating the pregnancy outcome of RIF patients after retransplantation.
Article
Full-text available
PurposeTo assess the effects PCOS on live birth rates when transferring a single fresh ideal blastocyst. MethodsA retrospective cohort study performed at the university-affiliated reproductive center. Women with PCOS and a control group of normal ovulatory women who underwent their first fresh embryo transfer with single ideal grade blastocyst were included in the study. Demographic, stimulation information and pregnancy outcomes were collected and analysed. The primary outcome was live birth rates, and secondary outcomes included pregnancy and clinical pregnancy rates.Results71 Women with PCOS and 272 normal ovulatory controls underwent their first embryo transfer and met the inclusion and exclusion criteria. PCOS patient were younger (31.0 ± 3.7 vs. 33.1 ± 3.2, p = 0.0001), with higher AFC (40.0 ± 9.3 vs. 13.3 ± 4.6, p = 0.0001), required lower dose of gonadotropins to stimulate (1198 ± 786 vs. 1891 ± 1224, p = 0.0001), and had higher serum testosterone levels (2.3 ± 0.7 vs. 1.1 ± 0.3, p = 0.0001). No significant difference was found between the two groups regarding the number of previous pregnancies, the number of previous full-term pregnancies, the level of basal serum FSH, estradiol level at triggering and the BMI. When compared by Chi squared testing pregnancy rates, clinical pregnancy rates and live birth rates did not differ. However, when controlling (with multivariate stepwise logistic regression) for confounders, live birth rates were lower among the women with PCOS (p = 0.035, CI: 0.18−0.92).Conclusion After controlling for confounders, when transferring a fresh single ideal blastocyst, live birth rates were lower among the women with PCOS than normal ovulatory controls.
Article
Placentation is a major determinant of the success of pregnancy. It is regulated by several factors such as cell adhesion molecules, tight junctions, and gap junctions. The cell adhesion molecules are integrins, cadherins, immunoglobulins, nectins, and selectins. The tight junctions are composed of claudins, occludin, and junction adhesion molecule proteins while the gap junctions are composed of connexins of varying molecular weights. During placentation, some of these molecules regulate trophoblast proliferation, trophoblast fusion, trophoblast migration, trophoblast invasion, trophoblast-endothelium adhesion, glandular remodeling, and spiral artery remodeling. There is a dysregulated placental expression of some of these molecules during obstetric complications. We have, hereby, indicated the expression patterns of the subunits of each of these molecules in the various trophoblast subtypes and in the decidua, and have highlighted their involvement in physiological and pathological placentation. The available evidence points to the relevance of these molecules as distinguishing markers of the various trophoblast lineages and as potential therapeutic targets in the management of malplacentation-mediated diseases. Keywords Trophoblast. Integrin. Cadherin. Immunoglobulin. Nectin. Selectin. Claudin. Occludin. Connexin. Preeclampsia Abbreviations PECAM1 platelet-endothelial cell adhesion molecule-1 VCAM1 vascular cell adhesion molecule-1 EMILIN1 elastin microfibril interface-located protein-1 ECM extracellular matrix Dsg2 desmoglein-2 Dsg3 desmoglein-3 PECAM1 platelet-endothelial cell adhesion molecule-1 NCAM neural cell adhesion molecule Mel-CAM melanoma cell adhesion molecule ICAM1 intercellular adhesion molecule-1 VCAM1 vascular cell adhesion molecule-1 ZO zonnula occludens CTB cytotrophoblast STB syncytiotrophoblast EVTs extravillous trophoblasts PCT proximal column trophoblast DCT distal column trophoblast iEVT intestitial extravillous trophoblast egEVT endoglandular extravillous trophoblast evEVT endovascular extravillous trophoblast GC giant cell dSC decidual stromal cell dNK decidual natural killer cell
Article
Purpose: To investigate the effect of androgen (A2) at different concentrations on human endometrial epithelial cancer cell line (HHUA) using iTRAQ and liquid chromatography–mass spectrometry (LCMS/MS) analysis. Methods: Human endometrial cells were cultured in Dulbecco Modified Eagle Medium. Total protein was isolated from human endometrial cells and HHUA cells were treated with A2 in three different concentrations (10-9 , 10-8 and 10-7 M). The proteins were digested using filter-aided sample preparation (FASP) method, and labeled using iTRAQ. The proteins were then subjected to LC-MS/MS. The resultant peptides were identified using Expasy tool, and the up-stream and down-stream proteins were confirmed. Moreover, the up-regulated proteins: isoform 2 of nuclear ubiquitous casein and cyclindependent kinase substrate 1 (NUCKS1), isoform 2 of endothelial differentiation-related factor 1 (EDF1), and acid phosphatase 1 (ACP1) were independently confirmed using western blotting and immunohistochemistry techniques. Results: Transforming growth factor (TGF), p38 mitogen-activated protein kinase (p38-MAPK), cyclindependent kinase (CDK), and tumor protein p53 (TP53) were the major regulators of the upstream process, and were upregulated RM-linked endometrial epithelial cells. Androgen-associated proteins NUCKS1, EDF1, and ACP1 were significantly responsible for miscarriage-related changes in endometrial tissues (p < 0.05). The results showed that a high level of androgen significantly (p < 0.05) distorts the expression levels of proteins associated with endometrium development, resulting in impaired endometrial accessibility which led to miscarriage. Conclusion: Androgen-associated proteins are responsible for changes in endometrial tissues which, in turn, lead to recurrent miscarriage.
Article
For patients with recurrent implantation failure in IVF, histologic or transcriptomic testing of the endometrium during the mid-secretory phase is often considered. Histological dating of endometrial biopsies (Noyes criteria) can determine if endometrial morphology is consistent with the period of receptivity. Alternatively, endometrial tissue can be sent for a commercial Endometrial Receptivity Array (ERA) test which characterizes the gene expression of the endometrium using a panel of 238 genes that have been implicated in endometrial receptivity. This study aimed to compare the two tests to assess their concordance and to examine the ability of the ERA to successfully predict implantation and pregnancy in a subsequent personalized embryo transfer. A retrospective review was done of 97 patients with a history of implantation failure who underwent an ERA, 35 of whom had histologic dating on the same sample. ERA and histology were classified as 'concordant' when samples were receptive by both tests or non-receptive by both tests. The ERA result was then used to personalize the embryo transfer day, and pregnancy rates from the first subsequent frozen transfer cycle were analyzed. The results indicated that there is poor concordance between ERA and histological dating with only 40.0% agreement and a kappa (95%CI) = -0.18 (-0.50, 0.14). According to the ERA, 48.5% of biopsies were receptive, 47.4% were non-receptive and 2.01% were insufficient tissue for analysis. The clinical pregnancy rate in patients shown to be receptive by ERA was 26.7% and non-receptive was 22.5% following the subsequent personalized ET (p = 0.66). This study concludes that there is a high degree of discordance between histological dating of the endometrium and molecular analysis by ERA. There was no evidence of clinical benefit when embryo transfer was personalized according to ERA in patients with a history of implantation failure.