Next Article in Journal
Spheroid Formation and Peritoneal Metastasis in Ovarian Cancer: The Role of Stromal and Immune Components
Next Article in Special Issue
Valproate Targets Mammalian Gastrulation Impairing Neural Tissue Differentiation and Development of the Placental Source In Vitro
Previous Article in Journal
Copper Modulates Mitochondrial Oxidative Phosphorylation to Enhance Dermal Papilla Cells Proliferation in Rex Rabbits
Previous Article in Special Issue
Melatonin Signaling Pathways Implicated in Metabolic Processes in Human Granulosa Cells (KGN)
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

MicroRNAs in the Regulation of Endometrial Receptivity for Embryo Implantation

Implantation and Pregnancy Research Laboratory, School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology, Bundoora, VIC 3083, Australia
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(11), 6210; https://doi.org/10.3390/ijms23116210
Submission received: 8 April 2022 / Revised: 13 May 2022 / Accepted: 28 May 2022 / Published: 1 June 2022
(This article belongs to the Special Issue Germ Cells Molecular Research and Application)

Abstract

:
Development of endometrial receptivity is crucial for successful embryo implantation and pregnancy initiation. Understanding the molecular regulation underpinning endometrial transformation to a receptive state is key to improving implantation rates in fertility treatments such as IVF. With microRNAs (miRNAs) increasingly recognized as important gene regulators, recent studies have investigated the role of miRNAs in the endometrium. Studies on miRNAs in endometrial disorders such as endometriosis and endometrial cancer have been reviewed previously. In this minireview, we aim to provide an up-to-date knowledge of miRNAs in the regulation of endometrial receptivity. Since endometrial remodelling differs considerably between species, we firstly summarised the key events of the endometrial cycle in humans and mice and then reviewed the miRNAs identified so far in these two species with likely functional significance in receptivity establishment. To date, 29 miRNAs have been reported in humans and 15 miRNAs in mice within various compartments of the endometrium that may potentially modulate receptivity; miRNAs regulating the Wnt signalling and those from the let-7, miR-23, miR-30, miR-200 and miR-183 families are found in both species. Future studies are warranted to investigate miRNAs as biomarkers and/or therapeutic targets to detect/improve endometrial receptivity in human fertility treatment.

1. Introduction

Embryo implantation requires a well-developed embryo implanting into the inner lining of the uterus called the endometrium. For implantation to succeed, the endometrium must undergo major molecular and cellular changes beforehand to transform from a nonreceptive to a receptive state. A blastocyst cannot implant if the endometrium is not ‘receptive’ [1]. The human endometrium is a dynamic tissue that undergoes cyclic remodelling across the menstrual cycle, becoming receptive for embryo implantation only for a short period of time in each cycle [1,2]. It is estimated that approximately one third of implantation failures in healthy women are likely due to inadequate endometrial receptivity [3]. Implantation failure is also commonly experienced by those who undergo fertility treatment such as in vitro fertilization (IVF) where ~60–70% of high-quality embryos fail to implant, and non- or sub-receptive endometrium is believed to be an important contributor [4,5]. Understanding the molecular regulation of endometrial receptivity is thus critical to improve fertility in general and to increase implantation rates in fertility treatment.
Endometrial remodelling differs between humans (undergoing the menstrual cycle) and animal species such as mice (undergoing the oestrous cycle) [6,7]. It is also known that the development of receptivity involves multifaceted changes within various endometrial cell types and the intrauterine environment [8,9,10]. This minireview firstly synopsizes the endometrial cycle in humans and mice. It then discusses microRNAs (miRNAs) that have been discovered in different compartments of the endometrium during the development of receptivity in both species. MiRNAs secreted from the endometrium and/or embryos that may influence endometrial receptivity are also briefly reviewed. Discussions are then made on miRNAs as potential biomarkers or therapeutic targets in human fertility treatment.

2. Brief Summary of the Endometrial Cycle in Humans and Mice

2.1. The Human Menstrual Cycle and Endometrial Receptivity

The human endometrium consists of two distinctive regions, the basalis and the functionalis [11,12] (Figure 1A). The basalis is a permanent layer that acts as the base for endometrial regeneration following menstruation [7], whereas the functionalis thickens and remodels during the menstrual cycle; it is this part of the endometrium that facilitates embryo implantation in a conceptual cycle but sheds in every nonconceptual cycle [1]. The menstrual cycle is regulated primarily by ovarian steroid hormones, especially estrogen and progesterone [7], and endometrial changes across an average 28-day cycle can be categorized into three phases (Figure 1B): menstrual, proliferative and secretory [7]. The menstrual phase involves the shedding of the functionalis following the rapid decline in estrogen and progesterone due to degeneration of the corpus luteum (CL) in the ovary. Menses eliminates the decidualised tissue which is irreversibly differentiated; the endometrium then regrows and reprepares for potential pregnancies in the subsequent cycle [13].
The proliferative phase is the regeneration of the functionalis. Major activities include re-epithelialisation while maintaining developmental plasticity to prevent endometrial scarring [14,15] and elongation of glands and vasculature to form a mature endometrium [14,16]. Sufficient tissue growth during this phase is crucial for the development of receptivity in the next phase, and estrogen produced by the developing ovarian follicles is the dominant hormone [7]. Estrogen receptors α (ER-α) and β (ER-β) are both expressed in the endometrium and peak during the proliferative phase [7,17], with ER-α in epithelial/stromal cells and blood vessels and ER-β predominantly in the epithelium [7,17].
Following ovulation, the thickened endometrial tissue enters the secretory phase, starting to differentiate in anticipation of embryo implantation (Figure 1B). Progesterone produced by the CL peaks in the mid-secretory phase and acts as the key driver of multiple molecular and morphological changes for the development of endometrial receptivity [2,18,19]. For instance, endometrial epithelial cells become highly secretory, while the stroma undergoes a terminal differentiation process known as decidualisation [1,20]. Decidualised cells secrete the necessary extracellular matrix required for a successful pregnancy [1,13,20]. Both ER-α and ER-β are downregulated by progesterone in the secretory phase [7,14,17].
The endometrium becomes transiently receptive during the mid-secretory phase (day 20–24), approximately 6–10 days after the ovulation (Figure 1B) [2,21], and this period is also known as the “window of implantation” [21,22]. During implantation (Figure 1C), the blastocyst firstly apposes itself to the surface of the receptive endometrium, establishing a loose connection between the trophectoderm of the blastocyst and the luminal epithelium of the endometrium [23,24]. Thereafter, the blastocyst firmly attaches to the luminal epithelium and then invades this layer of epithelial cells and moves to the stromal compartment. The luminal epithelium then reseals and encloses the embryo within the endometrial tissue [25].
Embryo transfer outside of the receptive window results in implantation failure or increased risk of early pregnancy loss [3,26], highlighting the importance of endometrial receptivity. To date, many molecular changes associated with endometrial receptivity have been found and have been reviewed previously [9,27,28,29,30,31,32]. Because the blastocyst directly interacts with the luminal epithelium to initiate implantation, changes in this layer of endometrial epithelial cells are essential for endometrial receptivity [23,24]. Past studies have shown that luminal epithelial cells are well-polarised during the nonreceptive state, but polarity-supporting factors are downregulated at receptivity [32]. Progesterone is known to upregulate adhesion molecules during the secretory phase [33]. Recently, we have discovered membrane protein podocalyxin (PODXL) as the key negative regulator of human endometrial epithelial receptivity [34]. PODXL is expressed in all epithelial (luminal and glandular) and endothelial cells in the nonreceptive phase but downregulated selectively in the luminal epithelium in the receptive phase [34]. Functionally, PODXL inhibits embryo attachment and invasion in in vitro models [34,35], and insufficient downregulation of PODXL in the luminal epithelium is associated with implantation failure in patients undergoing IVF treatment [35]. At the molecular level, PODXL promotes an impermeable/polarized epithelium and inhibits pro-implantation factors to negatively control endometrial epithelial receptivity [36]. PODXL is thus a potentially important regulator of human endometrial epithelial receptivity.

2.2. The Murine Estrous Cycle and the Key Differences in Endometrial Receptivity Development between Mice and Humans

The murine uterus, consisting of two horns, undergoes the oestrous cycle (Figure 2). Each cycle lasts approximately 4–5 days and comprises four phases: proestrus, oestrus, metestrus and diestrus [14,37]. Proestrus is characterised by increased blood flow and distension of the uterus, while epithelial cells and leukocytes begin to proliferate. At oestrus, leukocyte numbers drop significantly and epithelial cell proliferation increases. During metestrus, epithelial cells begin to degenerate, leukocytes proliferate once again and the uterus is no longer distended. The degenerated tissue is then reabsorbed by the uterus during diestrus [6,38,39]. Thus, the uterine tissue does not shed, and no bleeding occurs during the oestrous cycle.
The murine oestrous cycle is also controlled by ovarian hormones estrogen and progesterone. However, unlike humans in whom endometrial receptivity is established in the mid-secretory phase of the menstrual cycle, female mice are sexually active only during the oestrous phase, and the endometrium becomes receptive around 4.5 days post-coitus [38,40]. Among many molecular changes, a transient rise of oestradiol on day 4 of pregnancy, the nidatory oestradiol surge, is key to triggering the expression of several endometrial factors, such as the leukemia inhibitory factor (LIF), to fully induce endometrial receptivity for implantation [41].
Furthermore, while shifts in hormonal levels during the menstrual cycle induce spontaneous endometrial stromal decidualisation in humans [13], the murine uterus does not undergo decidualisation until after an embryo has contacted the uterine epithelium for implantation [42]. Additionally, the orientation of the blastocyst during implantation differs between humans and mice. In humans, the polar trophectoderm (located nearest to the inner cell mass, ICM) of the blastocyst interacts with the endometrial luminal epithelium (Figure 1C), but in mice, the mural trophectoderm (located farthest from the ICM) interacts with the endometrial luminal epithelium at implantation (Figure 2) [38]. In humans, the blastocyst invades between luminal epithelial cells, whereas in mice, luminal epithelial cells undergo apoptosis at the site of embryo attachment to make space for the implanting blastocyst [43,44].
Due to these differences, whilst some characteristics identified in mice can be translatable to humans, mice are often not the most appropriate animal model to study human endometrial receptivity, especially epithelial receptivity, for embryo implantation.

3. MicroRNAs and Endometrial Receptivity

MiRNAs are an important class of non-coding RNAs; they are endogenous and single-stranded regulatory RNAs which can vary between 18 and 25 nucleotides in length [45,46,47]. MiRNAs function as regulators of mRNA and mainly target the 3′ untranslated region (UTR) of gene transcripts [48,49]. They bind to Argonaute proteins to form RNA-induced silencing complexes and cause cleavage or translational repression of mRNA targets [50]; the degree of sequence complementation between the miRNA and the mRNA target dictates the strength of regulation [47]. Currently, over 2500 mature miRNAs have been identified [48], many of which are shown to be involved in reproduction and pregnancy.
Previous reviews discussed miRNAs in reproduction, covering both the male and female systems [51,52,53,54]; those on pregnancy placed more emphasis on miRNAs and maternal immune tolerance [55,56] or pregnancy complications [57,58,59]. MiRNAs in gynaecological diseases have also been reviewed [60,61,62]. However, the endometrium has often been overlooked in reviews concerning miRNAs, except when endometrial miRNAs are related to endometriosis [63,64,65,66,67,68,69,70] or endometrial cancer [71,72,73,74,75,76]. The remainder of this paper thus focuses on miRNAs in endometrial receptivity which has not been reviewed previously. Studies in both humans and mice are discussed.

3.1. MiRNAs Identified in the Endometrial Tissue

Not all studies of miRNAs in endometrial receptivity focused on a particular cell type; instead, many analysed endometrial tissues without revealing the exact location of miRNA production/action. The key miRNAs identified by this approach are listed in Table 1.
Two separate studies identified the LIF in the murine environment as a target gene of miRNAs, specifically of miR-181 [77] and miR-223 [78]. LIF has been well-established to play an important role in endometrial receptivity and implantation [79]. LIF is shown to be upregulated in fertile women compared to infertile women during the mid-secretory phase, suggesting its importance for receptivity [80]. In the studies assessing miRNAs that target LIF, upregulation of miR-181 and miR-223 supressed LIF expression in the murine endometrium and compromised embryo implantation [77,78]. Potential miRNA regulation of LIF was also identified in humans, specifically by miR-494 [81]. In silico prediction tools identified LIF as a target of miR-494 in the human endometrial tissue, where downregulation of miR-494 was associated with a high expression of LIF [81]. LIF appears to be a common target of multiple miRNAs in the development of endometrial receptivity; however, it is unknown whether some of these miRNAs are more physiologically important than others.
Another in silico study identified a further nineteen miRNAs that are potentially involved in receptivity development [3], in particular miR-130b, miR-548n/ah, miR-30c and miR-449, which are downregulated during the receptive phase [3]. It is of note that downregulation of miR-30c has also been associated with endometrial cancer [82]. It has been predicted that downregulation of these miRNAs during the receptive phase corresponds to the upregulation of 11 target genes, which include ephrin-A1 (EFNA1), growth arrest and DNA damage-inducible protein 45 alpha (GADD45A), annexin A4 (ANXA4), secreted phosphoprotein 1 (SPP1), AT-rich interaction domain 5B (ARID5B), interleukin 15 (IL15), Dickkopf-related protein 1 (DKK1), insulin-like growth factor binding protein 1 (IGFBP1) [3].
Other miRNAs found to regulate receptivity in the murine endometrial tissue include miR-101 and miR-199a, which target the cyclooxygenase-2 gene (Cox-2) [83]. Cox-2 was previously identified as a critical molecule for murine implantation and appears to have an inverse relationship with miR-101 and miR-199a [83]. However, another study reported that miR-199a may also target mucin-1 (MUC1), which has been established to maintain murine endometrial epithelial cells in a non-receptive state [84]. It is also reported that miR-193, which targets growth factor receptor-bound protein 7 (GRB7) [85], is involved in murine embryo implantation. However, the exact role of GRB7 in endometrial receptivity is unknown. Additionally, studies using human endometrial tissue identified miR-135b to be elevated in infertile women, and this miRNA is shown to downregulate homeobox A10 (HOXA10) expression [86].
A recent study profiled murine uterine miRNA transcriptomes at 1 day (non-receptive), 4 days (receptive) and 5 days (implantation) post-coitus [87]; 138 miRNAs were found to be differentially expressed across these timepoints, and the five most upregulated (miRNAs, miR-27a, miR-96, miR-106a, mir-23a and miR-200b) targeted a substantial proportion of the detected mRNAs [87]. Among these five miRNAs, miR-27a has also been detected in human serum samples; however, miR-27a is upregulated in patients with chronic endometritis [88] with no association with pregnancy outcomes.
Table 1. The miRNAs identified in endometrial tissues.
Table 1. The miRNAs identified in endometrial tissues.
miRNASpeciesTarget Gene/ProteinImproved/Impaired ReceptivityRef.
miR-30cHuman↓EFNA1, ARID5BImpaired[3]
miR-101MouseCox-2Impaired[83]
miR-130bHuman↓ANXA4, IL15Impaired[3]
miR-135bHuman↓HOXA10Impaired[86]
miR-181Mouse↓LIFImpaired[77]
miR-193MouseGRB7Impaired[85]
miR-199aMouseCox-2, MUC1Variable between gene targets[83,84]
miR-223Mouse↓LIFImpaired[78]
miR-449cHuman↓DKK1, IGFBP1Impaired[3]
miR-548ah/nHuman↓EFNA1, GADD45A, ANXA4, SPP1, ARID5BImpaired[3]

3.2. MiRNAs Identified in Endometrial Epithelial Cells

MiRNAs identified to promote endometrial epithelial receptivity include the let-7 family [89,90], miR-23b [91], miR-30b/d [81,92], miR-183-5p [93], miR-192 [94] and miR-494 [81] (Table 2). On the other hand, miR-23a-3p [95], miR-30a-3p [96], miR-30d [97], miR-125b [98], miR-145 [99,100], miR-200c [101], miR-429 and miR-5088 [102] are reported to hinder epithelial receptivity (Table 2).
While both the let-7-a and let-7-g families of miRNAs affect murine endometrial receptivity, only let-7-a is involved in human endometrial receptivity [89]. This particular study identified that Wnt signalling impairs uterine receptivity and that let-7-a/g, via downregulating β-catenin, represses Wnt to improve endometrial receptivity [89]. MiR-23b is also shown to indirectly influence Wnt signalling in women with recurring implantation failure (RIF) via targeting secreted frizzled-related protein 4 (sFRP-4), a modulatory protein of the Wnt pathway. Women with RIF are shown to have elevated levels of miR-23b, causing significantly lower levels of sFRP-4 in comparison to fertile women [91]. As sFRP-4 negatively regulates the Wnt pathway to increase endometrial receptivity, downregulation of sFRP-4 in RIF patients potentially impairs their receptivity [91]. Another miRNA, miR-183-5p, which targets catenin alpha 2 (CTNNA2), is reported to indirectly regulate the Wnt signalling pathway in the both murine and human endometrial epithelial cell line Ishikawa [93]. Both sFRP-4 and CTNNA2 negatively regulate the Wnt pathway to increase endometrial receptivity [91,93].
However, one study suggests that miR-23a downregulates cullin-3 (CUL3) to promote receptivity in Ishikawa cells [95], whereas other studies report that downregulation of CUL3 causes upregulation of β-catenin, ultimately impairing rather than increasing endometrial receptivity and implantation [89,91,93]. Therefore, there are conflicting results regarding the role of miR-23a in the regulation of endometrial receptivity.
Besides β-catenin, MUC1 has also been identified as a target of let-7-a/g in mice [90]. Downregulation of MUC1 by let-7 improves, whereas upregulation of MUC1 by miR-192 impairs endometrial receptivity in mice [94]. This indicates that in the murine endometrium, regulation of MUC1 via let-7, miR-192 and other miRNAs that are yet to be identified would modulate receptivity. MiR-192 also upregulates the adherens junction protein E-cadherin, which may also contribute to the impairment of endometrial receptivity in mice [94]. Furthermore, miR-192 downregulates Rho GTPase-activating protein 19 (ARHGAP19), which is involved in the remodelling of junctional proteins in murine endometrial epithelial cells [103].
The miR-30 family has been investigated in endometrial epithelial receptivity. Two extensive microarray and proteomics studies using receptive phase human endometrial epithelial cells have identified hundreds of gene targets of miR-30b/d, with varying degrees of up- or downregulation [81,92]. In mice, upregulation of miR-30a-3p and miR-30d has opposing impacts: elevated miR-30a-3p reduces receptivity by suppressing snail family transcriptional repressor 2 (Snai2) [96], whereas miR-30d deficiency impairs overall implantation [97].
In the previously mentioned study by Altmäe et al. [81], miR-494 also has hundreds of potential targets, but the following are particularly of note: calpastatin (CAST), cystic fibrosis transmembrane conductance regulator (CFTR), dihydropyrimidinase-like 2 (DPYSL2), F11 receptor (F11R), fibroblast growth factor receptor 2 (FGFR2), LIF, metal-regulatory transcription factor 1 (MTF1), neuronal PAS domain protein 2 (NPAS2), peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PPARGC1A), transforming acidic coiled-coil-containing protein 2 (TACC2) and RAB40B. While all display some degree of regulation by miR-494, the downregulation of miR-494 in the receptive phase leads to 5–10× greater upregulation of the CFTR and LIF than of the other target genes [81].
Multiple studies of RIF have identified miR-145 as an important regulator of endometrial receptivity [91,99,100,102]. All these studies have used human endometrial tissue, and studies by Liu et al. [100] and Kang et al. [99] also used the Ishikawa cell line to identify cell-specific roles of miR-145. This miRNA has been found to primarily play a role in cell–cell interaction and junctional proteins [91,99] and regulate ER-α [102] and plasminogen activator inhibitor (PAI-1) [100]. In the study by Shi et al. [102], miR-145 is lower in patients with RIF; the same study also identified miR-429 and miR-5088 being upregulated and miR-4668-5p downregulated in patients with RIF [102].
Furthermore, miR-125b [98] and miR-200c [101] are reported to impair epithelial receptivity in the murine epithelium. MiR-125b represses the expression of matrix metallopeptidase 26 (MMP26), a major player of endometrial remodelling, to reduce endometrial receptivity [98]. On the other hand, miR-200c influences endometrial receptivity via indirectly targeting the Wnt pathway [101]. Through targeting the gene fucosyltransferase 4 (FUT4), miR-200c is suggested to indirectly inactivate β-catenin/Wnt signalling, leading to impaired endometrial receptivity and implantation [101]. The β-catenin/Wnt signalling pathway is emerging to be important in the regulation of endometrial epithelial receptivity. However, whether the pathway helps or hinders endometrial receptivity appears to be in question as the related molecular mechanisms are not yet fully understood.

3.3. MiRNAs Identified in Endometrial Stromal Cells

While endometrial epithelial cells are the first point of contact by the blastocyst to initiate implantation, endometrial stromal cells are vital in subsequent stages of implantation. As decidualisation must occur for implantation to progress properly, studies have also investigated the role of miRNAs in decidualisation in humans and mice. MiRNAs that have been identified as the key players in promoting decidualisation include miR-21 [104], miR-96 [87,105], miR-181a [106] and miR-200 [107] (Table 3); miRNAs associated with impairment of decidualisation consist of miR-22 [108], miR-141 [109], miR-148a [110], miR-181b [111], miR-194 [112], miR-200a [113,114] and miR-542 [115] (Table 3).
Using primary stromal cells isolated from the human endometrium, the studies by Yan et al. [104] and Zhang et al. [106] both identified Kruppel-like factor 12 (KLF12) as the target gene of miR-21 and miR-181a. Both miRNAs downregulate KLF12 to promote decidualisation in vitro. KLF12 was previously identified as a negative regulator of decidualisation in patients with RIF [104], and overexpression of KLF12 is associated with downregulation of decidualisation markers [106]. MiR-200 is also reported to be associated with decidualisation in human endometrial stromal cells via repressing zinc finger E-box binding homeobox 1 (ZEB1) expression [107].
In murine endometrial stromal cells, miR-96 is shown to play an important role in decidualisation [87,105]. The study by Yang et al. [105] identified miR-96 levels inversely correlating with the expression of antiapoptotic protein B-cell lymphoma 2 (Bcl2). In the study of Chen et al. [87], miR-96 targets Kruppel-like factor 13 (Klf13) and potentially regulates the progesterone receptor (PGR) in mice. This indicates that miR-96 likely plays a vital role in the decidualisation of murine endometrial stromal cells.
However, the expression of certain miRNAs can cause dysregulation of decidualisation. Patients with RIF are reported to have elevated levels of miR-22 with reduced levels of Tiam/Rac1, a signal pathway considered to be potentially important in early stromal cell decidualisation [108]. Overexpression of miR-148a is also associated with impairment of decidualisation in patients with RIF via the suppression of homeobox C8 (HOXC8) [110]. Furthermore, miR-542 [115] and miR-194 [112] are reported to dysregulate decidualisation of human endometrial stromal cells. In a human endometrial stromal cell line, miR-181b downregulates tissue inhibitor of metalloproteinase 3 (TIMP-3), but overall does not significantly affect decidualisation [111].
Two different miRNAs have been found to target the same gene in murine endometrial stromal cells, where upregulation of miR-141 [109] and miR-200a [113] negatively regulate the phosphatase and tensin homolog (PTEN) expression. Like Bcl2, PTEN is also believed to control proliferation and apoptosis of stromal cells, thus maintaining the correct environment for decidualisation [113]. Upregulation of miR-200a has also been associated with indirect downregulation of progesterone signalling in mice, reducing the overall implantation capacity [114].

3.4. MiRNAs Secreted by the Endometrium and/or the Embryo That May Influence Endometrial Receptivity

The endometrium can also be influenced by the implanting embryo. Several studies identified miRNAs that are expressed by embryos and present in embryo secretions [116,117,118,119,120]. Due to the increasingly common use of IVF techniques, blastocyst spent media have been analysed for miRNA and other biomarkers [116,117,118,119,120]. A few previous reviews discussed miRNAs and embryos, often regarding embryo development but also implantation [116,117,118,119,120]. Other previous reviews deliberated the miRNAs secreted by preimplantation blastocysts in facilitating the embryo–endometrium crosstalk to promote implantation [118,120]. Hence, no extensive discussion on this topic is made here. Embryo-derived miRNAs have also been suggested to play a role in implantation failure. For instance, miR-661 is identified to be secreted by human blastocysts, but uptake of this miRNA by human epithelial endometrial cells (HEECs) reduces their ability for trophoblast spheroids attachment in in vitro implantation models [121]. Other studies have identified miR-519d [122] and miR-155 [123] as negative regulators of trophoblast proliferation and migration during implantation. However, it is unknown if miR-519d and miR-155 also regulate endometrial cells for implantation [122,123].
While mRNA relies on the modifications to the RNA strand to improve its stability [124], miRNAs are only 18–25 nucleotides long [45,46,47] and generally do not undergo these modifications. Some miRNAs can circulate freely within the body, but the vast majority are located within extracellular vesicles (EVs), either in the form of microvesicles or exosomes [125,126], and circulate through the body while ‘packaged’. Multiple reviews have discussed EVs that are secreted from the endometrium and/or embryos and contain miRNAs, some of which are found to potentially enhance endometrial receptivity and embryo implantation [126,127,128,129]. Not all of the studies used human samples, but of those that did, the miR-30 family was identified multiple times as important miRNAs in human endometrial EVs [127,129].
The endometrium is proposed to have a mechanism to screen embryo quality prior to implantation [130], and EVs secreted by embryos have been suggested as the mediator where the cargo in the EVs is exposed to and assessed by the endometrium for embryo quality [130]. Embryonic EVs have been suggested to epigenetically regulate endometrial cells via EV-mediated RNA transfer, and poor embryos are unable to establish the same epigenetic changes [131]. There is growing evidence suggesting that the endometrium also has an active role in selecting embryonic EVs based on the cargo profile, which could include miRNAs such as the let-7 family, miR-31, miR-101a, miR-199a, miR-200 and the aforementioned miR-661 [121,132]. Other studies have predicted that miRNAs derived from the blastocyst culture medium may potentially modulate endometrial epithelial proliferation and embryo attachment [31,130,133].

4. Potential Clinical Applications

Since the first successful IVF birth in 1978 [134], more than 8 million babies have been born using assisted reproductive technologies, with the technique becoming commonplace globally [135,136]. However, as discussed earlier, implantation failure limits IVF treatment, and inadequate endometrial receptivity is an important contributing factor. With the identification of miRNAs as important players in the establishment of endometrial receptivity, the presence of miRNAs in serum may allow for clinical diagnosis of fertility and receptivity [116,117,118]. Furthermore, as the use of miRNAs as therapeutics has been pushed into clinical trials in other fields [137,138], the utility of miRNAs in endometrial preparation in IVF can also be speculated.

4.1. MiRNAs for Detecting/Predicting Endometrial Receptivity

While many studies have sought to identify miRNAs in specific cell populations within the endometrium or in relevant bodily fluids, a few studies have investigated miRNAs as clinical biomarkers for infertility and receptivity (Table 4). Some of these studies have compared miRNA in the endometrium and in serum to identify physiologically relevant biomarkers. These include miR-27a [88], miR-31 [139], mir-152 and miR-155 [140], which are found to be expressed in the endometrium and detected in serum. In the study by Kresowik et al. [139], miR-31 is elevated in both the endometrium and serum of normal fertile women in the window of implantation. Upregulation of miR-31 is associated with the downregulation of FOXP3 and CXCL12, which may be the potential target genes [139]. On the other hand, miR-27a, miR-152 and miR-155 have been shown to be upregulated in the endometrium and serum of patients with RIF [140] or chronic endometritis [88]. Studies by Ghaebi et al. [141] also identified miR-155 as a potential biomarker. However, miR-155 is reported to be downregulated in patients with RIF, contradicting the findings by Drissennek et al. [140]. Other miRNAs which are significantly altered in the serum of patients with RIF include miR-25, miR-93, miR-106b (upregulated) and miR-146a (downregulated) [141].

4.2. MiRNAs for Improving Endometrial Receptivity

Currently, most miRNA therapeutics trials are likely to be miRNA inhibitors over miRNA mimics [142]. However, patent applications for both types do exist and tend to be cancer-focused [142,143]. While not yet available in the market, some miRNAs have been in clinical trials, with anti-miRNA drug Miravirsen targeting miR-122 for the treatment of the hepatitis C virus [137]. While there is some interest in developing treatments to target the endometrium [144], no miRNA therapeutics are currently in clinical trials. While technologies required for effectively transferring miRNAs are still being heavily researched, the vast majority relies on the use of nanoparticles to prevent degradation of miRNAs or miRNA inhibitors [144,145,146,147]. Previous studies showed that endogenous EVs do exist in the uterine space and can safely deliver miRNAs between tissues [126,128,129]. The use of synthetically produced exogenous EVs may allow transfer of miRNA therapeutics to the endometrium and avoid unnecessary off-target sites. However, this is still an emerging field, and the types of nanoparticles and their surface molecules differ between studies [144,145,146,147].

5. Conclusions

The endometrium is a complex and dynamic tissue; understanding the molecular changes that govern endometrial receptivity is vital to improve fertility treatment. This review summarised miRNAs that have been identified to date as potential regulators of endometrial receptivity in both mice and humans. Because of the key species-specific differences in the fundamental biology of endometrial remodelling, it is important to bear in mind that discoveries made in mice are not automatically applicable to humans. Encouragingly, a few miRNAs have been found to potentially regulate endometrial receptivity in both species; these include those that influence the Wnt signalling pathway and those from the let-7, miR-23, miR-30, miR-200 and miR-183 families. Future studies need to further investigate the clinical significance of these miRNAs.

Author Contributions

Conceptualisation, G.N.; investigation, M.S.; data curation, M.S.; writing—original draft preparation, M.S.; writing—review and editing, M.S., S.H. and G.N.; visualisation, M.S.; supervision, S.H. and G.N.; project administration, G.N.; funding acquisition, G.N. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by the National Health and Medical Research Council of Australia (grant No. 1156666 to G.N.) and the Australian Government Research Training Program (scholarship to M.S.).

Conflicts of Interest

The authors declare no conflict of interests.

References

  1. Evans, J.; Salamonsen, L.; Winship, A.; Menkhorst, E.; Nie, G.; Gargett, L.A.S.C.E.; Dimitriadis, E. Fertile ground: Human endometrial programming and lessons in health and disease. Nat. Rev. Endocrinol. 2016, 12, 654–667. [Google Scholar] [CrossRef] [PubMed]
  2. Gellersen, B.; Brosens, I.A.; Brosens, J.J. Decidualization of the Human Endometrium: Mechanisms, Functions, and Clinical Perspectives. Semin. Reprod. Med. 2007, 25, 445–453. [Google Scholar] [CrossRef] [PubMed]
  3. Altmäe, S.; Koel, M.; Võsa, U.; Adler, P.; Suhorutšenko, M.; Laisk-Podar, T.; Kukushkina, V.; Saare, M.; Velthut-Meikas, A.; Krjutškov, K.; et al. Meta-signature of human endometrial receptivity: A meta-analysis and validation study of transcriptomic biomarkers. Sci. Rep. 2017, 7, 10077. [Google Scholar] [CrossRef] [PubMed]
  4. Heng, S.; Cervero, A.; Simon, C.; Stephens, A.N.; Li, Y.; Zhang, J.; Paule, S.; Rainczuk, A.; Singh, H.; Quinonero, A.; et al. Proprotein Convertase 5/6 Is Critical for Embryo Implantation in Women: Regulating Receptivity by Cleaving EBP50, Modulating Ezrin Binding, and Membrane-Cytoskeletal Interactions. Endocrinology 2011, 152, 5041–5052. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Paulson, R.J.; Hatch, I.E.; Lobo, R.A.; Sauer, M.V. Cumulative conception and live birth rates after oocyte donation: Implications regarding endometrial receptivity. Hum. Reprod. 1997, 12, 835–839. [Google Scholar] [CrossRef]
  6. Boyd, K.L.; Muehlenbachs, A.; Rendi, M.H.; Garcia, R.L.; Gibson-Corley, K.N. 17—Female Reproductive System. In Comparative Anatomy and Histology, 2nd ed.; Treuting, P.M., Dintzis, S.M., Montine, K.S., Eds.; Academic Press: San Diego, CA, USA, 2018; pp. 303–334. [Google Scholar]
  7. Salamonsen, L.A. Menstruation and the estrous cycle. In Essential Reproduction; Johnson, M.H., Ed.; Blackwell Pub.: Malden, MA, USA, 2007; pp. 25–45. [Google Scholar]
  8. Salamonsen, L.A.; Nie, G.; Hannan, N.J.; Dimitriadis, E. Society for Reproductive Biology Founders’ Lecture 2009. Preparing fertile soil: The importance of endometrial receptivity. Reprod. Fertil. Dev. 2009, 21, 923–934. [Google Scholar] [CrossRef]
  9. Lessey, B.A.; Young, S.L. What exactly is endometrial receptivity? Fertil. Steril. 2019, 111, 611–617. [Google Scholar] [CrossRef]
  10. Ochoa-Bernal, M.A.; Fazleabas, A.T. Physiologic Events of Embryo Implantation and Decidualization in Human and Non-Human Primates. Int. J. Mol. Sci. 2020, 21, 1973. [Google Scholar] [CrossRef] [Green Version]
  11. Tortora, G.J.; Derrickson, B.H. Principles of Anatomy and Physiology; John Wiley & Sons: Hoboken, NJ, USA, 2008. [Google Scholar]
  12. Ellis, H. Anatomy of the uterus. Anaesth. Intensive Care Med. 2005, 6, 74–75. [Google Scholar] [CrossRef]
  13. Hannan, N.J.; Evans, J.; Salamonsen, L.A. Alternate roles for immune regulators: Establishing endometrial receptivity for implantation. Expert Rev. Clin. Immunol. 2011, 7, 789–802. [Google Scholar] [CrossRef]
  14. Salamonsen, L. Menstrual and estrous cycles. In The Endometrium: Molecular, Cellular and Clinical Perspectives; Aplin, J.D., Ed.; Informa Healthcare: London, UK, 2008. [Google Scholar]
  15. Young, S.L.; Evans-Hoeker, E.A. Endometrial Receptivity and Intrauterine Adhesive Disease. Semin. Reprod. Med. 2014, 32, 392–401. [Google Scholar] [CrossRef]
  16. Gambino, L.S.; Wreford, N.G.; Bertram, J.; Dockery, P.; Lederman, F.; Rogers, P.A. Angiogenesis occurs by vessel elongation in proliferative phase human endometrium. Hum. Reprod. 2002, 17, 1199–1206. [Google Scholar] [CrossRef] [PubMed]
  17. Salamonsen, L.A.; Jones, R.L. Endometrial Remodeling. In Encyclopedia of Hormones; Henry, H.L., Norman, A.W., Eds.; Academic Press: New York, NY, USA, 2003; pp. 504–512. [Google Scholar]
  18. Senger, P.L. Pathways to Pregnancy and Parturition; Current Conceptions, Inc.: Redmond, OR, USA, 2003. [Google Scholar]
  19. Knight, P.G.; Glister, C. TGF-β superfamily members and ovarian follicle development. Reproduction 2006, 132, 191–206. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Gellersen, B.; Brosens, J. Cyclic Decidualization of the Human Endometrium in Reproductive Health and Failure. Endocr. Rev. 2014, 35, 851–905. [Google Scholar] [CrossRef]
  21. Evans, J.; Hannan, N.J.; Hincks, C.; Rombauts, L.J.F.; Salamonsen, L.A. Defective Soil for a Fertile Seed? Altered Endometrial Development Is Detrimental to Pregnancy Success. PLoS ONE 2012, 7, e53098. [Google Scholar] [CrossRef]
  22. Cakmak, H.; Taylor, H.S. Implantation failure: Molecular mechanisms and clinical treatment. Hum. Reprod. Updat. 2010, 17, 242–253. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Kim, S.-M.; Kim, J.-S. A Review of Mechanisms of Implantation. Dev. Reprod. 2017, 21, 351–359. [Google Scholar] [CrossRef] [Green Version]
  24. Zhang, S.; Lin, H.; Kong, S.; Wang, S.; Wang, H.; Wang, H.; Armant, D.R. Physiological and molecular determinants of embryo implantation. Mol. Asp. Med. 2013, 34, 939–980. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. James, J.; Carter, A.M.; Chamley, L. Human placentation from nidation to 5 weeks of gestation. Part I: What do we know about formative placental development following implantation? Placenta 2012, 33, 327–334. [Google Scholar] [CrossRef]
  26. Gardner, D.K. In Vitro Fertilization: A Practical Approach; CRC Press: Boca Raton, FL, USA, 2006. [Google Scholar]
  27. Lessey, B.A. Assessment of endometrial receptivity. Fertil. Steril. 2011, 96, 522–529. [Google Scholar] [CrossRef]
  28. Campbell, K.L.; Rockett, J.C. Biomarkers of ovulation, endometrial receptivity, fertilisation, implantation and early pregnancy progression. Paediatr. Périnat. Epidemiol. 2006, 20, 13–25. [Google Scholar] [CrossRef] [PubMed]
  29. Craciunas, L.; Gallos, I.; Chu, J.; Bourne, T.; Quenby, S.; Brosens, J.J.; Coomarasamy, A. Conventional and modern markers of endometrial receptivity: A systematic review and meta-analysis. Hum. Reprod. Updat. 2019, 25, 202–223. [Google Scholar] [CrossRef]
  30. Whitby, S.; Zhou, W.; Dimitriadis, E. Alterations in Epithelial Cell Polarity During Endometrial Receptivity: A Systematic Review. Front. Endocrinol. 2020, 11, 596324. [Google Scholar] [CrossRef] [PubMed]
  31. Governini, L.; Luongo, F.P.; Haxhiu, A.; Piomboni, P.; Luddi, A. Main actors behind the endometrial receptivity and successful implantation. Tissue Cell 2021, 73, 101656. [Google Scholar] [CrossRef] [PubMed]
  32. Whitby, S.; Salamonsen, L.A.; Evans, J. The Endometrial Polarity Paradox: Differential Regulation of Polarity Within Secretory-Phase Human Endometrium. Endocrinology 2017, 159, 506–518. [Google Scholar] [CrossRef] [PubMed]
  33. Achache, H.; Revel, A. Endometrial receptivity markers, the journey to successful embryo implantation. Hum. Reprod. Updat. 2006, 12, 731–746. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Paule, S.G.; Heng, S.; Samarajeewa, N.; Li, Y.; Mansilla, M.; Webb, A.I.; Nebl, T.; Young, S.L.; Lessey, B.A.; Hull, M.L.; et al. Podocalyxin is a key negative regulator of human endometrial epithelial receptivity for embryo implantation. Hum. Reprod. 2021, 36, 1353–1366. [Google Scholar] [CrossRef]
  35. Heng, S.; Samarajeewa, N.; Aberkane, A.; Essahib, W.; Van de Velde, H.; Scelwyn, M.; Hull, M.L.; Vollenhoven, B.; Rombauts, L.J.; Nie, G. Podocalyxin inhibits human embryo implantation in vitro and luminal podocalyxin in putative receptive endometrium is associated with implantation failure in fertility treatment. Fertil. Steril. 2021, 116, 1391–1401. [Google Scholar] [CrossRef]
  36. Heng, S.; Samarajeewa, N.; Wang, Y.; Paule, S.G.; Breen, J.; Nie, G. Podocalyxin promotes an impermeable epithelium and inhibits pro-implantation factors to negatively regulate endometrial receptivity. Sci. Rep. 2021, 11, 24016. [Google Scholar] [CrossRef]
  37. Croy, B.A.; Yamada, A.T.; DeMayo, F.J.; Adamson, S.L. The Guide to Investigation of Mouse Pregnancy; Academic Press: Cambridge, MA, USA, 2013. [Google Scholar]
  38. Matsumoto, H. Molecular and cellular events during blastocyst implantation in the receptive uterus: Clues from mouse models. J. Reprod. Dev. 2017, 63, 445–454. [Google Scholar] [CrossRef] [Green Version]
  39. Caligioni, C.S. Assessing Reproductive Status/Stages in Mice. Curr. Protoc. Neurosci. 2009, 48, A.4I.1–A.4I.8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Kelleher, A.; Burns, G.W.; Behura, S.; Wu, G.; Spencer, T.E. Uterine glands impact uterine receptivity, luminal fluid homeostasis and blastocyst implantation. Sci. Rep. 2016, 6, 38078. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Hewitt, S.C.; Harrell, J.C.; Korach, K.S. Lessons in Estrogen Biology from Knockout and Transgenic Animals. Annu. Rev. Physiol. 2005, 67, 285–308. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Wang, H.; Dey, S.K. Roadmap to embryo implantation: Clues from mouse models. Nat. Rev. Genet. 2006, 7, 185–199. [Google Scholar] [CrossRef]
  43. Dey, S.K.; Lim, H.; Das, S.K.; Reese, J.; Paria, B.C.; Daikoku, T.; Wang, H. Molecular Cues to Implantation. Endocr. Rev. 2004, 25, 341–373. [Google Scholar] [CrossRef]
  44. Pampfer, S.; Donnay, I. Apoptosis at the time of embryo implantation in mouse and rat. Cell Death Differ. 1999, 6, 533–545. [Google Scholar] [CrossRef] [Green Version]
  45. Hammond, S.M. An overview of microRNAs. Adv. Drug Deliv. Rev. 2015, 87, 3–14. [Google Scholar] [CrossRef] [Green Version]
  46. Winter, J.; Jung, S.; Keller, S.; Gregory, R.I.; Diederichs, S. Many roads to maturity: MicroRNA biogenesis pathways and their regulation. Nat. Cell Biol. 2009, 11, 228–234. [Google Scholar] [CrossRef]
  47. Bartel, D.P. MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell 2004, 116, 281–297. [Google Scholar] [CrossRef] [Green Version]
  48. Gebert, L.F.R.; Macrae, I.J. Regulation of microRNA function in animals. Nat. Rev. Mol. Cell Biol. 2019, 20, 21–37. [Google Scholar] [CrossRef]
  49. O’Brien, J.; Hayder, H.; Zayed, Y.; Peng, C. Overview of MicroRNA Biogenesis, Mechanisms of Actions, and Circulation. Front. Endocrinol. 2018, 9, 402. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. O’Carroll, D.; Mecklenbrauker, I.; Das, P.P.; Santana, A.; Koenig, U.; Enright, A.J.; Miska, E.A.; Tarakhovsky, A. A Slicer-independent role for Argonaute 2 in hematopoiesis and the microRNA pathway. Genes Dev. 2007, 21, 1999–2004. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Imbar, T.; Galliano, D.; Pellicer, A.; Laufer, N. Introduction: MicroRNAs in human reproduction: Small molecules with crucial regulatory roles. Fertil. Steril. 2014, 101, 1514–1515. [Google Scholar] [CrossRef] [PubMed]
  52. Eisenberg, I.; Kotaja, N.; Goldman-Wohl, D.; Imbar, T. microRNA in Human Reproduction. In Advances in Experimental Medicine and Biology; Springer: Cham, Switzerland, 2015; Volume 888, pp. 353–387. [Google Scholar]
  53. Reza, A.M.M.T.; Choi, Y.-J.; Han, S.G.; Song, H.; Park, C.; Hong, K.; Kim, J.-H. Roles of microRNAs in mammalian reproduction: From the commitment of germ cells to peri-implantation embryos. Biol. Rev. 2018, 94, 415–438. [Google Scholar] [CrossRef] [PubMed]
  54. Hossain, M.M.; Sohel, M.H.; Schellander, K.; Tesfaye, D. Characterization and importance of microRNAs in mammalian gonadal functions. Cell Tissue Res. 2012, 349, 679–690. [Google Scholar] [CrossRef]
  55. Kamity, R.; Sharma, S.; Hanna, N. MicroRNA-Mediated Control of Inflammation and Tolerance in Pregnancy. Front. Immunol. 2019, 10, 718. [Google Scholar] [CrossRef] [Green Version]
  56. Bidarimath, M.; Khalaj, K.; Wessels, J.M.; Tayade, C. MicroRNAs, immune cells and pregnancy. Cell. Mol. Immunol. 2014, 11, 538–547. [Google Scholar] [CrossRef]
  57. Barchitta, M.; Maugeri, A.; Quattrocchi, A.; Agrifoglio, O.; Agodi, A. The Role of miRNAs as Biomarkers for Pregnancy Outcomes: A Comprehensive Review. J. Genom. 2017, 2017, 8067972. [Google Scholar] [CrossRef] [Green Version]
  58. Cai, M.; Kolluru, G.K.; Ahmed, A. Small Molecule, Big Prospects: MicroRNA in Pregnancy and Its Complications. J. Pregnancy 2017, 2017, 6972732. [Google Scholar] [CrossRef] [Green Version]
  59. Yang, H.; Ma, Q.; Wang, Y.; Tang, Z. Clinical application of exosomes and circulating microRNAs in the diagnosis of pregnancy complications and foetal abnormalities. J. Transl. Med. 2020, 18, 32. [Google Scholar] [CrossRef]
  60. Nothnick, W.B. The role of micro-RNAs in the female reproductive tract. Reproduction 2012, 143, 559–576. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Virant-Klun, I.; Ståhlberg, A.; Kubista, M.; Skutella, T. MicroRNAs: From Female Fertility, Germ Cells, and Stem Cells to Cancer in Humans. Stem Cells Int. 2015, 2016, 3984937. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Carletti, M.Z.; Christenson, L.K. MicroRNA in the ovary and female reproductive tract1. J. Anim. Sci. 2009, 87, E29–E38. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Aznaurova, Y.B.; Zhumataev, M.B.; Roberts, T.; Aliper, A.M.; Zhavoronkov, A.A. Molecular aspects of development and regulation of endometriosis. Reprod. Biol. Endocrinol. 2014, 12, 50. [Google Scholar] [CrossRef] [Green Version]
  64. Borghese, B.; Zondervan, K.T.; Abrao, M.S.; Chapron, C.; Vaiman, D. Recent insights on the genetics and epigenetics of endometriosis. Clin. Genet. 2016, 91, 254–264. [Google Scholar] [CrossRef] [Green Version]
  65. Guo, S.W. Epigenetics of endometriosis. Mol. Hum. Reprod. 2009, 15, 587–607. [Google Scholar] [CrossRef]
  66. Bjorkman, S.; Taylor, H.S. MicroRNAs in endometriosis: Biological function and emerging biomarker candidates. Biol. Reprod. 2019, 100, 1135–1146. [Google Scholar] [CrossRef]
  67. Ahn, S.H.; Singh, V.; Tayade, C. Biomarkers in endometriosis: Challenges and opportunities. Fertil. Steril. 2017, 107, 523–532. [Google Scholar] [CrossRef] [Green Version]
  68. Santamaria, X.; Taylor, H. MicroRNA and gynecological reproductive diseases. Fertil. Steril. 2014, 101, 1545–1551. [Google Scholar] [CrossRef]
  69. Teague, E.M.C.O.; Print, C.; Hull, M.L. The role of microRNAs in endometriosis and associated reproductive conditions. Hum. Reprod. Updat. 2009, 16, 142–165. [Google Scholar] [CrossRef] [Green Version]
  70. Pan, Q.; Chegini, N. MicroRNA signature and regulatory functions in the endometrium during normal and disease states. In Seminars in Reproductive Medicine; NIH Public Access: Bethesda, Maryland, 2008; Volume 26, p. 479. [Google Scholar]
  71. Matias-Guiu, X.; Prat, J. Molecular pathology of endometrial carcinoma. Histopathology 2012, 62, 111–123. [Google Scholar] [CrossRef]
  72. Delangle, R.; De Foucher, T.; Larsen, A.K.; Sabbah, M.; Azaïs, H.; Bendifallah, S.; Daraï, E.; Ballester, M.; Mehats, C.; Uzan, C.; et al. The Use of microRNAs in the Management of Endometrial Cancer: A Meta-Analysis. Cancers 2019, 11, 832. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Kanekura, K.; Nishi, H.; Isaka, K.; Kuroda, M. MicroRNA and gynecologic cancers. J. Obstet. Gynaecol. Res. 2016, 42, 612–617. [Google Scholar] [CrossRef] [PubMed]
  74. Vasilatou, D.; Sioulas, V.D.; Pappa, V.; Papageorgiou, S.G.; Vlahos, N.F. The role of miRNAs in endometrial cancer. Epigenomics 2015, 7, 951–959. [Google Scholar] [CrossRef]
  75. Sidorkiewicz, I.; Jóźwik, M.; Niemira, M.; Krętowski, A. Insulin Resistance and Endometrial Cancer: Emerging Role for microRNA. Cancers 2020, 12, 2559. [Google Scholar] [CrossRef] [PubMed]
  76. Favier, A.; Rocher, G.; Larsen, A.; Delangle, R.; Uzan, C.; Sabbah, M.; Castela, M.; Duval, A.; Mehats, C.; Canlorbe, G. MicroRNA as Epigenetic Modifiers in Endometrial Cancer: A Systematic Review. Cancers 2021, 13, 1137. [Google Scholar] [CrossRef]
  77. Chu, B.; Zhong, L.; Dou, S.; Wang, J.; Liangwen, Z.; Wang, M.; Shi, Q.; Mei, Y.; Wu, M. miRNA-181 regulates embryo implantation in mice through targeting leukemia inhibitory factor. J. Mol. Cell Biol. 2015, 7, 12–22. [Google Scholar] [CrossRef] [Green Version]
  78. Dong, X.; Sui, C.; Huang, K.; Wang, L.; Hu, D.; Xiong, T.; Wang, R.; Zhang, H. MicroRNA-223-3p suppresses leukemia inhibitory factor expression and pinopodes formation during embryo implantation in mice. Am. J. Transl. Res. 2016, 8, 1155–1163. [Google Scholar] [PubMed]
  79. Cavagna, M.; Mantese, J. Biomarkers of Endometrial Receptivity—A Review. Placenta 2003, 24, S39–S47. [Google Scholar] [CrossRef]
  80. Aghajanova, L. Leukemia Inhibitory Factor and Human Embryo Implantation. Ann. N. Y. Acad. Sci. 2004, 1034, 176–183. [Google Scholar] [CrossRef]
  81. Altmäe, S.; Martinez-Conejero, J.A.; Esteban, F.J.; Ruiz-Alonso, M.; Stavreus-Evers, A.; Horcajadas, J.A.; Salumets, A. MicroRNAs miR-30b, miR-30d, and miR-494 Regulate Human Endometrial Receptivity. Reprod. Sci. 2012, 20, 308–317. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Boren, T.; Xiong, Y.; Hakam, A.; Wenham, R.; Apte, S.; Wei, Z.; Kamath, S.; Chen, D.-T.; Dressman, H.; Lancaster, J.M. MicroRNAs and their target messenger RNAs associated with endometrial carcinogenesis. Gynecol. Oncol. 2008, 110, 206–215. [Google Scholar] [CrossRef] [PubMed]
  83. Chakrabarty, A.; Tranguch, S.; Daikoku, T.; Jensen, K.; Furneaux, H.; Dey, S.K. MicroRNA regulation of cyclooxygenase-2 during embryo implantation. Proc. Natl. Acad. Sci. USA 2007, 104, 15144–15149. [Google Scholar] [CrossRef] [Green Version]
  84. Inyawilert, W.; Fu, T.-Y.; Lin, C.-T.; Tang, P.-C. MicroRNA-199a mediates mucin 1 expression in mouse uterus during implantation. Reprod. Fertil. Dev. 2014, 26, 653–664. [Google Scholar] [CrossRef] [PubMed]
  85. Li, R.; He, J.; Chen, X.; Ding, Y.; Wang, Y.; Long, C.; Shen, L.; Liu, X. Mmu-miR-193 is involved in embryo implantation in mouse uterus by regulating GRB7 gene expression. Reprod. Sci. 2014, 21, 733–742. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Riyanti, A.; Febri, R.R.; Zakirah, S.C.; Harzif, A.K.; Rajuddin, R.; Muharam, R.; Asmarinah, A.; Wiweko, B. Suppressing HOXA-10 Gene Expression by MicroRNA 135b During the Window of Implantation in Infertile Women. J. Reprod. Infertil. 2020, 21, 217–221. [Google Scholar] [PubMed]
  87. Chen, K.; Chen, X.; He, J.; Ding, Y.; Geng, Y.; Liu, S.; Liu, X.; Wang, Y. Mouse Endometrium Temporal and Spatial Expression mRNA and MicroRNA Associated with Embryo Implantation. Reprod. Sci. 2015, 22, 1399–1408. [Google Scholar] [CrossRef] [PubMed]
  88. Di Pietro, C.; Caruso, S.; Battaglia, R.; Sareri, M.I.; La Ferlita, A.; Strino, F.; Bonaventura, G.; Di Mauro, M.; Barcellona, M.L.; Perciavalle, V.; et al. MiR-27a-3p and miR-124-3p, upregulated in endometrium and serum from women affected by Chronic Endometritis, are new potential molecular markers of endometrial receptivity. Am. J. Reprod. Immunol. 2018, 80, e12858. [Google Scholar] [CrossRef]
  89. Li, Q.; Liu, W.; Chiu, P.C.; Yeung, W.S. Mir-let-7a/g Enhances Uterine Receptivity via Suppressing Wnt/β-Catenin Under the Modulation of Ovarian Hormones. Reprod. Sci. 2020, 27, 1164–1174. [Google Scholar] [CrossRef]
  90. Inyawilert, W.; Fu, T.-Y.; Lin, C.-T.; Tang, P.-C. Let-7-mediated suppression of mucin 1 expression in the mouse uterus during embryo implantation. J. Reprod. Dev. 2015, 61, 138–144. [Google Scholar] [CrossRef] [Green Version]
  91. Revel, A.; Achache, H.; Stevens, J.; Smith, Y.; Reich, R. MicroRNAs are associated with human embryo implantation defects. Hum. Reprod. 2011, 26, 2830–2840. [Google Scholar] [CrossRef]
  92. Moreno-Moya, J.M.; Vilella, F.; Martínez, S.; Pellicer, A.; Simón, C. The transcriptomic and proteomic effects of ectopic overexpression of miR-30d in human endometrial epithelial cells. Mol. Hum. Reprod. 2014, 20, 550–566. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Akbar, R.; Ullah, K.; Rahman, T.U.; Cheng, Y.; Pang, H.-Y.; Jin, L.Y.; Wang, Q.-J.; Huang, H.-F.; Sheng, J.-Z. miR-183-5p regulates uterine receptivity and enhances embryo implantation. J. Mol. Endocrinol. 2020, 64, 43–52. [Google Scholar] [CrossRef] [PubMed]
  94. Liang, J.; Cao, D.; Zhang, X.; Liu, L.; Tan, Q.; Shi, S.; Chen, K.; Liang, J.; Wang, Z. miR-192-5p suppresses uterine receptivity formation through impeding epithelial transformation during embryo implantation. Theriogenology 2020, 157, 360–371. [Google Scholar] [CrossRef]
  95. Huang, K.; Chen, G.; Fan, W.; Hu, L. miR-23a-3p increases endometrial receptivity via CUL3 during embryo implantation. J. Mol. Endocrinol. 2020, 65, 35–44. [Google Scholar] [CrossRef] [PubMed]
  96. Li, L.; Gou, J.; Yi, T.; Li, Z.; Lin, L.; Jinhai, G.; Tao, Y.; Zhengyu, L. MicroRNA-30a-3p regulates epithelial-mesenchymal transition to affect embryo implantation by targeting Snai2. Biol. Reprod. 2019, 100, 1171–1179. [Google Scholar] [CrossRef] [PubMed]
  97. Balaguer, N.; Moreno, I.; Herrero, M.; Gonzáléz-Monfort, M.; Vilella, F.; Simón, C. MicroRNA-30d deficiency during preconception affects endometrial receptivity by decreasing implantation rates and impairing fetal growth. Am. J. Obstet. Gynecol. 2019, 221, 46.e1–46.e16. [Google Scholar] [CrossRef]
  98. Chen, C.; Zhao, Y.; Yu, Y.; Li, R.; Qiao, J. MiR-125b regulates endometrial receptivity by targeting MMP26 in women undergoing IVF-ET with elevated progesterone on HCG priming day. Sci. Rep. 2016, 6, 25302. [Google Scholar] [CrossRef] [Green Version]
  99. Kang, Y.-J.; Lees, M.; Matthews, L.C.; Kimber, S.; Forbes, K.; Aplin, J.D. miR-145 suppresses embryo-epithelial juxtacrine communication at implantation by modulating maternal IGF1R. J. Cell Sci. 2015, 128, 804–814. [Google Scholar]
  100. Liu, X.; Zhao, H.; Li, W.; Bao, H.; Qu, Q.; Ma, D. Up-regulation of miR-145 may contribute to repeated implantation failure after IVF–embryo transfer by targeting PAI-1. Reprod. Biomed. Online 2020, 40, 627–636. [Google Scholar] [CrossRef]
  101. Zheng, Q.; Zhang, D.; Cui, X.; Sun, J.; Liang, C.; Qin, H.; Yang, X.; Liu, S.; Yan, Q. MicroRNA-200c impairs uterine receptivity formation by targeting FUT4 and α1,3-fucosylation. Cell Death Differ. 2017, 24, 2161–2172. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Shi, C.; Shen, H.; Fan, L.-J.; Guan, J.; Zheng, X.-B.; Chen, X.; Liang, R.; Zhang, X.-W.; Cui, Q.-H.; Sun, K.-K.; et al. Endometrial MicroRNA Signature during the Window of Implantation Changed in Patients with Repeated Implantation Failure. Chin. Med. J. 2017, 130, 566–573. [Google Scholar] [CrossRef] [PubMed]
  103. Liang, J.; Li, K.; Chen, K.; Liang, J.; Qin, T.; He, J.; Shi, S.; Tan, Q.; Wang, Z. Regulation of ARHGAP19 in the endometrial epithelium: A possible role in the establishment of uterine receptivity. Reprod. Biol. Endocrinol. 2021, 19, 2. [Google Scholar] [CrossRef] [PubMed]
  104. Yan, Q.; Yan, G.; Zhang, C.; Wang, Z.; Huang, C.; Wang, J.; Zhou, J.; Liu, Y.; Ding, L.; Zhang, Q.; et al. miR-21 reverses impaired decidualization through modulation of KLF12 and NR4A1 expression in human endometrial stromal cells. Biol. Reprod. 2019, 100, 1395–1405. [Google Scholar] [CrossRef]
  105. Yang, Y.; Xie, Y.; Wu, M.; Geng, Y.; Li, R.; Xu, L.; Liu, X.; Pan, Y. Expression of mmu-miR-96 in the endometrium during early pregnancy and its regulatory effects on stromal cell apoptosis via Bcl2. Mol. Med. Rep. 2017, 15, 1547–1554. [Google Scholar] [CrossRef] [Green Version]
  106. Zhang, Q.; Zhang, H.; Jiang, Y.; Xue, B.; Diao, Z.; Ding, L.; Zhen, X.; Sun, H.; Yan, G.; Hu, Y. MicroRNA-181a is involved in the regulation of human endometrial stromal cell decidualization by inhibiting Krüppel-like factor 12. Reprod. Biol. Endocrinol. 2015, 13, 23. [Google Scholar] [CrossRef] [Green Version]
  107. Jimenez, P.T.; Mainigi, M.; Word, R.A.; Kraus, W.L.; Mendelson, C. miR-200 Regulates Endometrial Development During Early Pregnancy. Mol. Endocrinol. 2016, 30, 977–987. [Google Scholar] [CrossRef] [Green Version]
  108. Ma, H.L.; Gong, F.; Tang, Y.; Li, X.; Li, X.; Yang, X.; Lu, G. Inhibition of Endometrial Tiam1/Rac1 Signals Induced by miR-22 Up-Regulation Leads to the Failure of Embryo Implantation During the Implantation Window in Pregnant Mice. Biol. Reprod. 2015, 92, 152. [Google Scholar] [CrossRef]
  109. Liu, X.; Gao, R.; Chen, X.; Zhang, H.; Zheng, A.; Yang, D.; Ding, Y.; Wang, Y.; He, J. Possible Roles of mmu-miR-141 in the Endometrium of Mice in Early Pregnancy Following Embryo Implantation. PLoS ONE 2013, 8, e67382. [Google Scholar]
  110. Zhang, Q.; Ni, T.; Dang, Y.; Ding, L.; Jiang, J.; Li, J.; Xia, M.; Yu, N.; Ma, J.; Yan, J.; et al. MiR-148a-3p may contribute to flawed decidualization in recurrent implantation failure by modulating HOXC8. J. Assist. Reprod. Genet. 2020, 37, 2535–2544. [Google Scholar] [CrossRef]
  111. Graham, A.; Bs, J.H.; Nothnick, W.B. miR-181b-5p Modulates Cell Migratory Proteins, Tissue Inhibitor of Metalloproteinase 3, and Annexin A2 During In Vitro Decidualization in a Human Endometrial Stromal Cell Line. Reprod. Sci. 2016, 24, 1264–1274. [Google Scholar] [CrossRef] [PubMed]
  112. Pei, T.; Liu, C.; Liu, T.; Xiao, L.; Luo, B.; Tan, J.; Li, X.; Zhou, G.; Duan, C.; Huang, W. miR-194-3p Represses the Progesterone Receptor and Decidualization in Eutopic Endometrium from Women with Endometriosis. Endocrinology 2018, 159, 2554–2562. [Google Scholar] [CrossRef] [PubMed]
  113. Shen, L.J.; He, J.L.; Yang, D.H.; Ding, Y.B.; Chen, X.M.; Geng, Y.Q.; Liu, S.J.; Liu, X.Q.; Wang, Y.X. Mmu-microRNA-200a overexpression leads to implantation defect by targeting phosphatase and tensin homolog in mouse uterus. Reprod. Sci. 2013, 20, 1518–1528. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Haraguchi, H.; Saito-Fujita, T.; Hirota, Y.; Egashira, M.; Matsumoto, L.; Matsuo, M.; Hiraoka, T.; Koga, K.; Yamauchi, N.; Fukayama, M.; et al. MicroRNA-200a locally attenuates progesterone signaling in the cervix, preventing embryo implantation. Mol. Endocrinol. 2014, 28, 1108–1117. [Google Scholar] [CrossRef] [Green Version]
  115. Tochigi, H.; Kajihara, T.; Mizuno, Y.; Mizuno, Y.; Tamaru, S.; Kamei, Y.; Okazaki, Y.; Brosens, J.J.; Ishihara, O. Loss of MIR-542-3p enhances IGFBP-1 expression in decidualizing human endometrial stromal cells. Sci. Rep. 2017, 7, 40001. [Google Scholar] [CrossRef]
  116. Paul, A.B.M.; Sadek, S.T.; Mahesan, A.M. The role of microRNAs in human embryo implantation: A review. J. Assist. Reprod. Genet. 2018, 36, 179–187. [Google Scholar] [CrossRef]
  117. Galliano, D.; Pellicer, A. MicroRNA and implantation. Fertil. Steril. 2014, 101, 1531–1544. [Google Scholar] [CrossRef]
  118. Liu, W.; Niu, Z.; Li, Q.; Pang, R.T.; Chiu, P.C.; Yeung, W.S.B. Micro RNA and Embryo Implantation. Am. J. Reprod. Immunol. 2016, 75, 263–271. [Google Scholar] [CrossRef]
  119. Liang, J.; Wang, S.; Wang, Z. Role of microRNAs in embryo implantation. Reprod. Biol. Endocrinol. 2017, 15, 90. [Google Scholar] [CrossRef]
  120. Zhou, W.; Dimitriadis, E. Secreted MicroRNA to Predict Embryo Implantation Outcome: From Research to Clinical Diagnostic Application. Front. Cell Dev. Biol. 2020, 8, 939. [Google Scholar] [CrossRef]
  121. Cuman, C.; Van Sinderen, M.; Gantier, M.P.; Rainczuk, K.; Sorby, K.; Rombauts, L.; Osianlis, T.; Dimitriadis, E. Human Blastocyst Secreted microRNA Regulate Endometrial Epithelial Cell Adhesion. EBioMedicine 2015, 2, 1528–1535. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Xie, L.; Mouillet, J.-F.; Chu, T.; Parks, W.T.; Sadovsky, E.; Knöfler, M.; Sadovsky, Y. C19MC MicroRNAs Regulate the Migration of Human Trophoblasts. Endocrinology 2014, 155, 4975–4985. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Dai, Y.; Qiu, Z.; Diao, Z.; Shen, L.; Xue, P.; Sun, H.; Hu, Y. MicroRNA-155 inhibits proliferation and migration of human extravillous trophoblast derived HTR-8/SVneo cells via down-regulating cyclin D1. Placenta 2012, 33, 824–829. [Google Scholar] [CrossRef] [PubMed]
  124. Boo, S.H.; Kim, Y.K. The emerging role of RNA modifications in the regulation of mRNA stability. Exp. Mol. Med. 2020, 52, 400–408. [Google Scholar] [CrossRef] [Green Version]
  125. Nik Mohamed Kamal, N.N.S.B.; Shahidan, W.N.S. Non-Exosomal and Exosomal Circulatory MicroRNAs: Which Are More Valid as Biomarkers? Front. Pharmacol. 2020, 10, 1500. [Google Scholar] [CrossRef] [Green Version]
  126. Homer, H.; Rice, G.E.; Salomon, C. Review: Embryo- and endometrium-derived exosomes and their potential role in assisted reproductive treatments–liquid biopsies for endometrial receptivity. Placenta 2017, 54, 89–94. [Google Scholar] [CrossRef] [Green Version]
  127. Andronico, F.; Battaglia, R.; Ragusa, M.; Barbagallo, D.; Purrello, M.; Di Pietro, C. Extracellular Vesicles in Human Oogenesis and Implantation. Int. J. Mol. Sci. 2019, 20, 2162. [Google Scholar] [CrossRef] [Green Version]
  128. Machtinger, R.; Laurent, L.C.; Baccarelli, A.A. Extracellular vesicles: Roles in gamete maturation, fertilization and embryo implantation. Hum. Reprod. Update 2016, 22, 182–193. [Google Scholar] [CrossRef]
  129. Nguyen, H.P.T.; Simpson, R.J.; Salamonsen, L.A.; Greening, D.W. Extracellular Vesicles in the Intrauterine Environment: Challenges and Potential Functions. Biol. Reprod. 2016, 95, 109. [Google Scholar] [CrossRef]
  130. Mishra, A.; Ashary, N.; Sharma, R.; Modi, D. Extracellular vesicles in embryo implantation and disorders of the endometrium. Am. J. Reprod. Immunol. 2020, 85, e13360. [Google Scholar] [CrossRef]
  131. Chen, K.; Liang, J.; Qin, T.; Zhang, Y.; Chen, X.; Wang, Z. The Role of Extracellular Vesicles in Embryo Implantation. Front. Endocrinol. 2022, 13, 809596. [Google Scholar] [CrossRef] [PubMed]
  132. Jiang, N.-X.; Li, X.-L. The Complicated Effects of Extracellular Vesicles and Their Cargos on Embryo Implantation. Front. Endocrinol. 2021, 12, 681266. [Google Scholar] [CrossRef]
  133. Godakumara, K.; Ord, J.; Lättekivi, F.; Dissanayake, K.; Viil, J.; Boggavarapu, N.R.; Faridani, O.R.; Jääger, K.; Velthut-Meikas, A.; Jaakma, Ü.; et al. Trophoblast derived extracellular vesicles specifically alter the transcriptome of endometrial cells and may constitute a critical component of embryo-maternal communication. Reprod. Biol. Endocrinol. 2021, 19, 115. [Google Scholar] [CrossRef] [PubMed]
  134. Steptoe, P.; Edwards, R. Birth After the Reimplantation of a Human Embryo. Lancet 1978, 312, 366. [Google Scholar] [CrossRef]
  135. Edwards, R.G. An astonishing journey into reproductive genetics since the 1950’s. Reprod. Nutr. Dev. 2005, 45, 299–306. [Google Scholar] [CrossRef] [PubMed]
  136. Kamel, R.M. Assisted Reproductive Technology after the Birth of Louise Brown. J. Reprod. Infertil. 2013, 14, 96–109. [Google Scholar] [CrossRef] [Green Version]
  137. Gebert, L.F.R.; Rebhan, M.A.E.; Crivelli, S.E.M.; Denzler, R.; Stoffel, M.; Hall, J. Miravirsen (SPC3649) can inhibit the biogenesis of miR-122. Nucleic Acids Res. 2013, 42, 609–621. [Google Scholar] [CrossRef] [Green Version]
  138. Chakraborty, C.; Sharma, A.R.; Sharma, G.; Doss, C.G.P.; Lee, S.-S. Therapeutic miRNA and siRNA: Moving from Bench to Clinic as Next Generation Medicine. Mol. Ther.-Nucleic Acids 2017, 8, 132–143. [Google Scholar] [CrossRef] [Green Version]
  139. Kresowik, J.D.; Devor, E.J.; Van Voorhis, B.J.; Leslie, K.K. MicroRNA-31 is Significantly Elevated in Both Human Endometrium and Serum During the Window of Implantation: A Potential Biomarker for Optimum Receptivity1. Biol. Reprod. 2014, 91, 17. [Google Scholar] [CrossRef] [Green Version]
  140. Drissennek, L.; Baron, C.; Brouillet, S.; Entezami, F.; Hamamah, S.; Haouzi, D. Endometrial miRNome profile according to the receptivity status and implantation failure. Hum. Fertil. 2020. [Google Scholar] [CrossRef]
  141. Ghaebi, M.; Abdolmohammadi-Vahid, S.; Ahmadi, M.; Eghbal-Fard, S.; Dolati, S.; Nouri, M.; Talebi, M.; Hamdi, K.; Marofi, F.; Aghebati-Maleki, L.; et al. T cell Subsets in Peripheral Blood of Women with Recurrent Implantation Failure. J. Reprod. Immunol. 2018, 131, 21–29. [Google Scholar] [CrossRef] [PubMed]
  142. Bansal, P.; Christopher, A.F.; Kaur, R.P.; Kaur, G.; Kaur, A.; Gupta, V. MicroRNA therapeutics: Discovering novel targets and developing specific therapy. Perspect. Clin. Res. 2016, 7, 68–74. [Google Scholar] [CrossRef] [PubMed]
  143. Hanna, J.; Hossain, G.S.; Kocerha, J. The Potential for microRNA Therapeutics and Clinical Research. Front. Genet. 2019, 10, 478. [Google Scholar] [CrossRef] [Green Version]
  144. Yanokura, M.; Banno, K.; Iida, M.; Irie, H.; Umene, K.; Masuda, K.; Kobayashi, Y.; Tominaga, E.; Aoki, D. MicroRNAS in endometrial cancer: Recent advances and potential clinical applications. EXCLI J. 2015, 14, 190–198. [Google Scholar] [PubMed]
  145. Broderick, J.A.; Zamore, P.D. MicroRNA therapeutics. Gene Ther. 2011, 18, 1104–1110. [Google Scholar] [CrossRef]
  146. Rupaimoole, R.; Han, H.-D.; Lopez-Berestein, G.; Sood, A.K. MicroRNA therapeutics: Principles, expectations, and challenges. Chin. J. Cancer 2011, 30, 368–370. [Google Scholar] [CrossRef]
  147. Lanford, R.E.; Hildebrandt-Eriksen, E.S.; Petri, A.; Persson, R.; Lindow, M.; Munk, M.E.; Kauppinen, S.; Ørum, H. Therapeutic silencing of microRNA-122 in primates with chronic hepatitis C virus infection. Science 2010, 327, 198–201. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Schematic illustration of the human endometrium, its changes across the menstrual cycle and the initial process of embryo implantation. (A) The endometrium, the inner lining of the uterus, consists of the basalis and the functionalis and contains multiple cell types including luminal and glandular epithelial cells. (B) Endometrial changes across the menstrual cycle. The menstrual phase, lasting 1–5 days depending on the individual, marks the start of the cycle. The endometrium regrows in the proliferative phase and differentiates in the secretory phase. The fall in progesterone triggers the initiation of menses and the next cycle. (C) The apposition, attachment and invasion steps of embryo implantation. The embryo loosely apposes and then firmly attaches to the endometrial luminal epithelium; it then invades the stroma. Created with BioRender.com.
Figure 1. Schematic illustration of the human endometrium, its changes across the menstrual cycle and the initial process of embryo implantation. (A) The endometrium, the inner lining of the uterus, consists of the basalis and the functionalis and contains multiple cell types including luminal and glandular epithelial cells. (B) Endometrial changes across the menstrual cycle. The menstrual phase, lasting 1–5 days depending on the individual, marks the start of the cycle. The endometrium regrows in the proliferative phase and differentiates in the secretory phase. The fall in progesterone triggers the initiation of menses and the next cycle. (C) The apposition, attachment and invasion steps of embryo implantation. The embryo loosely apposes and then firmly attaches to the endometrial luminal epithelium; it then invades the stroma. Created with BioRender.com.
Ijms 23 06210 g001
Figure 2. Schematic illustration of the murine uterus and the close contact between the blastocyst and the endometrium at the start of implantation. Shown are the two horns and a cross-section of the uterus, as well as the implantation site where an embryo is in the implantation chamber on pregnancy d4.5. Created with BioRender.com.
Figure 2. Schematic illustration of the murine uterus and the close contact between the blastocyst and the endometrium at the start of implantation. Shown are the two horns and a cross-section of the uterus, as well as the implantation site where an embryo is in the implantation chamber on pregnancy d4.5. Created with BioRender.com.
Ijms 23 06210 g002
Table 2. The miRNAs identified in endometrial epithelial cells.
Table 2. The miRNAs identified in endometrial epithelial cells.
miRNASpeciesTarget Gene/ProteinImproved/Impaired
Receptivity
Ref.
let-7a/gHuman, mouse↓β-catenin, MUC1Improved[89,90]
miR-23a/bHuman, mouse↓CUL3, ↓sFRP-4Variable between subtypes[91,95]
miR-30a/b/dHuman, mouseSnai2, ↓P4HA4Variable between subtypes[81,92,96,97]
miR-125bMouse↓MMP26Impaired[98]
miR-145Human↓N-cadherin, IGF1R H2AFX, Netrin-4, ER-α, PAI-1Impaired[91,99,100,102]
miR-183Human, mouse↓CTNNA2Improved[93]
miR-192Mouse↑E-cadherin, Muc1, ↓ARHGAP19Impaired[94,103]
miR-200cMouse↓FUT4Impaired[101]
miR-429Human↓DPP4, SERPING1, AQP3Impaired[102]
miR-494Human↓CAST, CFTR, DPYSL2, F11R, FGFR2, LIF, MTF1, NPAS2, PPARGC1A, TACC2, RAB40BImpaired[81]
miR-4668Humann/aImproved[102]
miR-5088Human↓DPP4, SERPING1, AQP3Impaired[102]
Table 3. The miRNAs identified in endometrial stromal cells.
Table 3. The miRNAs identified in endometrial stromal cells.
miRNASpeciesTarget Gene/ProteinImproved/Impaired ReceptivityRef.
miR-21Human↓KLF12Improved[104]
miR-22HumanTiam/Rac1Impaired[108]
miR-96Mouse↓Bcl2, Klf13Improved[87,105]
miR-141Mouse↓PTENImpaired[109]
miR-148aHuman↓HOXC8Impaired[110]
miR-181a/bHuman↓KLF12, TIMP-3Variable between subtypes[106,111]
miR-194Human↓PRImpaired[112]
miR-200/aHuman, mouse↓ZEB1, PTEN, PGR, Bcl2Variable between subtypes[87,107,113,114]
miR-542Human↓IGFBP-1Impaired[115]
Table 4. miRNAs identified as potential serum biomarkers.
Table 4. miRNAs identified as potential serum biomarkers.
miRNASpeciesTarget Gene/ProteinImproved/Impaired ReceptivityRef.
miR-25Humann/aImpaired[141]
miR-27aHuman↓IGF1Impaired[88]
miR-31Human↓FOXP3, CXCL12Improved[139]
miR-93Humann/aImpaired[141]
miR-106bHumann/aImpaired[141]
miR-146aHuman↓STAT1Improved[141]
miR-152Humann/aImpaired[140]
miR-155Human↓SOCS1Variable between papers[140,141]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Shekibi, M.; Heng, S.; Nie, G. MicroRNAs in the Regulation of Endometrial Receptivity for Embryo Implantation. Int. J. Mol. Sci. 2022, 23, 6210. https://doi.org/10.3390/ijms23116210

AMA Style

Shekibi M, Heng S, Nie G. MicroRNAs in the Regulation of Endometrial Receptivity for Embryo Implantation. International Journal of Molecular Sciences. 2022; 23(11):6210. https://doi.org/10.3390/ijms23116210

Chicago/Turabian Style

Shekibi, Manizha, Sophea Heng, and Guiying Nie. 2022. "MicroRNAs in the Regulation of Endometrial Receptivity for Embryo Implantation" International Journal of Molecular Sciences 23, no. 11: 6210. https://doi.org/10.3390/ijms23116210

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop