Next Article in Journal
Automatic Segmentation for Favourable Delineation of Ten Wrist Bones on Wrist Radiographs Using Convolutional Neural Network
Next Article in Special Issue
Modeling Endometrium Biology and Disease
Previous Article in Journal
The Present and Future of Allergen Immunotherapy in Personalized Medicine
Previous Article in Special Issue
A Revised Stem Cell Theory for the Pathogenesis of Endometriosis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Endometrial Stem/Progenitor Cells in Recurrent Reproductive Failure

1
Centre for Women’s Health Research, Department of Women’s and Children’s Health, Institute of Life Course and Medical Sciences, University of Liverpool, Member of Liverpool Health Partners, Liverpool L8 7SS, UK
2
Hewitt Centre for Reproductive Medicine, Liverpool Women’s NHS Foundation Trust, Liverpool L8 7SS, UK
3
Liverpool Women’s NHS Foundation Trust, Member of Liverpool Health Partners, Liverpool L8 7SS, UK
*
Author to whom correspondence should be addressed.
J. Pers. Med. 2022, 12(5), 775; https://doi.org/10.3390/jpm12050775
Submission received: 20 April 2022 / Revised: 6 May 2022 / Accepted: 9 May 2022 / Published: 11 May 2022
(This article belongs to the Special Issue Endometrial Stem/Progenitor Cell Biology: Prospects and Challenges)

Abstract

:
Recurrent implantation failure (RIF) and recurrent pregnancy loss (RPL), collectively referred to as recurrent reproductive failure (RRF), are both challenging conditions with many unanswered questions relating to causes and management options. Both conditions are proposed to be related to an aberrant endometrial microenvironment, with different proposed aetiologies related to a restrictive or permissive endometrium for an invading embryo. The impressive regenerative capacity of the human endometrium has been well-established and has led to the isolation and characterisation of several subtypes of endometrial stem/progenitor cells (eSPCs). eSPCs are known to be involved in the pathogenesis of endometrium-related disorders (such as endometriosis) and have been proposed to be implicated in the pathogenesis of RRF. This review appraises the current knowledge of eSPCs, and their involvement in RRF, highlighting the considerable unknown aspects in this field, and providing avenues for future research to facilitate much-needed advances in the diagnosis and management of millions of women suffering with RRF.

1. Introduction

The endometrium is a highly dynamic, complex, and critical organ for successful human reproduction [1]. A multitude of harmonious processes allow for menstrual shedding, repair, and remodelling of the endometrium on a monthly basis, throughout a female’s reproductive lifespan, with receptivity of an embryo occurring for a finite time period in each cycle [1,2,3]. During this ‘window of implantation’ (WOI), the human endometrium, and viable blastocyst, must undergo an elaborate and tightly orchestrated crosstalk to achieve successful implantation, and for the endometrium to support an ongoing pregnancy [4].
Recurrent reproductive failure (RRF) encompasses two separate but related conditions, recurrent implantation failure (RIF), and recurrent pregnancy loss (RPL) [3,5,6,7]. RIF affects around 10% of women undergoing embryo transfers (ETs) during in vitro fertilisation (IVF) treatment [8], and the generally accepted definition is three or more failed ET attempts [9,10]. RPL has a prevalence of 1–2% [11,12], with around 50% of unexplained aetiology [13]. RPL has no universally accepted definition when referring to the number of pregnancy losses; however, the loss of either two [11,12] or three [14] consecutive pregnancies are most widely accepted. Both conditions pose major challenges for clinicians who are left with no clear definitive or effective management options, causing extreme distress for patients who may also have many unanswered questions [6,7,15]. The causes of RRF are multifactorial [16,17]; however, the proposed pathophysiological mechanisms vary according to both maternal age and gestational age when the pregnancy loss occurs [18]. Risk factors associated with both RIF and RPL include uterine abnormalities [19,20], tubal pathology [21], acquired and hereditary thrombophilia [22], endometriosis [23], suboptimal endometrial thickness [24,25], metabolic and autoimmune disorders [26,27,28], chronic infection and immunological and lifestyle factors, etc. [12,29,30,31,32,33] (Figure 1A). RIF is thought to be related to decreased endometrial receptivity [18,34,35] and RPL to an unselective or permissive endometrium with an impaired decidualisation, leading to disrupted embryo-endometrial dialogue, with a consequential lack of natural embryo selection and subsequent inappropriate implantation of a non-viable embryo [12,36,37]. Increasing evidence is now emerging that suggests a strong endometrial cause for RRF, with reports suggesting that two-thirds of RIF is secondary to inadequate endometrial receptivity [3,6,18,38], and many other studies also propose an association between unexplained RPL and a defective endometrium [3,6,17,32,39] (Figure 1B).
Endometrial glands are essential for the establishment of a pregnancy, with glandular topography and secretions integral to embryo attachment, and thus, are vital for the subsequent establishment of the decidua [40,41,42,43,44]. So far, studies of epithelial endometrial stem/progenitor cells (eSPCs) have been based on the long-accepted presumption that the human endometrial glandular architecture arise from single blunt-ended tubes within the basalis, similar to the architecture of the intestinal crypts [45,46,47,48,49]. Recent three-dimensional (3D) reconstruction of the endometrium has altered our understanding of the glandular microarchitecture, showing mycelium-like, horizontal branching networks within the deep basalis layer [48,50,51,52]. This newly discovered glandular architecture is important to consider in light of pathologies such as RRF. Endometrial glands and stroma are thought to arise and regenerate from SPC populations that are located within the relatively static basalis layer, which is undisturbed at menstrual shedding [50,53,54,55,56,57]. Aberrant eSPCs have been associated with endometrial proliferative disorders such as endometriosis and endometrial carcinoma [54,58,59,60,61]. It is possible that RRF may also arise from an abnormal glandular network, secondary to aberrant eSPCs. Dysfunctional endometrial repair may present in women with infertility, RIF, or RPL, and may be implicated in the aetiology of clinical conditions such as Asherman’s syndrome (AS) and a persistently thin endometrium [62]. AS is a condition whereby intrauterine adhesions result in a thin or defective endometrium, due to probable extensive destruction of the basalis glands (usually protected from external influence), thereby impacting the functionalis, and consequentially embryo implantation and pregnancy [48,63]. Evidence is emerging that RPL is associated with a depletion of highly proliferative mesenchymal cells that act as decidual precursors and are likely to originate from bone marrow-derived mesenchymal stem/stromal cells (BMDMSCs) [64]. The use of exogenous, autologous, or allogenic stem cells has been proposed in regenerative medicine, including the treatment of atrophic or scarred endometrium [62,65]. These have shown promise for the clinical application of SPC therapy in endometrium-related gynaecological pathologies and fertility, with reports of BMDMSCs used to improve endometrial thickness and vascularity, restore menses, and aid conception in patients with AS or endometrial atrophy [65,66,67,68,69].
Although evidence is now emerging on the possible link between non-eSPCs and RRF, little is known about the resident eSPCs and their association with these pathologies [62,70]. Evidently, much work is required in the characterisation and clinical application of eSPCs in such disorders. However, their key role in the regeneration of endometrial glands and luminal epithelium (LE) [50,71], and their role in stromal decidualisation [72,73], would suggest that they are likely to have an important function in endometrial receptivity, embryo implantation, and the support of an ongoing pregnancy.

2. Scope of This Review

This review sought to examine and summarise the current knowledge base for eSPCs and their potential involvement in RRF, to identify important voids in the literature, and to highlight avenues for future research.
Non-resident endometrial SPCs (including BMDMSCs) and non-endometrial MSCs (such as umbilical cord-derived, amniotic-derived, or adipose-derived MSCs) are other additional stem cell populations that have been previously explored in relation to endometrial regeneration [62], and lie beyond the scope of this review. Only those SPCs derived from the endometrial cell types (e.g., epithelial and stromal) will be included in this review with regards to the aetiology of RRF.

3. Materials and Methods

An extensive literature search was performed across multiple databases, including PubMed, Web of Science, EMBASE, and Scopus, and spanned publications from inception to February 2022. Studies were selected using keywords associated with eSPCs, RRF, RIF, and RPL. Additionally, all references cited within other relevant publications were screened. Publications related to all keywords were included, and this included both human and animal studies.

4. Endometrial Stem/Progenitor Cells

The remarkable regenerative capacity of the endometrium is widely believed to arise from SPCs residing within the deeper basalis layer, which remains intact following menstruation and the menopause [48,54,56,57,74,75,76,77,78]. The functionalis layer can re-grow from a thickness of 1–2 mm, following menstruation, to 14 mm during the secretory phase of the cycle [79]. Remarkable restoration is also seen following parturition, iatrogenic surgical destruction of the endometrium (such as endometrial ablation), and within the post-menopausal endometrium following exposure to oestrogen hormone replacement therapy [77,80,81,82]. eSPCs are thought to be the key players in driving this impressive proliferative capacity and cell turnover [48,54,56,57,83,84].
Since the existence of endometrial adult stem cells (ASCs) was first postulated [74,75], much work has been invested into identifying and characterising eSPCs in both human endometrium and in mouse models [50,53,54,71,85,86,87,88,89,90,91,92,93,94,95,96]. In 2004, rare populations of clonogenic epithelial and stromal cells were first reported [53]. Identifying and examining human eSPCs is challenging due to their scarcity within the tissue, their lack of specific markers allowing for their isolation, and their change in phenotype when taken out of their highly unique endometrial microenvironment [57,97]. Studies attempting to identify these cells have mainly explored their distinguishing functional properties, such as their clonogenic ability, self-renewal capacity, proliferation and differentiation capacity, label retention, and tissue reconstitution assays [50,54,56,71,87,88,89,90,98,99,100,101].
Work identifying endometrial stromal ASCs (also known as endometrial mesenchymal stem cells (eMSCs)) was first initiated by Gargett and colleagues, who examined cells displaying higher clonogenic colony-forming capacity in vitro [53]. Endometrial stromal ASCs are seen to show similar properties to BMDMSCs and are multipotent, displaying the capability to differentiate into fat, bone, cartilage, skeletal muscle, and smooth muscle [100,101,102,103]. Endometrial stromal ASCs have been well characterised, with CD146+ platelet-derived growth factor receptor beta (PDGFRβ+) [93,102] and sushi domain containing-2 (SUSD2+), as specific markers for their enrichment [96]. More recently, nucleoside triphosphate diphosphohydrolase 2 (NTPDase2) has been detected in just the perivascular SUSD2+ cells and not the rest of the stromal fraction, and thus has been proposed as a marker for eMSCs located in the endometrial basal layer [104]. SUSD2+ cells (also referred to as W5C5+) are found to increase during the proliferative phase, suggesting their involvement in regeneration of the functional stroma [96]. They are also seen within postmenopausal endometrium treated with oestrogen, and reconstitute endometrium under the kidney capsule of xenografted mice [96]. Their location is now widely accepted to be within the pericyte and perivascular cells of the endometrial basalis and functionalis, with transcriptomics and secretomics of SUSD2+ cells confirming their perivascular phenotype [56,93,96,104].
Unlike endometrial stromal ASCs, epithelial eSPCs are yet to be conclusively defined, with specific universal markers remaining elusive [57]. Work focused on characterising the epithelial eSPC population was first initiated within our laboratories, with the breakthrough discovery of cell surface marker stage-specific embryonic antigen-1 (SSEA-1) to demarcate the basalis epithelial cells [54]. The proposed SSEA-1+ epithelial eSPCs show higher telomerase activity and longer telomere lengths, a greater propensity to generate spheroids in 3D cell culture, and low expression of steroid hormone receptors [54,61]. Subsequent work has also suggested N-cadherin to be an epithelial eSPC marker. It enriches for clonogenic epithelial cells showing greater self-renewal capacity and quiescence, with the ability to generate large gland-like spheroids [105] (Figure 2). These characteristics correlate with a more primitive or SPC phenotype. With in vivo immunostaining of SSEA-1+ and N-cadherin+ endometrial epithelial cells in full thickness endometrium, Nguyen et al. have proposed that there is a spatial relationship between the two cell types, with SSEA-1+ cells located closer to the basalis–functionalis interface, whilst N-cadherin+ cells situated deeper in the basalis. With this observation, the authors proposed a possible differentiation hierarchy amongst the epithelial eSPCs [105]. Prior to these findings, Musashi-1 (an ASC marker) had also been immunolocalised to single epithelial cells within endometrial glands, and small clusters of stromal cells [94]. Musashi-1+ cells were found mainly within the basalis of the proliferative phase endometrium, supporting its possible SPC role; however, no functional data is available for these cells, thus the evidence for Musashi-1 as an eSPC marker is limited [94]. Nuclear WNT signalling pathway molecules, axis inhibition protein 2 (AXIN2) [106,107], SRY-box 9 (SOX9) [54], and nuclear ß-catenin [54] are also seen to be expressed by basalis glands; however, nuclear markers are not useful for the prospective isolation of epithelial eSPCs for important functional assessment, and therefore surface markers for their identification are required. More recently, leucine-rich repeat-containing G protein-coupled receptor 5 (LGR5) (a surface marker for intestinal epithelial stem cells) has been demonstrated to be expressed by a subset of epithelial cells within the LE, as well as in the glands of the basalis layer. However, due to the lack of reliable antibodies for immunology-based sorting, functional data for these cells are not yet available [71]. The proposed current understanding of eSPCs is summarised within the pictorial representation below (Figure 2).
eSPCs have been postulated to be involved in the pathogenesis of proliferative gynaecological disorders such as endometriosis [61,74,84,108,109]. Studies have demonstrated the expression of proposed epithelial stem cell markers SSEA-1 and nuclear SOX9, nuclear ß-catenin, and Musashi-1 in ectopic endometriotic lesions [54,61,94], which has shed light on the pathogenesis of the disease [61]. Endometriosis is associated with infertility [110], suggesting that the presence of atypical endometrial ASCs may be a possible common aetiology between these disorders, resulting in abnormal eutopic endometrium, as well as giving rise to the ectopic endometriotic lesions in endometriosis patients. In addition to the proliferative disorders of the endometrium, it is therefore likely that other disorders, associated with aberrant regeneration and remodelling of the functional layer of the endometrium, including infertility, RIF, and RPL [70], arise from abnormalities within the endometrial ASC population. Similar detailed examination of the involvement of eSPCs in RRF is expected to enhance our understanding of the pathogenesis of the disease.

5. Endometrial Stem/Progenitor Cells and Recurrent Reproductive Failure (RRF)

Two studies have identified the role of eSPCs in RRF (RIF and RPL), with current knowledge limited to eMSCs. To date, eMSCs have been suggested to play a role in endometrial receptivity and regeneration, both of which are essential to successful pregnancy.

5.1. Recurrent Implantation Failure (RIF)

Considering the well-established endometrial contribution to RIF, eSPC abnormalities are expected to be involved in generating a persistently abnormal functionalis layer. Unlike in RPL, the endometrium of women with RIF is proposed to be hostile and less receptive to embryo-attachment [18,34,35]. Studies have suggested that eMSCs express markers of endometrial receptivity, such as adhesion molecules. Endometrial stromal cells are known to express receptivity markers HoxA11 and Noggin during the WOI [111]. In 2020, five markers of receptivity (ITGβ1 [112], RAC1 [113], HOXA11 [111], ITGβ3 [112,114], and NOGGIN [111]) were compared between menstrual blood samples taken from women with RIF, versus fertile controls, revealing different patterns of expression. This observation led the authors to propose that aberrant eMSCs contribute to altered endometrial receptivity in women with RIF [115]. eMSCs from menstrual blood were isolated and characterised based on their morphology and behaviour, expression of specific surface markers (CD44, CD31, CD34, CD73, CD90, and CD105), and capacity to differentiate, all of which was based on the International Society for Cellular Therapy statement [115,116]. Notably, significantly higher expression levels of both HOXA11 and ITGβ3 were seen in the eMSCs of women with RIF [115]. HOXA11 encodes a transcription factor and is involved in proliferation, differentiation, and embryologic development of the endometrium [117], therefore, HOXA11 is thought to be important in establishing pregnancy [111,118]. Rac1 expression is necessary for eMSC migration and is associated with increased cell motility at the site of implantation [113]. Integrins are adhesive molecules also highly expressed by MSCs and were found to play a role in embryo implantation and development [39,112,114,119]. Integrins play a vital role in extracellular matrix adhesion and therefore are vital in embryo implantation [112,120], with integrin β3 proposed as a useful tool to predict the success rate of assisted reproductive technology (ART) [112]. In agreement with this manuscript, recent work from another group also identified an increased expression of HOXA11 antisense RNA in the endometrium of women with RIF, leading to downregulated decidualisation [121]. Contradictory to this, Hoxa11 knockout mice are known to be associated with uterine factor infertility [117]. As for integrin β3, the available evidence is conflicting: higher expression has been shown in eMSC from RIF endometrial tissue samples [115], but, in a larger study, low expression in endometrial tissue has been associated with lower pregnancy rates [122]. It is apparent that HOXA11 and integrin β3 contribute to uterine receptivity and are therefore relevant to RIF. Hoxa11 is expressed in mouse uterine stromal cells during the receptive phase and during post-implantation decidualisation, suggesting a possible role in receptivity, implantation, and decidualisation [118]. Hoxa11-/- mice have hypoplastic uteri, fewer endometrial glands, and do not express Lif, which is essential for the progression of a normal pregnancy [111,123]. However, the mechanisms behind how specific eSPCs contribute to the observed abnormal endometrial HOXA11 and integrin β3 levels, remains undefined. The results from Esmaeilzadeh et al. (the only study concentrating on eMSCs and RIF) should be interpreted with caution as the number of women who participated in the study was very small (n = 5) and the control group utilised (n = 3) was extremely heterogenous, almost overlapping with the RIF group (history of less than three miscarriages and at least one live birth after ART cycle(s) (two of the controls had a child after their second ART cycle and the other after their third cycle) [115]. In addition, the technique used to isolate and identify the eMSCs is questionable as they did not use the previously characterised eMSC markers (CD146+PDGFRβ+, SUSD2+, or NTPDase2), raising concerns regarding the appropriateness of the methodology and reliability of only eMSCs being isolated.

5.2. Recurrent Pregnancy Loss (RPL)

Similarly to RIF, aberrant endometrium is expected to contribute to the pathology of RPL. RPL is proposed to be associated with a hyper-receptive endometrium, permissive to even a sub-optimal blastocyst [124], with possible eSPC aberrations in the LE and functionalis, which are the first layers of contact for an incoming embryo [48]. To explore the eMSC population in women with RPL, Lucas et al. (2016) isolated perivascular and non-perivascular human endometrial stromal cells (W5C5+ and W5C5, respectively) from luteinising hormone (LH) timed mid-luteal phase endometrial biopsies [125]. W5C5 was identified in 2012 as a single marker capable of purifying eMSCs possessing MSC properties and reconstituting endometrial stromal tissues in vivo [96]. No differences in the number of W5C5+ perivascular cells between the women with RPL and control samples were noted. However, there was a significant decrease in the clonogenicity of W5C5+ and W5C5 cell populations from RPL samples when compared to fertile controls. No clonogenic W5C5 cells were recovered from 42% of the RPL sample versus 11% of the control samples. In the cohort studied, the presence of clonogenic W5C5+ or W5C5 cells were adversely associated with the number of previous miscarriages [125]. This means that controlling an eMSC deficiency could be considered a possible approach to prevent RPL. However, the contribution of the epithelial eSPCs to RPL has not yet been examined.
The discovery that a deficiency of eMSCs is connected to RPL has revealed new insights into pregnancy failure mechanisms. This has posed new questions concerning the pathways that control the maintenance of eMSCs from one pregnancy to another pregnancy.

6. Applications in Clinical Management of RRF

eSPCs have been found to play a crucial role in the regeneration and repair of the endometrium [54,126,127]. We therefore would envisage eSPCs to be involved in the persistent aberrations in the functionalis layer of each successive cycle, associated with the implantation process and subsequent pregnancy establishment in RRF. However, the available evidence for detailed knowledge from basic science studies on functional or phenotypic abnormalities of these specialised cells is still lacking to guide their safe clinical translation. It is therefore unsurprising that trials of eSPC therapies for women with RRF are currently not available. Therefore, studies are urgently required to explore the fundamental role of eSPCs in the pathogenesis of RIF and RPL, as well as to assess their therapeutic potential in the management of these conditions. Furthermore, common and challenging clinical conditions that are related to RRF include thin endometrium and AS, for which studies have been undertaken to aid clinical management with the use of eSPCs [62,126].
Persistently thin endometrium is a major challenge within the fertility setting [128,129,130,131], exemplified by the findings that, with each millimetre of decrease in endometrial thickness under 8 mm, a significant decrease in clinical pregnancy and live birth rates and dramatically increased pregnancy loss rates after achieving a clinical pregnancy occur [132,133,134]. The causes of persistently thin endometrium are unknown, with no studies to report if the deficiency is of the functionalis or basalis, epithelial eSPC or eMSC. However, the malfunctioning of eSPCs is postulated to have a role [135], and many studies exist targeting SPCs as a possible treatment avenue [136,137,138,139,140]. Exogenous oestradiol supplementation and various adjuvant therapies have been proposed to promote endometrial regeneration (such as aspirin, sildenafil, tamoxifen, vitamin E, pentoxifylline, L-arginine, or platelet-rich plasma) presumably by influencing eSPC activity; however, they all lack robust evidence and definitive efficacy for their proposed use [24,141]. A thin endometrium is known to be associated with RRF [24] and it may be idiopathic in nature [142,143] or possibly associated with inflammatory/iatrogenic causes such as intrauterine adhesions after a surgical procedure [24]. Post-surgical thin endometrium may be due to the removal of the stem cell rich basalis during extensive curettage, or following ablation [135]. The use of eSPCs to enhance regeneration of the thin endometrium can be expected to provide a novel solution to the ongoing dilemma of ‘thin endometrium’ by encouraging more efficient regrowth. We postulate that the eSPC would provide a stock of eSPCs able to integrate into the endometrium, thus increasing the resident eSPC pool. Alternatively, it may provide the appropriate niche for the existing eSPCs to function more efficiently, with the desired end point being the availability of the appropriate bulk of functional eSPCs, and provision of a sufficient amount of their progeny, on a monthly basis. In 2019, Hu et al. investigated the effect of menstrual blood-derived stem cells (MenSCs) on endometrial repair following mechanical injury in mice [144]. They found that mice treated with MenSC exhibited significantly higher expression of endometrial keratin, vimentin, and vascular endothelial growth factor, with an increased endometrial thickness, and increased pregnancy rates [144]. This was potentially due to the MenSC providing a better scaffold for cells with improved blood supply, cellular integrity, and resistance to cell damage. This evidence supports the notion that MenSC therapy improves the eSPC niche. Zhao et al. and Jing et al. also demonstrated that injecting BMDMSCs into the endometrial cavities of rats, resulted in increased endometrial thickness [137,145]. Injecting primitive MenSC or BMDSC cells seem to be beneficial with an intriguing mechanism of action that is yet to be clarified. We could hypothesise that the injected cells cause activation of quiescent resident eSPCs, enhancing the important support from the stem cell niche, or they may promote recruitment of new SPCs to the endometrium, rectifying a postulated deficient eSPC pool.
Currently, treatment for AS is usually surgical, with hysteroscopic adhesiolysis [146,147]. In severe cases, however, surgical treatments often fail, requiring multiple repeat hysteroscopic procedures, a 29% risk of recurrence, and a low post-treatment pregnancy rate of a mere 25% in these severely affected cases [148,149]. In 2021, the phenotypic and biological characteristics of eMSCs (co-expression of CD140b and CD146) from healthy fertile women, and women with intrauterine adhesions was compared [150]. The number of eMSCs in women with intrauterine adhesions was reported to be significantly lower compared with healthy fertile women [150]. Furthermore, the colony-forming capacity, migration, invasion, angiogenic/capillary formation, and immunosuppressive abilities of the eMSCs was also significantly lower, possibly affecting their ability to migrate and regenerate at the site of injury [150]. No evidence is available regarding the loss of eSPC stem cell niche being a reason for the deficient endometrium, and additional work needs to be done on this plausible theory. Although Min et al.’s study showed a significant decrease in the number and biological function of eMSCs in women with intrauterine adhesions, whether these disorders can be treated by eMSC supplementation requires further validation. Due to the main features of AS being the lack of basalis stem cell niche and fibrotic adhesions between the opposing uterine myometrial surfaces, the number of cells available to be harvested from AS women are likely to be low. Since the functional assays used in the experiments presented in the above study are directly relevant to the number of cells that can be harvested and used in in vitro assays, the expected reduction in number of cells harvested from AS endometrium could directly affect the assay data. In 2016, Zhang et al. applied human eMSCs derived from menstrual blood into mouse models. The authors concluded that stem cells derived from menstrual blood may have a role in repairing the damaged endometrium, which could be due to the engagement of angiogenesis mediated by stem cells derived from menstrual blood [151]. Therapy with autologous eMSCs derived from menstrual blood has also shown promise, accelerating the healing process of damaged endometrium by inducing self-renewal, differentiation, angiogenesis, anti-inflammation, and immunomodulation [127]. Numerous issues must, however, be addressed before treatments can be introduced to clinical practice, such as transplanted cell dosage, and administration route.
RPL has been associated with the loss of eMSCs [125]. In 2020, Tewary et al. conducted a randomised, double-blind, placebo-controlled feasibility trial, administering either sitagliptin or placebo to women with RPL, proposing to target BMDMSCs and eMSCs [152]. Sitagliptin is a dipeptidyl peptidase 4 (DPP4) inhibitor, which may encourage the migration and engraftment of BMDMSCs to the site of tissue injury. DPP4 inhibitors are commonly used oral hypoglycaemic medications, which work by blocking the action of DDP4, thereby increasing concentrations of active incretin hormones, and improving glycaemic control [153,154]. They have been seen to promote tissue regeneration following injury [152,155]. Their aim was to determine the impact of sitagliptin on the abundance of eMSCs and on endometrial decidualisation in women with RPL. By using colony forming unit assays, the authors concluded that sitagliptin increases eMSCs and decreases decidual senescence in women with RPL during the mid-luteal phase [152]. Decidual cells, but not senescent decidual cells, are required for a continuing pregnancy, and are dependent on continuous progesterone signalling, therefore, the decreased decidual senescence would lead to increased promotion of decidualisation by progesterone, successful implantation, and potentially an increased ability to maintain a pregnancy. Although further work is required, these findings have shown promise that eSPCs could be a potential target for treatment in women with RRF. The available evidence that endometrial plasticity can be pharmacologically enhanced through improving the number of eMSCs is encouraging.

7. Avenues for Future Research

Although we strongly believe that eSPCs have a critical role to play in the pathology of RRF, evidence for direct eSPCs involvement in RRF is currently lacking. Available data is limited, sometimes contradictory, and of varying quality. Furthermore, the lack of clarity on the precise phenotypical and functional properties, origin, and location of eSPCs contributing to normal endometrial regeneration, hinders progress in the identification of eSPC aberration, specific to RRF, and thus, impedes the advancement of potential eSPC-based therapeutic avenues.
The importance of understanding the critical role of the endometrium in the implantation and success of ongoing pregnancy is acutely apparent by the knowledge that only 50% of euploid embryos accomplish implantation [156]. In addition, 1–2% of women who do conceive (either spontaneously or using ART methods) are found to experience an unexplained loss of a karyotypically normal early pregnancy [12].
Currently, the majority of research exploring stem cells in unexplained infertility, RRF, or endometrial regenerative medicine is mainly restricted to non-endometrial stem cells [62]. This primarily includes MSCs derived from the bone marrow [67,68,157,158], umbilical cord [140,159,160,161], placenta [162,163,164,165,166], or adipose tissue [167,168,169,170]. Very limited data exists on eMSCs and their relationship to RRF, with no current evidence for the role of human epithelial eSPCs in this devastating condition. Whilst it is important to understand the role of all types of stem cells within the endometrium of women with RRF, it is essential to understand the role of resident eSPCs from which the two primary endometrial cell types arise [100]. Further work is required to investigate the role of human stromal and epithelial eSPCs in both RIF and RPL, to aid in diagnosis and provide translatable treatment options.
The LE (the endometrial layer that communicates first with the incoming blastocyst) and endometrial glands (which provide an abundance of secretions to enable successful implantation) have been proven to be essential for pregnancy establishment [40,41]. The LE and endometrial glands are both thought to originate from epithelial eSPCs within the basalis layer [50,54,57,71]. More work is evidently required to identify and characterise the epithelial eSPC population within the normal endometrium of fertile women, which will allow, in the future, further study on their, possibly, aberrant role in women with RIF and RPL. If/when robust epithelial eSPC surface markers can be identified, a possibility to selectively isolate these eSPCs, and use them in diagnostic and therapeutic technologies, would become apparent. Our new understanding of the endometrial glandular microarchitecture [48,50,51,52] supports the theory that primitive, quiescent eSPCs reside in the mycelium-like basalis glands and give rise to the functional glands that grow vertically from these branches. Characterisation and localisation of epithelial eSPCs should be re-considered and re-examined in light of this new discovery. It may be that patients who experience RIF and RPL have an aberrant or underdeveloped horizontal network of basalis glands, and therefore an abnormal eSPC population, leading to defective regeneration of the functional endometrium and LE [48,51,52].
There are many technical and ethical challenges to overcome in defining the intricacies of human embryo implantation, and there are still many aspects that are not fully understood. Much of our knowledge of embryo implantation has stemmed from animal studies and in vitro two-dimensional (2D) monolayer culture systems, neither of which accurately represent the in vivo endometrial or embryo physiology, or environment [48,171]. 3D investigation of the human endometrium and embryo implantation is an important avenue for future research [48,51,52]. The advent of ‘organoids’ has led to huge advances in in vitro 3D modelling techniques [171,172,173]. Endometrial organoids arise from single or small clusters of cells and resemble endometrial tissue ex vivo [174]. They are grown in a supportive hydrogel droplet (such as Matrigel), which acts to mimic the extracellular matrix in vivo, alongside complex growth media designed to recapitulate the endometrial niche [172,174]. This 3D culture system has been successfully adapted to derive embryo-like organoids (known as blastoids) [175,176,177], trophoblast organoids modelling placental development [178,179], and gland-like organoids originating from endometrial epithelial cells [54,172,180,181,182,183]. Future research can utilise these 3D modelling systems in order to study endometrial glandular development and embryo implantation in patients with RIF and RPL.

8. Discussion

RIF and RPL are devastating and challenging conditions, with many unanswered questions remaining. Evidence is now starting to emerge that eSPCs may be implicated in the aetiology of these conditions, giving rise to an abnormal endometrium, and therefore a suboptimal environment to support an ongoing pregnancy. Advances in endometrial stem cell biology and novel seminal discoveries, such as the 3D histoarchitecture of human endometrial glands [50,51], have allowed us to start to identify and isolate different eSPC populations. More work is, however, required in order to fully characterise them in vivo and establish the stem cell hierarchy. In particular, new highlights on the 3D microarchitecture of the human endometrium have provided novel insights into the accurate structure and arrangement of the endometrial glands [48,50,51,52]. This may provide new insight and explanations for the location, mechanisms and possible aberrations of endometrium-associated conditions, such as RRF.

Author Contributions

Conceptualisation of the manuscript by H.A.-L., N.T., and D.K.H.; H.A.-L., N.T., C.J.H., F.T., T.P., A.J.D., and D.K.H. wrote, edited, and reviewed the manuscript; C.J.H. prepared the figures. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by Wellbeing of Women (grant number RTF510, RG1073, and RG2137). H.A.-L was supported by The Hewitt Fertility Centre/Liverpool Women’s Hospital NHS Trust. N.T was supported by an Academic Clinical Lectureship from the National Institute of Health Research.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Acknowledgments

Authors would like to acknowledge Lisa Xue at the Royal College of Obstetricians and Gynaecologists who kindly assisted with accessing the literature for this review.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Okada, H.; Tsuzuki, T.; Murata, H. Decidualization of the Human Endometrium. Reprod. Med. Biol. 2018, 17, 220–227. [Google Scholar] [CrossRef] [Green Version]
  2. Diedrich, K.; Fauser, B.C.J.M.; Devroey, P.; Griesinger, G.; on behalf of the Evian Annual Reproduction (EVAR) Workshop Group. The Role of the Endometrium and Embryo in Human Implantation. Hum. Reprod. Update 2007, 13, 365–377. [Google Scholar] [CrossRef]
  3. Hapangama, D.K.; Turner, M.A.; Drury, J.A.; Martin-Ruiz, C.; Von Zglinicki, T.; Farquharson, R.G.; Quenby, S. Endometrial Telomerase Shows Specific Expression Patterns in Different Types of Reproductive Failure. Reprod. Biomed. Online 2008, 17, 416–424. [Google Scholar] [CrossRef]
  4. Kim, S.-M.; Kim, J.-S. A Review of Mechanisms of Implantation. Dev. Reprod. 2017, 21, 351–359. [Google Scholar] [CrossRef] [Green Version]
  5. Quenby, S.; Nik, H.; Innes, B.; Lash, G.; Turner, M.; Drury, J.; Bulmer, J. Uterine Natural Killer Cells and Angiogenesis in Recurrent Reproductive Failure. Hum. Reprod. 2009, 24, 45–54. [Google Scholar] [CrossRef] [Green Version]
  6. Tempest, N.; Batchelor, E.; Hill, C.J.; Al-Lamee, H.; Drury, J.; Drakeley, A.J.; Hapangama, D.K. Anterior Gradient Protein 3 and S100 Calcium-Binding Protein P Levels in Different Endometrial Epithelial Compartments May Play an Important Role in Recurrent Pregnancy Failure. Int. J. Mol. Sci. 2021, 22, 3835. [Google Scholar] [CrossRef]
  7. Al-Lamee, H.; Ellison, A.; Drury, J.; Hill, C.J.; Drakeley, A.J.; Hapangama, D.K.; Tempest, N. Altered Endometrial Oestrogen-Responsiveness and Recurrent Reproductive Failure. Reprod. Fertil. 2022, 3, 30–38. [Google Scholar] [CrossRef]
  8. Busnelli, A.; Reschini, M.; Cardellicchio, L.; Vegetti, W.; Somigliana, E.; Vercellini, P. How Common Is Real Repeated Implantation Failure? An Indirect Estimate of the Prevalence. Reprod. Biomed. Online 2020, 40, 91–97. [Google Scholar] [CrossRef] [Green Version]
  9. Busnelli, A.; Somigliana, E.; Cirillo, F.; Baggiani, A.; Levi-Setti, P.E. Efficacy of Therapies and Interventions for Repeated Embryo Implantation Failure: A Systematic Review and Meta-Analysis. Sci. Rep. 2021, 11, 1747. [Google Scholar] [CrossRef]
  10. Polanski, L.T.; Baumgarten, M.N.; Quenby, S.; Brosens, J.; Campbell, B.K.; Raine-Fenning, N.J. What Exactly Do We Mean by “Recurrent Implantation Failure”? A Systematic Review and Opinion. Reprod. Biomed. Online 2014, 28, 409–423. [Google Scholar] [CrossRef] [Green Version]
  11. Practice Committee of the American Society for Reproductive Medicine. Definitions of Infertility and Recurrent Pregnancy Loss: A Committee Opinion. Fertil. Steril. 2013, 99, 63. [Google Scholar] [CrossRef]
  12. ESHRE Guideline Group on RPL; Bender Atik, R.; Christiansen, O.B.; Elson, J.; Kolte, A.M.; Lewis, S.; Middeldorp, S.; Nelen, W.; Peramo, B.; Quenby, S.; et al. ESHRE Guideline: Recurrent Pregnancy Loss. Hum. Reprod. Open 2018, 2018, hoy004. [Google Scholar] [CrossRef]
  13. El Hachem, H.; Crepaux, V.; May-Panloup, P.; Descamps, P.; Legendre, G.; Bouet, P.-E. Recurrent Pregnancy Loss: Current Perspectives. Int. J. Women’s Health 2017, 9, 331–345. [Google Scholar] [CrossRef] [Green Version]
  14. Royal College of Obstetritians and Gynaecologists (RCOG). The Investigation and Treatment of Couples with Recurrent First-Trimester and Second-Trimester Miscarriage; RCOG: London, UK, 2011. [Google Scholar]
  15. Farren, J.; Jalmbrant, M.; Falconieri, N.; Mitchell-Jones, N.; Bobdiwala, S.; Al-Memar, M.; Tapp, S.; Van Calster, B.; Wynants, L.; Timmerman, D.; et al. Posttraumatic Stress, Anxiety and Depression Following Miscarriage and Ectopic Pregnancy: A Multicenter, Prospective, Cohort Study. Am. J. Obstet. Gynecol. 2020, 222, 367.e1–367.e22. [Google Scholar] [CrossRef]
  16. Margalioth, E.J.; Ben-Chetrit, A.; Gal, M.; Eldar-Geva, T. Investigation and Treatment of Repeated Implantation Failure Following IVF-ET. Hum. Reprod. 2006, 21, 3036–3043. [Google Scholar] [CrossRef] [Green Version]
  17. Dimitriadis, E.; Menkhorst, E.; Saito, S.; Kutteh, W.H.; Brosens, J.J. Recurrent Pregnancy Loss. Nat. Rev. Dis. Primers 2020, 6, 98. [Google Scholar] [CrossRef]
  18. Bashiri, A.; Halper, K.I.; Orvieto, R. Recurrent Implantation Failure-Update Overview on Etiology, Diagnosis, Treatment and Future Directions. Reprod. Biol. Endocrinol. 2018, 16, 121. [Google Scholar] [CrossRef] [Green Version]
  19. Demirol, A.; Gurgan, T. Effect of Treatment of Intrauterine Pathologies with Office Hysteroscopy in Patients with Recurrent IVF Failure. Reprod. Biomed. Online 2004, 8, 590–594. [Google Scholar] [CrossRef]
  20. Turocy, J.M.; Rackow, B.W. Uterine Factor in Recurrent Pregnancy Loss. Semin. Perinatol. 2019, 43, 74–79. [Google Scholar] [CrossRef]
  21. Meyer, W.R.; Castelbaum, A.J.; Somkuti, S.; Sagoskin, A.W.; Doyle, M.; Harris, J.E.; Lessey, B.A. Hydrosalpinges Adversely Affect Markers of Endometrial Receptivity. Hum. Reprod. 1997, 12, 1393–1398. [Google Scholar] [CrossRef] [Green Version]
  22. Backos, M.; Rai, R.; Regan, L. Antiphospholipid Antibodies and Infertility. Hum. Fertil. 2002, 5, 30–34. [Google Scholar] [CrossRef]
  23. Pirtea, P.; Cicinelli, E.; De Nola, R.; de Ziegler, D.; Ayoubi, J.M. Endometrial Causes of Recurrent Pregnancy Losses: Endometriosis, Adenomyosis, and Chronic Endometritis. Fertil. Steril. 2021, 115, 546–560. [Google Scholar] [CrossRef]
  24. Mahajan, N.; Sharma, S. The Endometrium in Assisted Reproductive Technology: How Thin Is Thin? J. Hum. Reprod. Sci. 2016, 9, 3–8. [Google Scholar] [CrossRef]
  25. Vartanyan, E.; Tsaturova, K.; Devyatova, E. Thin Endometrium Problem in IVF Programs. Gynecol. Endocrinol. 2020, 36, 24–27. [Google Scholar] [CrossRef]
  26. Bellver, J.; Soares, S.R.; Alvarez, C.; Muñoz, E.; Ramírez, A.; Rubio, C.; Serra, V.; Remohí, J.; Pellicer, A. The Role of Thrombophilia and Thyroid Autoimmunity in Unexplained Infertility, Implantation Failure and Recurrent Spontaneous Abortion. Hum. Reprod. 2008, 23, 278–284. [Google Scholar] [CrossRef] [Green Version]
  27. Alecsandru, D.; Barrio, A.; Andia, V.; Cruz, E.; Aparicio, P.; Serna, J.; Cruz, M.; Pellicer, A.; Garcia-Velasco, J.A. Pancreatic Autoimmunity: An Unknown Etiology on Patients with Assisted Reproductive Techniques (ART)-Recurrent Reproductive Failure. PLoS ONE 2018, 13, e0203446. [Google Scholar] [CrossRef]
  28. Sheikhansari, G.; Soltani-Zangbar, M.S.; Pourmoghadam, Z.; Kamrani, A.; Azizi, R.; Aghebati-Maleki, L.; Danaii, S.; Koushaeian, L.; Hojat-Farsangi, M.; Yousefi, M. Oxidative Stress, Inflammatory Settings, and MicroRNA Regulation in the Recurrent Implantation Failure Patients with Metabolic Syndrome. Am. J. Reprod. Immunol. 2019, 82, e13170. [Google Scholar] [CrossRef]
  29. Broughton, D.E.; Moley, K.H. Obesity and Female Infertility: Potential Mediators of Obesity’s Impact. Fertil. Steril. 2017, 107, 840–847. [Google Scholar] [CrossRef] [Green Version]
  30. Vitagliano, A.; Saccardi, C.; Noventa, M.; Di Spiezio Sardo, A.; Saccone, G.; Cicinelli, E.; Pizzi, S.; Andrisani, A.; Litta, P.S. Effects of Chronic Endometritis Therapy on In Vitro Fertilization Outcome in Women with Repeated Implantation Failure: A Systematic Review and Meta-Analysis. Fertil. Steril. 2018, 110, 103–112.e1. [Google Scholar] [CrossRef]
  31. Odendaal, J.; Quenby, S.; Sammaritano, L.; Macklon, N.; Branch, D.W.; Rosenwaks, Z. Immunologic and Rheumatologic Causes and Treatment of Recurrent Pregnancy Loss: What Is the Evidence? Fertil. Steril. 2019, 112, 1002–1012. [Google Scholar] [CrossRef] [Green Version]
  32. Ticconi, C.; Pietropolli, A.; Di Simone, N.; Piccione, E.; Fazleabas, A. Endometrial Immune Dysfunction in Recurrent Pregnancy Loss. Int. J. Mol. Sci. 2019, 20, 5332. [Google Scholar] [CrossRef] [Green Version]
  33. Vomstein, K.; Voss, P.; Molnar, K.; Ainsworth, A.; Daniel, V.; Strowitzki, T.; Toth, B.; Kuon, R.-J. Two of a Kind? Immunological and Clinical Risk Factors Differ between Recurrent Implantation Failure and Recurrent Miscarriage. J. Reprod. Immunol. 2020, 141, 103166. [Google Scholar] [CrossRef] [PubMed]
  34. Valdes, C.T.; Schutt, A.; Simon, C. Implantation Failure of Endometrial Origin: It Is Not Pathology, but Our Failure to Synchronize the Developing Embryo with a Receptive Endometrium. Fertil. Steril. 2017, 108, 15–18. [Google Scholar] [CrossRef] [Green Version]
  35. Santamaria, X.; Simón, C. Endometrial Factor in Unexplained Infertility and Recurrent Implantation Failure. Semin. Reprod. Med. 2021, 39, 227–232. [Google Scholar] [CrossRef] [PubMed]
  36. Salker, M.; Teklenburg, G.; Molokhia, M.; Lavery, S.; Trew, G.; Aojanepong, T.; Mardon, H.J.; Lokugamage, A.U.; Rai, R.; Landles, C.; et al. Natural Selection of Human Embryos: Impaired Decidualization of Endometrium Disables Embryo-Maternal Interactions and Causes Recurrent Pregnancy Loss. PLoS ONE 2010, 5, e10287. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Teklenburg, G.; Salker, M.; Heijnen, C.; Macklon, N.S.; Brosens, J.J. The Molecular Basis of Recurrent Pregnancy Loss: Impaired Natural Embryo Selection. Mol. Hum. Reprod. 2010, 16, 886–895. [Google Scholar] [CrossRef] [Green Version]
  38. Simon, A.; Laufer, N. Repeated Implantation Failure: Clinical Approach. Fertil. Steril. 2012, 97, 1039–1043. [Google Scholar] [CrossRef]
  39. Quenby, S.; Anim-Somuah, M.; Kalumbi, C.; Farquharson, R.; Aplin, J.D. Different Types of Recurrent Miscarriage Are Associated with Varying Patterns of Adhesion Molecule Expression in Endometrium. Reprod. Biomed. Online 2007, 14, 224–234. [Google Scholar] [CrossRef]
  40. Yuan, J.; Cha, J.; Deng, W.; Bartos, A.; Sun, X.; Ho, H.-Y.H.; Borg, J.-P.; Yamaguchi, T.P.; Yang, Y.; Dey, S.K. Planar Cell Polarity Signaling in the Uterus Directs Appropriate Positioning of the Crypt for Embryo Implantation. Proc. Natl. Acad. Sci. USA 2016, 113, E8079–E8088. [Google Scholar] [CrossRef] [Green Version]
  41. Yuan, J.; Deng, W.; Cha, J.; Sun, X.; Borg, J.-P.; Dey, S.K. Tridimensional Visualization Reveals Direct Communication between the Embryo and Glands Critical for Implantation. Nat. Commun. 2018, 9, 603. [Google Scholar] [CrossRef]
  42. Salamonsen, L.A.; Evans, J.; Nguyen, H.P.T.; Edgell, T.A. The Microenvironment of Human Implantation: Determinant of Reproductive Success. Am. J. Reprod. Immunol. 2016, 75, 218–225. [Google Scholar] [CrossRef] [PubMed]
  43. Filant, J.; Spencer, T.E. Uterine Glands: Biological Roles in Conceptus Implantation, Uterine Receptivity and Decidualization. Int. J. Dev. Biol. 2014, 58, 107–116. [Google Scholar] [CrossRef] [PubMed]
  44. Kelleher, A.M.; Milano-Foster, J.; Behura, S.K.; Spencer, T.E. Uterine Glands Coordinate On-Time Embryo Implantation and Impact Endometrial Decidualization for Pregnancy Success. Nat. Commun. 2018, 9, 2435. [Google Scholar] [CrossRef]
  45. Nogales, F.; Martinez, H.; Parache, J. Shedding and reconstruction of the human endometrium. Gynakol. Rundsch. 1969, 7, 292–312. [Google Scholar] [CrossRef] [PubMed]
  46. Nogales-Ortiz, F.; Puerta, J.; Nogales, F.F. The Normal Menstrual Cycle. Chronology and Mechanism of Endometrial Desquamation. Obstet. Gynecol. 1978, 51, 259–264. [Google Scholar] [CrossRef] [PubMed]
  47. Ferenczy, A. Studies on the Cytodynamics of Human Endometrial Regeneration. I. Scanning Electron Microscopy. Am. J. Obstet. Gynecol. 1976, 124, 64–74. [Google Scholar] [CrossRef]
  48. Tempest, N.; Hill, C.J.; Maclean, A.; Marston, K.; Powell, S.G.; Al-Lamee, H.; Hapangama, D.K. Novel Microarchitecture of Human Endometrial Glands: Implications in Endometrial Regeneration and Pathologies. Hum. Reprod. Update 2022, 28, 153–171. [Google Scholar] [CrossRef]
  49. Kurman, R.J.; Ellenson, L.H.; Ronnett, B.M. Blaustein’s Pathology of the Female Genital Tract; Springer: Cham, Switzerland, 2019; ISBN 978-3-319-46333-9. [Google Scholar]
  50. Tempest, N.; Jansen, M.; Baker, A.-M.; Hill, C.J.; Hale, M.; Magee, D.; Treanor, D.; Wright, N.A.; Hapangama, D.K. Histological 3D Reconstruction and in Vivo Lineage Tracing of the Human Endometrium. J. Pathol. 2020, 251, 440–451. [Google Scholar] [CrossRef]
  51. Yamaguchi, M.; Yoshihara, K.; Suda, K.; Nakaoka, H.; Yachida, N.; Ueda, H.; Sugino, K.; Mori, Y.; Yamawaki, K.; Tamura, R.; et al. Three-Dimensional Understanding of the Morphological Complexity of the Human Uterine Endometrium. iScience 2021, 24, 102258. [Google Scholar] [CrossRef]
  52. Yamaguchi, M.; Yoshihara, K.; Yachida, N.; Suda, K.; Tamura, R.; Ishiguro, T.; Enomoto, T. The New Era of Three-Dimensional Histoarchitecture of the Human Endometrium. J. Pers. Med. 2021, 11, 713. [Google Scholar] [CrossRef]
  53. Chan, R.W.S.; Schwab, K.E.; Gargett, C.E. Clonogenicity of Human Endometrial Epithelial and Stromal Cells. Biol. Reprod. 2004, 70, 1738–1750. [Google Scholar] [CrossRef] [PubMed]
  54. Valentijn, A.J.; Palial, K.; Al-Lamee, H.; Tempest, N.; Drury, J.; Von Zglinicki, T.; Saretzki, G.; Murray, P.; Gargett, C.E.; Hapangama, D.K. SSEA-1 Isolates Human Endometrial Basal Glandular Epithelial Cells: Phenotypic and Functional Characterization and Implications in the Pathogenesis of Endometriosis. Hum. Reprod. 2013, 28, 2695–2708. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Fayazi, M.; Salehnia, M.; Ziaei, S. Characteristics of Human Endometrial Stem Cells in Tissue and Isolated Cultured Cells: An Immunohistochemical Aspect. Iran. Biomed. J. 2016, 20, 109–116. [Google Scholar] [CrossRef] [PubMed]
  56. Gargett, C.E.; Schwab, K.E.; Deane, J.A. Endometrial Stem/Progenitor Cells: The First 10 Years. Hum. Reprod. Update 2016, 22, 137–163. [Google Scholar] [CrossRef] [Green Version]
  57. Tempest, N.; Maclean, A.; Hapangama, D.K. Endometrial Stem Cell Markers: Current Concepts and Unresolved Questions. Int. J. Mol. Sci. 2018, 19, 3240. [Google Scholar] [CrossRef] [Green Version]
  58. Hapangama, D.K.; Turner, M.A.; Drury, J.A.; Quenby, S.; Saretzki, G.; Martin-Ruiz, C.; Von Zglinicki, T. Endometriosis Is Associated with Aberrant Endometrial Expression of Telomerase and Increased Telomere Length. Hum. Reprod. 2008, 23, 1511–1519. [Google Scholar] [CrossRef] [Green Version]
  59. Kyo, S.; Maida, Y.; Inoue, M. Stem Cells in Endometrium and Endometrial Cancer: Accumulating Evidence and Unresolved Questions. Cancer Lett. 2011, 308, 123–133. [Google Scholar] [CrossRef]
  60. Cousins, F.L.; Dorien, F.O.; Gargett, C.E. Endometrial Stem/Progenitor Cells and Their Role in the Pathogenesis of Endometriosis. Best Pract. Res. Clin. Obstet. Gynaecol. 2018, 50, 27–38. [Google Scholar] [CrossRef]
  61. Hapangama, D.K.; Drury, J.; Da Silva, L.; Al-Lamee, H.; Earp, A.; Valentijn, A.J.; Edirisinghe, D.P.; Murray, P.A.; Fazleabas, A.T.; Gargett, C.E. Abnormally Located SSEA1+/SOX9+ Endometrial Epithelial Cells with a Basalis-like Phenotype in the Eutopic Functionalis Layer May Play a Role in the Pathogenesis of Endometriosis. Hum. Reprod. 2019, 34, 56–68. [Google Scholar] [CrossRef]
  62. Strug, M.; Aghajanova, L. Making More Womb: Clinical Perspectives Supporting the Development and Utilization of Mesenchymal Stem Cell Therapy for Endometrial Regeneration and Infertility. J. Pers. Med. 2021, 11, 1364. [Google Scholar] [CrossRef]
  63. Dreisler, E.; Kjer, J.J. Asherman’s Syndrome: Current Perspectives on Diagnosis and Management. Int. J. Women’s Health 2019, 11, 191–198. [Google Scholar] [CrossRef] [PubMed]
  64. Diniz-da-Costa, M.; Kong, C.-S.; Fishwick, K.J.; Rawlings, T.; Brighton, P.J.; Hawkes, A.; Odendaal, J.; Quenby, S.; Ott, S.; Lucas, E.S.; et al. Characterization of Highly Proliferative Decidual Precursor Cells during the Window of Implantation in Human Endometrium. Stem Cells 2021, 39, 1067–1080. [Google Scholar] [CrossRef] [PubMed]
  65. De Miguel-Gómez, L.; Ferrero, H.; López-Martínez, S.; Campo, H.; López-Pérez, N.; Faus, A.; Hervás, D.; Santamaría, X.; Pellicer, A.; Cervelló, I. Stem Cell Paracrine Actions in Tissue Regeneration and Potential Therapeutic Effect in Human Endometrium: A Retrospective Study. BJOG Int. J. Obstet. Gynaecol. 2020, 127, 551–560. [Google Scholar] [CrossRef] [PubMed]
  66. Alawadhi, F.; Du, H.; Cakmak, H.; Taylor, H.S. Bone Marrow-Derived Stem Cell (BMDSC) Transplantation Improves Fertility in a Murine Model of Asherman’s Syndrome. PLoS ONE 2014, 9, e96662. [Google Scholar] [CrossRef] [PubMed]
  67. Singh, N.; Mohanty, S.; Seth, T.; Shankar, M.; Bhaskaran, S.; Dharmendra, S. Autologous Stem Cell Transplantation in Refractory Asherman’s Syndrome: A Novel Cell Based Therapy. J. Hum. Reprod. Sci. 2014, 7, 93–98. [Google Scholar] [CrossRef]
  68. Singh, N.; Shekhar, B.; Mohanty, S.; Kumar, S.; Seth, T.; Girish, B. Autologous Bone Marrow-Derived Stem Cell Therapy for Asherman’s Syndrome and Endometrial Atrophy: A 5-Year Follow-up Study. J. Hum. Reprod. Sci. 2020, 13, 31–37. [Google Scholar] [CrossRef]
  69. Yi, K.W.; Mamillapalli, R.; Sahin, C.; Song, J.; Tal, R.; Taylor, H.S. Bone Marrow-Derived Cells or C-X-C Motif Chemokine 12 (CXCL12) Treatment Improve Thin Endometrium in a Mouse Model. Biol. Reprod. 2019, 100, 61–70. [Google Scholar] [CrossRef]
  70. Lucas, E.S.; Dyer, N.P.; Fishwick, K.; Ott, S.; Brosens, J.J. Success after Failure: The Role of Endometrial Stem Cells in Recurrent Miscarriage. Reproduction 2016, 152, R159–R166. [Google Scholar] [CrossRef] [Green Version]
  71. Tempest, N.; Baker, A.M.; Wright, N.A.; Hapangama, D.K. Does Human Endometrial LGR5 Gene Expression Suggest the Existence of Another Hormonally Regulated Epithelial Stem Cell Niche? Hum. Reprod. 2018, 33, 1052–1062. [Google Scholar] [CrossRef] [Green Version]
  72. Valatkaitė, E.; Baušytė, R.; Vitkevičienė, A.; Ramašauskaitė, D.; Navakauskienė, R. Decidualization Potency and Epigenetic Changes in Human Endometrial Origin Stem Cells During Propagation. Front. Cell Dev. Biol. 2021, 9, 765265. [Google Scholar] [CrossRef]
  73. Gorsek Sparovec, T.; Markert, U.R.; Reif, P.; Schoell, W.; Moser, G.; Feichtinger, J.; Mihalic, Z.N.; Kargl, J.; Gargett, C.E.; Gold, D. The Fate of Human SUSD2+ Endometrial Mesenchymal Stem Cells during Decidualization. Stem Cell Res. 2022, 60, 102671. [Google Scholar] [CrossRef] [PubMed]
  74. Papanicolaou, G.N. Epithelial Regeneration in the Uterine Glands and on the Surface of the Uterus. Am. J. Obstet. Gynecol. 1933, 25, 30–37. [Google Scholar] [CrossRef]
  75. Prianishnikov, V.A. A Functional Model of the Structure of the Epithelium of Normal, Hyperplastic and Malignant Human Endometrium: A Review. Gynecol. Oncol. 1978, 6, 420–428. [Google Scholar] [CrossRef]
  76. Padykula, H.A.; Coles, L.G.; Okulicz, W.C.; Rapaport, S.I.; McCracken, J.A.; King, N.W.; Longcope, C.; Kaiserman-Abramof, I.R. The Basalis of the Primate Endometrium: A Bifunctional Germinal Compartment. Biol. Reprod. 1989, 40, 681–690. [Google Scholar] [CrossRef] [Green Version]
  77. Gargett, C.E. Uterine Stem Cells: What Is the Evidence? Hum. Reprod. Update 2007, 13, 87–101. [Google Scholar] [CrossRef]
  78. Cousins, F.L.; Pandoy, R.; Jin, S.; Gargett, C.E. The Elusive Endometrial Epithelial Stem/Progenitor Cells. Front. Cell Dev. Biol. 2021, 9, 868. [Google Scholar] [CrossRef]
  79. Fleischer, A.C. Sonographic Assessment of Endometrial Disorders. Semin. Ultrasound CT MRI 1999, 20, 259–266. [Google Scholar] [CrossRef]
  80. Tresserra, F.; Grases, P.; Ubeda, A.; Pascual, M.A.; Grases, P.J.; Labastida, R. Morphological Changes in Hysterectomies after Endometrial Ablation. Hum. Reprod. 1999, 14, 1473–1477. [Google Scholar] [CrossRef] [Green Version]
  81. Gargett, C.E. Identification and Characterisation of Human Endometrial Stem/Progenitor Cells. Aust. N. Z. J. Obstet. Gynaecol. 2006, 46, 250–253. [Google Scholar] [CrossRef]
  82. Hapangama, D.K.; Kamal, A.M.; Bulmer, J.N. Estrogen Receptor β: The Guardian of the Endometrium. Hum. Reprod. Update 2015, 21, 174–193. [Google Scholar] [CrossRef] [Green Version]
  83. Gargett, C.E.; Chan, R.W.S.; Schwab, K.E. Endometrial Stem Cells. Curr. Opin. Obstet. Gynecol. 2007, 19, 377–383. [Google Scholar] [CrossRef] [PubMed]
  84. Sourial, S.; Tempest, N.; Hapangama, D.K. Theories on the Pathogenesis of Endometriosis. Int. J. Reprod. Med. 2014, 2014, 179515. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Taylor, H.S. Endometrial Cells Derived from Donor Stem Cells in Bone Marrow Transplant Recipients. JAMA 2004, 292, 81–85. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Schwab, K.E.; Chan, R.W.S.; Gargett, C.E. Putative Stem Cell Activity of Human Endometrial Epithelial and Stromal Cells during the Menstrual Cycle. Fertil. Steril. 2005, 84 (Suppl. S2), 1124–1130. [Google Scholar] [CrossRef] [PubMed]
  87. Chan, R.W.S.; Gargett, C.E. Identification of Label-Retaining Cells in Mouse Endometrium. Stem Cells 2006, 24, 1529–1538. [Google Scholar] [CrossRef]
  88. Cervelló, I.; Martínez-Conejero, J.A.; Horcajadas, J.A.; Pellicer, A.; Simón, C. Identification, Characterization and Co-Localization of Label-Retaining Cell Population in Mouse Endometrium with Typical Undifferentiated Markers. Hum. Reprod. 2007, 22, 45–51. [Google Scholar] [CrossRef] [Green Version]
  89. Cervelló, I.; Gil-Sanchis, C.; Mas, A.; Delgado-Rosas, F.; Martínez-Conejero, J.A.; Galán, A.; Martínez-Romero, A.; Martínez, S.; Navarro, I.; Ferro, J.; et al. Human Endometrial Side Population Cells Exhibit Genotypic, Phenotypic and Functional Features of Somatic Stem Cells. PLoS ONE 2010, 5, e10964. [Google Scholar] [CrossRef] [Green Version]
  90. Cervelló, I.; Mas, A.; Gil-Sanchis, C.; Peris, L.; Faus, A.; Saunders, P.T.K.; Critchley, H.O.D.; Simón, C. Reconstruction of Endometrium from Human Endometrial Side Population Cell Lines. PLoS ONE 2011, 6, e21221. [Google Scholar] [CrossRef]
  91. Du, H.; Taylor, H.S. Contribution of Bone Marrow-Derived Stem Cells to Endometrium and Endometriosis. Stem Cells 2007, 25, 2082–2086. [Google Scholar] [CrossRef]
  92. Kato, K.; Yoshimoto, M.; Kato, K.; Adachi, S.; Yamayoshi, A.; Arima, T.; Asanoma, K.; Kyo, S.; Nakahata, T.; Wake, N. Characterization of Side-Population Cells in Human Normal Endometrium. Hum. Reprod. 2007, 22, 1214–1223. [Google Scholar] [CrossRef] [Green Version]
  93. Schwab, K.E.; Gargett, C.E. Co-Expression of Two Perivascular Cell Markers Isolates Mesenchymal Stem-like Cells from Human Endometrium. Hum. Reprod. 2007, 22, 2903–2911. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Götte, M.; Wolf, M.; Staebler, A.; Buchweitz, O.; Kelsch, R.; Schüring, A.N.; Kiesel, L. Increased Expression of the Adult Stem Cell Marker Musashi-1 in Endometriosis and Endometrial Carcinoma. J. Pathol. 2008, 215, 317–329. [Google Scholar] [CrossRef] [PubMed]
  95. Du, H.; Naqvi, H.; Taylor, H.S. Ischemia/Reperfusion Injury Promotes and Granulocyte-Colony Stimulating Factor Inhibits Migration of Bone Marrow-Derived Stem Cells to Endometrium. Stem Cells Dev. 2012, 21, 3324–3331. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Masuda, H.; Anwar, S.S.; Bühring, H.-J.; Rao, J.R.; Gargett, C.E. A Novel Marker of Human Endometrial Mesenchymal Stem-like Cells. Cell Transplant. 2012, 21, 2201–2214. [Google Scholar] [CrossRef]
  97. Kim, J.Y.; Tavaré, S.; Shibata, D. Counting Human Somatic Cell Replications: Methylation Mirrors Endometrial Stem Cell Divisions. Proc. Natl. Acad. Sci. USA 2005, 102, 17739–17744. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Potten, C.S.; Loeffler, M. Stem Cells: Attributes, Cycles, Spirals, Pitfalls and Uncertainties. Lessons for and from the Crypt. Development 1990, 110, 1001–1020. [Google Scholar] [CrossRef]
  99. Gargett, C.E. Review Article: Stem Cells in Human Reproduction. Reprod. Sci. 2007, 14, 405–424. [Google Scholar] [CrossRef]
  100. Gargett, C.E.; Schwab, K.E.; Zillwood, R.M.; Nguyen, H.P.T.; Wu, D. Isolation and Culture of Epithelial Progenitors and Mesenchymal Stem Cells from Human Endometrium. Biol. Reprod. 2009, 80, 1136–1145. [Google Scholar] [CrossRef] [Green Version]
  101. Fayazi, M.; Salehnia, M.; Ziaei, S. Differentiation of Human CD146-Positive Endometrial Stem Cells to Adipogenic-, Osteogenic-, Neural Progenitor-, and Glial-like Cells. In Vitro Cell. Dev. Biol.-Anim. 2015, 51, 408–414. [Google Scholar] [CrossRef]
  102. Azami, M.; Ai, J.; Ebrahimi-Barough, S.; Farokhi, M.; Fard, S.E. In Vitro Evaluation of Biomimetic Nanocomposite Scaffold Using Endometrial Stem Cell Derived Osteoblast-like Cells. Tissue Cell 2013, 45, 328–337. [Google Scholar] [CrossRef]
  103. Shoae-Hassani, A.; Sharif, S.; Seifalian, A.M.; Mortazavi-Tabatabaei, S.A.; Rezaie, S.; Verdi, J. Endometrial Stem Cell Differentiation into Smooth Muscle Cell: A Novel Approach for Bladder Tissue Engineering in Women. BJU Int. 2013, 112, 854–863. [Google Scholar] [CrossRef] [PubMed]
  104. Trapero, C.; Vidal, A.; Rodríguez-Martínez, A.; Sévigny, J.; Ponce, J.; Coroleu, B.; Matias-Guiu, X.; Martín-Satué, M. The Ectonucleoside Triphosphate Diphosphohydrolase-2 (NTPDase2) in Human Endometrium: A Novel Marker of Basal Stroma and Mesenchymal Stem Cells. Purinergic Signal. 2019, 15, 225–236. [Google Scholar] [CrossRef] [PubMed]
  105. Nguyen, H.P.T.; Xiao, L.; Deane, J.A.; Tan, K.-S.; Cousins, F.L.; Masuda, H.; Sprung, C.N.; Rosamilia, A.; Gargett, C.E. N-Cadherin Identifies Human Endometrial Epithelial Progenitor Cells by In Vitro Stem Cell Assays. Hum. Reprod. 2017, 32, 2254–2268. [Google Scholar] [CrossRef] [Green Version]
  106. Nguyen, H.P.T.; Sprung, C.N.; Gargett, C.E. Differential Expression of Wnt Signaling Molecules between Pre- and Postmenopausal Endometrial Epithelial Cells Suggests a Population of Putative Epithelial Stem/Progenitor Cells Reside in the Basalis Layer. Endocrinology 2012, 153, 2870–2883. [Google Scholar] [CrossRef]
  107. Syed, S.M.; Kumar, M.; Ghosh, A.; Tomasetig, F.; Ali, A.; Whan, R.M.; Alterman, D.; Tanwar, P.S. Endometrial Axin2+ Cells Drive Epithelial Homeostasis, Regeneration, and Cancer Following Oncogenic Transformation. Cell Stem Cell 2020, 26, 64–80.e13. [Google Scholar] [CrossRef]
  108. Sampson, J.A. Metastatic or Embolic Endometriosis, Due to the Menstrual Dissemination of Endometrial Tissue into the Venous Circulation. Am. J. Pathol. 1927, 3, 93–110.43. [Google Scholar]
  109. Leyendecker, G.; Herbertz, M.; Kunz, G.; Mall, G. Endometriosis Results from the Dislocation of Basal Endometrium. Hum. Reprod. 2002, 17, 2725–2736. [Google Scholar] [CrossRef] [Green Version]
  110. Macer, M.L.; Taylor, H.S. Endometriosis and Infertility: A Review of the Pathogenesis and Treatment of Endometriosis-Associated Infertility. Obstet. Gynecol. Clin. N. Am. 2012, 39, 535–549. [Google Scholar] [CrossRef] [Green Version]
  111. Wang, H.; Dey, S.K. Roadmap to Embryo Implantation: Clues from Mouse Models. Nat. Rev. Genet. 2006, 7, 185–199. [Google Scholar] [CrossRef]
  112. Achache, H.; Revel, A. Endometrial Receptivity Markers, the Journey to Successful Embryo Implantation. Hum. Reprod. Update 2006, 12, 731–746. [Google Scholar] [CrossRef] [Green Version]
  113. Grewal, S.; Carver, J.G.; Ridley, A.J.; Mardon, H.J. Implantation of the Human Embryo Requires Rac1-Dependent Endometrial Stromal Cell Migration. Proc. Natl. Acad. Sci. USA 2008, 105, 16189–16194. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Lessey, B.A.; Castelbaum, A.J.; Sawin, S.W.; Sun, J. Integrins as Markers of Uterine Receptivity in Women with Primary Unexplained Infertility. Fertil. Steril. 1995, 63, 535–542. [Google Scholar] [CrossRef]
  115. Esmaeilzadeh, S.; Mohammadi, A.; Mahdinejad, N.; Ghofrani, F.; Ghasemzadeh-Hasankolaei, M. Receptivity Markers in Endometrial Mesenchymal Stem Cells of Recurrent Implantation Failure and Non-Recurrent Implantation Failure Women: A Pilot Study. J. Obstet. Gynaecol. Res. 2020, 46, 1393–1402. [Google Scholar] [CrossRef]
  116. Dominici, M.; Le Blanc, K.; Mueller, I.; Slaper-Cortenbach, I.; Marini, F.; Krause, D.; Deans, R.; Keating, A.; Prockop, D.; Horwitz, E. Minimal Criteria for Defining Multipotent Mesenchymal Stromal Cells. The International Society for Cellular Therapy Position Statement. Cytotherapy 2006, 8, 315–317. [Google Scholar] [CrossRef] [PubMed]
  117. Chau, Y.M.; Pando, S.; Taylor, H.S. HOXA11 Silencing and Endogenous HOXA11 Antisense Ribonucleic Acid in the Uterine Endometrium. J. Clin. Endocrinol. Metab. 2002, 87, 2674–2680. [Google Scholar] [CrossRef] [PubMed]
  118. He, B.; Ni, Z.-L.; Kong, S.-B.; Lu, J.-H.; Wang, H.-B. Homeobox Genes for Embryo Implantation: From Mouse to Human. Anim. Models Exp. Med. 2018, 1, 14–22. [Google Scholar] [CrossRef] [Green Version]
  119. Docheva, D.; Popov, C.; Mutschler, W.; Schieker, M. Human Mesenchymal Stem Cells in Contact with Their Environment: Surface Characteristics and the Integrin System. J. Cell. Mol. Med. 2007, 11, 21–38. [Google Scholar] [CrossRef]
  120. Merviel, P.; Challier, J.C.; Carbillon, L.; Foidart, J.M.; Uzan, S. The Role of Integrins in Human Embryo Implantation. Fetal Diagn. Ther. 2001, 16, 364–371. [Google Scholar] [CrossRef]
  121. Zhao, H.; Hu, S.; Qi, J.; Wang, Y.; Ding, Y.; Zhu, Q.; He, Y.; Lu, Y.; Yao, Y.; Wang, S.; et al. Increased Expression of HOXA11-AS Attenuates Endometrial Decidualization in Recurrent Implantation Failure Patients. Mol. Ther. 2022, 30, 1706–1720. [Google Scholar] [CrossRef]
  122. Revel, A.; Koler, M.; Prus, D.; Tsafrir, A.; Laufer, N.; Reich, R. Implementation of Integrin Β3 Level as Predictor of Implantation in an IVF Program. Fertil. Steril. 2005, 84, S144. [Google Scholar] [CrossRef]
  123. Gendron, R.L.; Paradis, H.; Hsieh-Li, H.M.; Lee, D.W.; Potter, S.S.; Markoff, E. Abnormal Uterine Stromal and Glandular Function Associated with Maternal Reproductive Defects in Hoxa-11 Null Mice. Biol. Reprod. 1997, 56, 1097–1105. [Google Scholar] [CrossRef] [PubMed]
  124. Quenby, S.; Vince, G.; Farquharson, R.; Aplin, J. Recurrent Miscarriage: A Defect in Nature’s Quality Control? Hum. Reprod. 2002, 17, 1959–1963. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Lucas, E.S.; Dyer, N.P.; Murakami, K.; Lee, Y.H.; Chan, Y.-W.; Grimaldi, G.; Muter, J.; Brighton, P.J.; Moore, J.D.; Patel, G.; et al. Loss of Endometrial Plasticity in Recurrent Pregnancy Loss. Stem Cells 2016, 34, 346–356. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Gargett, C.E.; Nguyen, H.P.T.; Ye, L. Endometrial Regeneration and Endometrial Stem/Progenitor Cells. Rev. Endocr. Metab. Disord. 2012, 13, 235–251. [Google Scholar] [CrossRef]
  127. He, Y.; Han, Y.; Ye, Y. Therapeutic Potential of Menstrual Blood-Derived Stem Cell Transplantation for Intrauterine Adhesions. Front. Surg. 2022, 9, 847213. [Google Scholar] [CrossRef]
  128. Lebovitz, O.; Orvieto, R. Treating Patients with “Thin” Endometrium—An Ongoing Challenge. Gynecol. Endocrinol. 2014, 30, 409–414. [Google Scholar] [CrossRef]
  129. Timeva, T.; Shterev, A.; Kyurkchiev, S. Recurrent Implantation Failure: The Role of the Endometrium. J. Reprod. Infertil. 2014, 15, 173–183. [Google Scholar]
  130. Liu, S.-M.; Zhou, Y.-Z.; Wang, H.-B.; Sun, Z.-Y.; Zhen, J.-R.; Shen, K.; Deng, C.-Y.; Lang, J.-H. Factors Associated with Effectiveness of Treatment and Reproductive Outcomes in Patients with Thin Endometrium Undergoing Estrogen Treatment. Chin. Med. J. 2015, 128, 3173–3177. [Google Scholar] [CrossRef]
  131. Liu, K.E.; Hartman, M.; Hartman, A. Management of Thin Endometrium in Assisted Reproduction: A Clinical Practice Guideline from the Canadian Fertility and Andrology Society. Reprod. Biomed. Online 2019, 39, 49–62. [Google Scholar] [CrossRef] [Green Version]
  132. Vaegter, K.K.; Lakic, T.G.; Olovsson, M.; Berglund, L.; Brodin, T.; Holte, J. Which Factors Are Most Predictive for Live Birth after In Vitro Fertilization and Intracytoplasmic Sperm Injection (IVF/ICSI) Treatments? Analysis of 100 Prospectively Recorded Variables in 8400 IVF/ICSI Single-Embryo Transfers. Fertil. Steril. 2017, 107, 641–648.e2. [Google Scholar] [CrossRef] [Green Version]
  133. Gallos, I.D.; Khairy, M.; Chu, J.; Rajkhowa, M.; Tobias, A.; Campbell, A.; Dowell, K.; Fishel, S.; Coomarasamy, A. Optimal Endometrial Thickness to Maximize Live Births and Minimize Pregnancy Losses: Analysis of 25,767 Fresh Embryo Transfers. Reprod. Biomed. Online 2018, 37, 542–548. [Google Scholar] [CrossRef] [PubMed]
  134. Liu, K.E.; Hartman, M.; Hartman, A.; Luo, Z.-C.; Mahutte, N. The Impact of a Thin Endometrial Lining on Fresh and Frozen-Thaw IVF Outcomes: An Analysis of over 40,000 Embryo Transfers. Hum. Reprod. 2018, 33, 1883–1888. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Gharibeh, N.; Aghebati-Maleki, L.; Madani, J.; Pourakbari, R.; Yousefi, M.; Ahmadian Heris, J. Cell-Based Therapy in Thin Endometrium and Asherman Syndrome. Stem Cell Res. Ther. 2022, 13, 33. [Google Scholar] [CrossRef] [PubMed]
  136. Deane, J.A.; Gualano, R.C.; Gargett, C.E. Regenerating Endometrium from Stem/Progenitor Cells: Is It Abnormal in Endometriosis, Asherman’s Syndrome and Infertility? Curr. Opin. Obstet. Gynecol. 2013, 25, 193–200. [Google Scholar] [CrossRef] [PubMed]
  137. Jing, Z.; Qiong, Z.; Yonggang, W.; Yanping, L. Rat Bone Marrow Mesenchymal Stem Cells Improve Regeneration of Thin Endometrium in Rat. Fertil. Steril. 2014, 101, 587–594. [Google Scholar] [CrossRef] [PubMed]
  138. Azizi, R.; Aghebati-Maleki, L.; Nouri, M.; Marofi, F.; Negargar, S.; Yousefi, M. Stem Cell Therapy in Asherman Syndrome and Thin Endometrium: Stem Cell- Based Therapy. Biomed. Pharmacother. 2018, 102, 333–343. [Google Scholar] [CrossRef]
  139. Abuwala, N.; Tal, R. Endometrial Stem Cells: Origin, Biological Function, and Therapeutic Applications for Reproductive Disorders. Curr. Opin. Obstet. Gynecol. 2021, 33, 232–240. [Google Scholar] [CrossRef]
  140. Zhang, Y.; Shi, L.; Lin, X.; Zhou, F.; Xin, L.; Xu, W.; Yu, H.; Li, J.; Pan, M.; Pan, Y.; et al. Unresponsive Thin Endometrium Caused by Asherman Syndrome Treated with Umbilical Cord Mesenchymal Stem Cells on Collagen Scaffolds: A Pilot Study. Stem Cell Res. Ther. 2021, 12, 420. [Google Scholar] [CrossRef]
  141. Mouanness, M.; Ali-Bynom, S.; Jackman, J.; Seckin, S.; Merhi, Z. Use of Intra-Uterine Injection of Platelet-Rich Plasma (PRP) for Endometrial Receptivity and Thickness: A Literature Review of the Mechanisms of Action. Reprod. Sci. 2021, 28, 1659–1670. [Google Scholar] [CrossRef]
  142. Strohmer, H.; Obruca, A.; Radner, K.M.; Feichtinger, W. Relationship of the Individual Uterine Size and the Endometrial Thickness in Stimulated Cycles. Fertil. Steril. 1994, 61, 972–975. [Google Scholar] [CrossRef]
  143. Scioscia, M.; Lamanna, G.; Lorusso, F.; Serrati, G.; Selvaggi, L.E.; Depalo, R. Characterization of Endometrial Growth in Proliferative and Early Luteal Phase in IVF Cycles. Reprod. Biomed. Online 2009, 18, 73–78. [Google Scholar] [CrossRef]
  144. Hu, J.; Song, K.; Zhang, J.; Zhang, Y.; Tan, B.-Z. Effects of Menstrual Blood-Derived Stem Cells on Endometrial Injury Repair. Mol. Med. Rep. 2019, 19, 813–820. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Zhao, J.; Zhang, Q.; Wang, Y.; Li, Y. Uterine Infusion with Bone Marrow Mesenchymal Stem Cells Improves Endometrium Thickness in a Rat Model of Thin Endometrium. Reprod. Sci. 2015, 22, 181–188. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Conforti, A.; Alviggi, C.; Mollo, A.; De Placido, G.; Magos, A. The Management of Asherman Syndrome: A Review of Literature. Reprod. Biol. Endocrinol. 2013, 11, 118. [Google Scholar] [CrossRef] [Green Version]
  147. Khan, Z.; Goldberg, J.M. Hysteroscopic Management of Asherman’s Syndrome. J. Minim. Invasive Gynecol. 2018, 25, 218–228. [Google Scholar] [CrossRef]
  148. Hanstede, M.M.F.; van der Meij, E.; Goedemans, L.; Emanuel, M.H. Results of Centralized Asherman Surgery, 2003–2013. Fertil. Steril. 2015, 104, 1561–1568.e1. [Google Scholar] [CrossRef] [Green Version]
  149. Chen, L.; Zhang, H.; Wang, Q.; Xie, F.; Gao, S.; Song, Y.; Dong, J.; Feng, H.; Xie, K.; Sui, L. Reproductive Outcomes in Patients With Intrauterine Adhesions Following Hysteroscopic Adhesiolysis: Experience From the Largest Women’s Hospital in China. J. Minim. Invasive Gynecol. 2017, 24, 299–304. [Google Scholar] [CrossRef]
  150. Min, J.; Lu, N.; Huang, S.; Chai, X.; Wang, S.; Peng, L.; Wang, J. Phenotype and Biological Characteristics of Endometrial Mesenchymal Stem/Stromal Cells: A Comparison between Intrauterine Adhesion Patients and Healthy Women. Am. J. Reprod. Immunol. 2021, 85, e13379. [Google Scholar] [CrossRef]
  151. Zhang, Y.; Lin, X.; Dai, Y.; Hu, X.; Zhu, H.; Jiang, Y.; Zhang, S. Endometrial Stem Cells Repair Injured Endometrium and Induce Angiogenesis via AKT and ERK Pathways. Reproduction 2016, 152, 389–402. [Google Scholar] [CrossRef] [Green Version]
  152. Tewary, S.; Lucas, E.S.; Fujihara, R.; Kimani, P.K.; Polanco, A.; Brighton, P.J.; Muter, J.; Fishwick, K.J.; Da Costa, M.J.M.D.; Ewington, L.J.; et al. Impact of Sitagliptin on Endometrial Mesenchymal Stem-like Progenitor Cells: A Randomised, Double-Blind Placebo-Controlled Feasibility Trial. EBioMedicine 2020, 51, 102597. [Google Scholar] [CrossRef] [Green Version]
  153. Vella, A. Mechanism of Action of DPP-4 Inhibitors—New Insights. J. Clin. Endocrinol. Metab. 2012, 97, 2626–2628. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Kieffer, T.J.; Mc Intosh, C.H.S.; Pederson, R.A. Degradation of Glucose-Dependent Insulinotropic Polypeptide and Truncated Glucagon-like Peptide 1 In Vitro and In Vivo by Dipeptidyl Peptidase Iv. Endocrinology 1995, 136, 3585–3596. [Google Scholar] [CrossRef] [PubMed]
  155. Zhong, J.; Rajagopalan, S. Dipeptidyl Peptidase-4 Regulation of SDF-1/CXCR4 Axis: Implications for Cardiovascular Disease. Front. Immunol. 2015, 6, 477. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Munné, S.; Kaplan, B.; Frattarelli, J.L.; Child, T.; Nakhuda, G.; Shamma, F.N.; Silverberg, K.; Kalista, T.; Handyside, A.H.; Katz-Jaffe, M.; et al. Preimplantation Genetic Testing for Aneuploidy versus Morphology as Selection Criteria for Single Frozen-Thawed Embryo Transfer in Good-Prognosis Patients: A Multicenter Randomized Clinical Trial. Fertil. Steril. 2019, 112, 1071–1079.e7. [Google Scholar] [CrossRef]
  157. Nagori, C.B.; Panchal, S.Y.; Patel, H. Endometrial Regeneration Using Autologous Adult Stem Cells Followed by Conception by In Vitro Fertilization in a Patient of Severe Asherman’s Syndrome. J. Hum. Reprod. Sci. 2011, 4, 43–48. [Google Scholar] [CrossRef]
  158. Santamaria, X.; Cabanillas, S.; Cervelló, I.; Arbona, C.; Raga, F.; Ferro, J.; Palmero, J.; Remohí, J.; Pellicer, A.; Simón, C. Autologous Cell Therapy with CD133+ Bone Marrow-Derived Stem Cells for Refractory Asherman’s Syndrome and Endometrial Atrophy: A Pilot Cohort Study. Hum. Reprod. 2016, 31, 1087–1096. [Google Scholar] [CrossRef] [Green Version]
  159. Zhang, L.; Li, Y.; Guan, C.-Y.; Tian, S.; Lv, X.-D.; Li, J.-H.; Ma, X.; Xia, H.-F. Therapeutic Effect of Human Umbilical Cord-Derived Mesenchymal Stem Cells on Injured Rat Endometrium during Its Chronic Phase. Stem Cell Res. Ther. 2018, 9, 36. [Google Scholar] [CrossRef] [Green Version]
  160. Xin, L.; Lin, X.; Pan, Y.; Zheng, X.; Shi, L.; Zhang, Y.; Ma, L.; Gao, C.; Zhang, S. A Collagen Scaffold Loaded with Human Umbilical Cord-Derived Mesenchymal Stem Cells Facilitates Endometrial Regeneration and Restores Fertility. Acta Biomater. 2019, 92, 160–171. [Google Scholar] [CrossRef]
  161. Cao, Y.; Sun, H.; Zhu, H.; Zhu, X.; Tang, X.; Yan, G.; Wang, J.; Bai, D.; Wang, J.; Wang, L.; et al. Allogeneic Cell Therapy Using Umbilical Cord MSCs on Collagen Scaffolds for Patients with Recurrent Uterine Adhesion: A Phase I Clinical Trial. Stem Cell Res. Ther. 2018, 9, 192. [Google Scholar] [CrossRef] [Green Version]
  162. Gan, L.; Duan, H.; Xu, Q.; Tang, Y.-Q.; Li, J.-J.; Sun, F.-Q.; Wang, S. Human Amniotic Mesenchymal Stromal Cell Transplantation Improves Endometrial Regeneration in Rodent Models of Intrauterine Adhesions. Cytotherapy 2017, 19, 603–616. [Google Scholar] [CrossRef]
  163. Li, B.; Zhang, Q.; Sun, J.; Lai, D. Human Amniotic Epithelial Cells Improve Fertility in an Intrauterine Adhesion Mouse Model. Stem Cell Res. Ther. 2019, 10, 257. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Ouyang, X.; You, S.; Zhang, Y.; Zhang, C.; Zhang, G.; Shao, X.; He, F.; Hu, L. Transplantation of Human Amnion Epithelial Cells Improves Endometrial Regeneration in Rat Model of Intrauterine Adhesions. Stem Cells Dev. 2020, 29, 1346–1362. [Google Scholar] [CrossRef] [PubMed]
  165. Bai, X.; Liu, J.; Yuan, W.; Liu, Y.; Li, W.; Cao, S.; Yu, L.; Wang, L. Therapeutic Effect of Human Amniotic Epithelial Cells in Rat Models of Intrauterine Adhesions. Cell Transplant. 2020, 29, 963689720908495. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Choi, J.; Jo, M.; Lee, E.; Oh, Y.K.; Choi, D. The Role of Autophagy in Human Endometrium. Biol. Reprod. 2012, 86, 70. [Google Scholar] [CrossRef]
  167. Kilic, S.; Yuksel, B.; Pinarli, F.; Albayrak, A.; Boztok, B.; Delibasi, T. Effect of Stem Cell Application on Asherman Syndrome, an Experimental Rat Model. J. Assist. Reprod. Genet. 2014, 31, 975–982. [Google Scholar] [CrossRef] [Green Version]
  168. Shao, X.; Ai, G.; Wang, L.; Qin, J.; Li, Y.; Jiang, H.; Zhang, T.; Zhou, L.; Gao, Z.; Cheng, J.; et al. Adipose-Derived Stem Cells Transplantation Improves Endometrial Injury Repair. Zygote 2019, 27, 367–374. [Google Scholar] [CrossRef]
  169. Monsef, F.; Artimani, T.; Alizadeh, Z.; Ramazani, M.; Solgi, G.; Yavangi, M.; Asl, S.S. Comparison of the Regenerative Effects of Bone Marrow/Adipose-Derived Stem Cells in the Asherman Model Following Local or Systemic Administration. J. Assist. Reprod. Genet. 2020, 37, 1861–1868. [Google Scholar] [CrossRef]
  170. Lee, S.Y.; Shin, J.E.; Kwon, H.; Choi, D.H.; Kim, J.H. Effect of Autologous Adipose-Derived Stromal Vascular Fraction Transplantation on Endometrial Regeneration in Patients of Asherman’s Syndrome: A Pilot Study. Reprod. Sci. 2020, 27, 561–568. [Google Scholar] [CrossRef]
  171. Rawlings, T.M.; Makwana, K.; Tryfonos, M.; Lucas, E.S. Organoids to Model the Endometrium: Implantation and Beyond. Reprod. Fertil. 2021, 2, R85–R101. [Google Scholar] [CrossRef]
  172. Turco, M.Y.; Gardner, L.; Hughes, J.; Cindrova-Davies, T.; Gomez, M.J.; Farrell, L.; Hollinshead, M.; Marsh, S.G.E.; Brosens, J.J.; Critchley, H.O.; et al. Long-Term, Hormone-Responsive Organoid Cultures of Human Endometrium in a Chemically Defined Medium. Nat. Cell Biol. 2017, 19, 568–577. [Google Scholar] [CrossRef]
  173. Luddi, A.; Pavone, V.; Semplici, B.; Governini, L.; Criscuoli, M.; Paccagnini, E.; Gentile, M.; Morgante, G.; Leo, V.D.; Belmonte, G.; et al. Organoids of Human Endometrium: A Powerful In Vitro Model for the Endometrium-Embryo Cross-Talk at the Implantation Site. Cells 2020, 9, 1121. [Google Scholar] [CrossRef] [PubMed]
  174. Kim, J.; Koo, B.-K.; Knoblich, J.A. Human Organoids: Model Systems for Human Biology and Medicine. Nat. Rev. Mol. Cell Biol. 2020, 21, 571–584. [Google Scholar] [CrossRef] [PubMed]
  175. Zheng, Y.; Xue, X.; Shao, Y.; Wang, S.; Esfahani, S.N.; Li, Z.; Muncie, J.M.; Lakins, J.N.; Weaver, V.M.; Gumucio, D.L.; et al. Controlled Modelling of Human Epiblast and Amnion Development Using Stem Cells. Nature 2019, 573, 421–425. [Google Scholar] [CrossRef] [PubMed]
  176. Liu, X.; Tan, J.P.; Schröder, J.; Aberkane, A.; Ouyang, J.F.; Mohenska, M.; Lim, S.M.; Sun, Y.B.Y.; Chen, J.; Sun, G.; et al. Modelling Human Blastocysts by Reprogramming Fibroblasts into IBlastoids. Nature 2021, 591, 627–632. [Google Scholar] [CrossRef] [PubMed]
  177. Yu, L.; Wei, Y.; Duan, J.; Schmitz, D.A.; Sakurai, M.; Wang, L.; Wang, K.; Zhao, S.; Hon, G.C.; Wu, J. Blastocyst-like Structures Generated from Human Pluripotent Stem Cells. Nature 2021, 591, 620–626. [Google Scholar] [CrossRef]
  178. Haider, S.; Meinhardt, G.; Saleh, L.; Kunihs, V.; Gamperl, M.; Kaindl, U.; Ellinger, A.; Burkard, T.R.; Fiala, C.; Pollheimer, J.; et al. Self-Renewing Trophoblast Organoids Recapitulate the Developmental Program of the Early Human Placenta. Stem Cell Rep. 2018, 11, 537–551. [Google Scholar] [CrossRef] [Green Version]
  179. Turco, M.Y.; Gardner, L.; Kay, R.G.; Hamilton, R.S.; Prater, M.; Hollinshead, M.S.; McWhinnie, A.; Esposito, L.; Fernando, R.; Skelton, H.; et al. Trophoblast Organoids as a Model for Maternal-Fetal Interactions during Human Placentation. Nature 2018, 564, 263–267. [Google Scholar] [CrossRef] [Green Version]
  180. Rinehart, C.A.; Lyn-Cook, B.D.; Kaufman, D.G. Gland Formation from Human Endometrial Epithelial Cells In Vitro. In Vitro Cell. Dev. Biol. 1988, 24, 1037–1041. [Google Scholar] [CrossRef]
  181. Bläuer, M.; Heinonen, P.K.; Martikainen, P.M.; Tomás, E.; Ylikomi, T. A Novel Organotypic Culture Model for Normal Human Endometrium: Regulation of Epithelial Cell Proliferation by Estradiol and Medroxyprogesterone Acetate. Hum. Reprod. 2005, 20, 864–871. [Google Scholar] [CrossRef] [Green Version]
  182. Boretto, M.; Maenhoudt, N.; Luo, X.; Hennes, A.; Boeckx, B.; Bui, B.; Heremans, R.; Perneel, L.; Kobayashi, H.; Van Zundert, I.; et al. Patient-Derived Organoids from Endometrial Disease Capture Clinical Heterogeneity and Are Amenable to Drug Screening. Nat. Cell Biol. 2019, 21, 1041–1051. [Google Scholar] [CrossRef]
  183. Hill, C.J.; Alijassim, F.; Phelan, M.M.; Tempest, N.; Hapangama, D.K. Bipartite Collagen-Agarose Scaffolds to Model Eutopic Embryo Implantation. In Proceedings of the Reproductive Sciences, Denver, CO, USA, 1 March 2022; Volume 29, p. 185. [Google Scholar]
Figure 1. Aetiology of recurrent reproductive failure. (A) Risk factors for recurrent reproductive failure (RRF). (B) Demonstrable causes of recurrent implantation failure and recurrent pregnancy loss. The contribution of basalis-resident stem/progenitor cells and glandular architecture to RRF pathophysiology has yet to be fully explored.
Figure 1. Aetiology of recurrent reproductive failure. (A) Risk factors for recurrent reproductive failure (RRF). (B) Demonstrable causes of recurrent implantation failure and recurrent pregnancy loss. The contribution of basalis-resident stem/progenitor cells and glandular architecture to RRF pathophysiology has yet to be fully explored.
Jpm 12 00775 g001
Figure 2. Proposed stem cell niches of the human endometrium. Stromal stem cells reside throughout the superficial and deep endometrial tissue as CD146+, sushi domain containing-2 (SUSD2+), and platelet-derived growth factor receptor β positive (PDGFRβ+) perivascular cells. Putative epithelial stem cell populations have been described in the basalis layer, a region characterised by expression of progenitor markers, including leucine-rich repeat-containing G protein-coupled receptor 5 (LGR5), SRY-box 9 (SOX9), axis inhibition protein 2 (AXIN2), and nuclear β-catenin. Specific cells within the deeper basalis glands express N-cadherin, whilst stage-specific embryonic antigen-1 (SSEA-1) is characteristic of more superficial basalis regions.
Figure 2. Proposed stem cell niches of the human endometrium. Stromal stem cells reside throughout the superficial and deep endometrial tissue as CD146+, sushi domain containing-2 (SUSD2+), and platelet-derived growth factor receptor β positive (PDGFRβ+) perivascular cells. Putative epithelial stem cell populations have been described in the basalis layer, a region characterised by expression of progenitor markers, including leucine-rich repeat-containing G protein-coupled receptor 5 (LGR5), SRY-box 9 (SOX9), axis inhibition protein 2 (AXIN2), and nuclear β-catenin. Specific cells within the deeper basalis glands express N-cadherin, whilst stage-specific embryonic antigen-1 (SSEA-1) is characteristic of more superficial basalis regions.
Jpm 12 00775 g002
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Al-Lamee, H.; Hill, C.J.; Turner, F.; Phan, T.; Drakeley, A.J.; Hapangama, D.K.; Tempest, N. The Role of Endometrial Stem/Progenitor Cells in Recurrent Reproductive Failure. J. Pers. Med. 2022, 12, 775. https://doi.org/10.3390/jpm12050775

AMA Style

Al-Lamee H, Hill CJ, Turner F, Phan T, Drakeley AJ, Hapangama DK, Tempest N. The Role of Endometrial Stem/Progenitor Cells in Recurrent Reproductive Failure. Journal of Personalized Medicine. 2022; 12(5):775. https://doi.org/10.3390/jpm12050775

Chicago/Turabian Style

Al-Lamee, Hannan, Christopher J. Hill, Florence Turner, Thuan Phan, Andrew J. Drakeley, Dharani K. Hapangama, and Nicola Tempest. 2022. "The Role of Endometrial Stem/Progenitor Cells in Recurrent Reproductive Failure" Journal of Personalized Medicine 12, no. 5: 775. https://doi.org/10.3390/jpm12050775

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