Next Article in Journal
Update on the Management of Diabetic Retinopathy: Anti-VEGF Agents for the Prevention of Complications and Progression of Nonproliferative and Proliferative Retinopathy
Next Article in Special Issue
Cook with Different Pots, but Similar Taste? Comparison of Phase Angle Using Bioelectrical Impedance Analysis According to Device Type and Examination Posture
Previous Article in Journal
Ageing People Living with HIV/AIDS, PLWHA; More Dental Challenges; the Romanian Dental Professional’s Point of View
Previous Article in Special Issue
Systematic Review and Meta-Analysis: Recurrent Laryngeal Nerve Variants and Their Implication in Surgery and Neck Pathologies, Using the Anatomical Quality Assurance (AQUA) Checklist
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Immune Cell Functionality during Decidualization and Potential Clinical Application

1
Department of Gynecology and Gynecological Oncology, University Hospital Bonn, 53127 Bonn, Germany
2
Department of Gynecological Endocrinology and Reproductive Medicine, University Hospital Bonn, 53127 Bonn, Germany
*
Author to whom correspondence should be addressed.
Life 2023, 13(5), 1097; https://doi.org/10.3390/life13051097
Submission received: 21 March 2023 / Revised: 20 April 2023 / Accepted: 26 April 2023 / Published: 27 April 2023
(This article belongs to the Special Issue Feature Papers in Medical Research)

Abstract

:
Due to a vast influx in the secretory phase of the menstrual cycle, leukocytes represent 40–50% of the decidua at the time of implantation. Their importance for the implantation, maintenance of pregnancy, and parturition are known yet not fully understood. Thus, in idiopathic infertility, decidual immune-related factors are speculated to be the cause. In this review, the immune cell functions in the decidua were summarized, and clinical diagnostics, as well as interventions, were discussed. There is a rising number of commercially available diagnostic tools. However, the intervention options are still limited and/or poorly studied. In order for us to make big steps towards the proper use of reproductive immunology findings, we need to understand the mechanisms and especially support translational research.

1. Introduction

For a successful pregnancy free of complications, maternal immune factors, including cells and soluble factors, must be precisely balanced [1]. On the one hand, the semi-allogeneic fetus expressing both maternal and paternal antigens must be tolerated. On the other hand, tissue remodeling is constantly accompanied by both destructive (type 1 inflammation) and constructive (type 2 inflammation) immune mechanisms (see Figure 1). Thus, implantation requires inflammatory actions, while protection against foreign antigens and pathogens must not be impaired at the fetomaternal interface [2].
Active tolerance mechanisms mediate the prevention of rejection of the sperm and, later, also of the conceptus. In contrast to passive tolerance due to a lack of immune mechanisms, active tolerance is involved in pregnancy [3]. The maternal immune milieu is characterized by immunomodulatory and anti-inflammatory factors, e.g., interleukin (IL)-4, IL-10, transforming growth factor (TGF)-β, leukemia inhibitory factor (LIF) and human chorionic gonadotropin (hCG). Their secretion occurs by the cells of the reproductive tract, embryonal cells, as well as by immune cells themselves. Interestingly, the reproductive, immune cells are adapted to their reproductive tasks and are, therefore, distinct from their peripheral pendants. They recognize foreign antigens but induce active tolerogenic responses and, thereby, induce the development of regulatory T helper cells and B cells [3,4,5].
Decidualization of the endometrium includes all processes that prepare the tissue for the implantation of blastocysts during the menstrual cycle [6]. The endometrium is composed of epithelial and stromal cells as well as secretory cells that form the glands. Furthermore, the endometrium is permeated by numerous vessels and has a strong blood supply. In relation to the ovarian cycle, decidualization begins after ovulation, when both hormones, progesterone and estradiol, increase. By that, specific processes are initiated [7]. Endometrial stroma cells differentiate into decidual stroma cells. In response to the hormones, decidual cells proliferate, and a receptive microenvironment is formed [6,8]. During decidualization, stromal cells secrete the Insulin-like growth factor binding protein (IGFBP)-1 and prolactin. These factors are also used as markers for decidualization in vitro. IGFBP-1 controls growth and development—especially under hypoxic conditions [9], as found in early pregnancy. Moreover, due to the decidual transition of the stroma cells, they secrete increasing amounts of IL-15 [10].
The process of decidualization and implantation parallels the initiation and progression of benign and malignant neoplasms. While cancer cells transform from epithelial to mesenchymal cells, the reverse takes place during decidualization. Endometrial fibroblastic stromal cells undergo mesenchymal-to-epithelial transformation [11], becoming epithelial-like cells. Similar to cancer cells, these exhibit high proliferative, anti-apoptotic capacities [12,13]. These processes are hormonally driven to varying extents [13,14,15]. Signaling cascades are also shared, including key regulators of cell growth (mitogen-activated protein (MAP) kinases, neurogenic locus notch homolog protein 1 (Notch-1), and Dickkopf-related protein 1 (Dkk1) [16,17,18,19,20]), cell motility (Notch 1 and homeobox protein A10 (HOXA10) [12,15,21,22]) and the interaction between the immune system. Similarly, to immune cells in the decidua, angiogenetic and invasive processes can be supported by immune cells in the tumor microenvironment. Both tumor and trophoblastic cells express immune inhibitory ligands, including B7 family molecules such as programmed cell death ligand (PD-L) 1, PD-L2, CD80, and CD86 [23,24], and T cell immunoglobulin and mucin-domain containing-3 ligand (TIM-3L) [25]. Similar to the immune cells, such as macrophages and NK cells, which support trophoblast invasion, the presence of tumor-associated macrophages (TAMs) is associated with tumor progression and metastasis [26,27]. In contrast, the presence of NK cells per se is no marker of tumor progression unless the phenotype is considered. Whereas cytotoxic NK cells show anti-tumoral effects, low-cytotoxic NK cells rather support tumor progress [25]. Moreover, higher prolactin levels, as found during decidualization, are also observed in several tumor types, especially in breast cancer [28].
The stromal cell reprogramming includes the downregulation of inflammatory capacity [29]. Moreover, immune cells are recruited to the decidualized tissue progressively, which participates in the functionalization of the decidua. On the one hand, immune cells are ultimately instrumental in implantation [6,30,31]. Natural killer (NK) cells and macrophages are found in close proximity to implanting trophoblast cells and support invading processes [32] accompanied by rather inflammatory factors. On the other hand, the semi-allogenic fetus must be tolerated. Several tolerance mechanisms take part in this process. The prerequisite for this is the general shift of the local immune micro milieu toward tolerance, which is accomplished by various factors. Decidual leukocytes are affected by seminal plasma if exposed to it, which contains several immunomodulating factors (e.g., TGF-β, soluble human leukocyte antigen (HLA)-G). Although foreign antigens of the semen lead to the activation of immune responses, these factors shift the leukocytes towards tolerogenic behavior. In humans, decidualization starts independently of conception in every menstrual cycle. After conception, decidualization continues and is additionally affected by the secreted factors of the embryo. Trophoblastic cells secrete TGF-β and especially hCG, which further affect the decidua and leukocytes. Moreover, in the first trimester before placental perfusion, vascularization and plugging of the maternal arteries prevents blood flow to the placental intervillous space. This creates and maintains a hypoxic environment [33]. Hypoxia also affects the decidual leukocytes by the stabilization of the transcription factor HIF (hypoxia-inducible factor). HIF regulates over 70 targets directly and, thereby, promotes angiogenic as well as tolerogenic milieu [34].
Due to the vast recruitment, the early decidua contains approximately 30–40% leukocytes, of which NK cells represent the largest subpopulation at 70%. The second most abundant leukocytic cell type is macrophages (20%) [35,36]. Other decidual immune cells include T cells, B cells, dendritic cells, and other innate lymphoid cells [35,37,38]. Immune cells support the processes of decidualization, implantation, and placentation.
The aim of this review is to provide an overview of the importance of the immune balance, which does not only include pro- and anti-inflammation but type 1 inflammation, type 2 inflammation (often named as anti-inflammatory because of its inhibitory actions on type 1 inflammation) and the tolerogenic mechanisms. It also provides a summary of the idea behind diagnostic efforts and their limitations, as well as the need for improvement concerning therapeutical options and especially the definition of suitable guidelines.

2. Decidual Innate Lymphoid Cells

The most abundant leukocytes in the decidua are NK cells (50–70%), which makes the uterus the organ with the highest frequency of NK cells in the body [33]. Interestingly, uterine NK cells differ vastly from blood NK cells. In the periphery, NK cells live up to their name as cytotoxic defenders—especially against infected and tumor cells. In contrast to peripheral blood NK cells, uterine NK cells do barely act cytotoxic but produce cytokines, growth factors, and chemokines during decidualization, receptivity, and implantation [38] (see Figure 2). Their metabolism and protein profile are unique when compared to peripheral NK cells [39]. Several factors contribute to the decidual phenotype of NK cells, including IL-15, TGF-β [40], and hypoxia [41].
Blood NK cells are activated through the Fc receptor CD16/FcγRIIIa to initiate antibody-dependent cell-mediated cytotoxicity (ADCC), which is not expressed in the uterine subset (CD16-CD56hi). Moreover, NK cells detect target cells with downregulated major histocompatibility complex (MHC)I escaping the recognition by cytotoxic T cells (referred to as “missing self”). Thereby, the cytotoxic behavior of NK cells is inhibited by MHCI. This mechanism plays a pivotal role in fertility. To maintain the decidual phenotype of NK cells, fetal extravillous cells express HLA-G and -E isoforms [42], which are less polymorphic, well-conserved, non-classical MHCI molecules. These pregnancy-related immunomodulators induce inhibitory decidual NK cell signaling and suppress the anti-fetal immune response [42,43]. In addition, HLA-G stimulates decidual NK cells to secrete various factors, such as vascular endothelial growth factor (VEGF), IL-6, and IL-8 [44,45]. These cytokines promote invasion and angiogenesis and are, therefore, central to decidualization and, eventually, placentation [46,47]. This makes NK cells essential in the remodeling of spiral arteries, which is necessary for proper placentation [48]. Moreover, decidual NK cells produce both pro- and anti-inflammatory cytokines, including interferon (IFN)-γ and tumor necrosis factor (TNF)-α as well as TGF-β and IL-10 [33,49], contributing to the local immune balance. Chemokines produced by decidual NK cells involving CCL5, CXCL10, and CXCL8 (IL-8) comply with several tasks. On the one hand, leukocytes are recruited to the decidua. On the other hand, NK cells guide the trophoblast during implantation in terms of the right direction and invasion depth into the decidua [50,51,52]. Therefore, NK cells are located in close proximity to trophoblast cells [32].
NK cells belong to the group of innate lymphoid cells (ILC). Recently, the involvement of the other ILC subtypes in processes at the fetomaternal interface (<1%; [53]) has been described [53,54]. ILCs correspond to T cells in terms of cytokine secretion but do not express T cell receptors (lin-CD127+ [55]; NK cells—cytotoxic T cells, ILC1-T helper (Th)1, ILC2-Th2, ILC3-Th17). However, the function and occurrence of the ILC 1–3 subtypes during pregnancy are largely unclear [53,56,57]. ILC3 has been the most studied and is the second most abundant ILC subtype in the decidua after NK cells. They produce the cytokines IL-8, IL-17, IL-22, and granulocyte-macrophage colony-stimulating factor (GM-CSF, CSF2), which also play important roles in pregnancy [58,59]. IL-8 and IL-17 support trophoblast invasion [60]. Low IL-22 and GM-CSF levels can be correlated with recurrent pregnancy loss [61,62,63,64]. Because ILC3 also promotes tolerance to intestinal commensals, similar tolerance-stimulating properties are also thought to occur during the stages of pregnancy. Although ILC3 can express MHCII, it is specifically downregulated in the uterus of mice and in the human decidua at term. TGF-β, hCG, and hypoxic conditions, which are major regulators in decidualization and early pregnancy, cause a downregulation of MHCII, which might contribute to fetal tolerance [55,65].

3. Decidual Macrophages

Since macrophages are key cells in tissue remodeling in both destructing and constructing processes, they are vital leukocytes due to the course of the menstrual cycle. In the decidua, macrophages form the second most abundant leukocyte population after NK cells (20%) [66]. Macrophages are phagocytes keeping homeostasis, can mediate antigen presentation, and participate in creating the immune milieu by cytokine production [67,68] (see Figure 2). During pregnancy, different macrophage subtypes accomplish the diverse tasks in the decidua [69,70]. However, the predominant differentiation stage of the decidual macrophages varies depending on the gestational age [71]. The beginning of gestation is more characterized by inflammatory mechanisms in the context of invasion and tissue rearrangement. At the time of implantation, macrophages resemble mainly pro-inflammatory M1 macrophages [72], which soon develop into tissue-remodeling M2a macrophages. During placentation, macrophages are located in the stroma near the invading trophoblasts and spiral arteries [32,73]. There, they support trophoblast invasion and spiral artery remodeling [74] by the secretion and regulation of the activity of matrix metalloproteinases (MMPs) [75]. These MMPs mediate the breakdown of the extracellular matrix, loosening the tissue integrity in order to rearrange it. Similar to decidual NK cells, HLA-G from extravillous trophoblasts induces macrophages to produce IL-6 and IL-8 in the first trimester [45]. By that, macrophages support angiogenesis and trophoblast invasion [76,77]. In addition, decidual macrophages secrete chemotactic molecules, cytokines, and growth factors to support placentation [78]. Furthermore, clearance of apoptotic degradation bodies by macrophages occurs [79,80,81,82].
After the implantation and placentation phase, immune cells mainly mediate fetal tolerance. This prevents fetal rejection. Trophoblastic cells secrete TGF-β, CXCL16, PD-L1, IL-10, and macrophage colony-stimulating factor (M-CSF, CSF1) for macrophage stimulation [81,82,83,84]. These factors differentiate the macrophages into an M2c-like phenotype. M2c macrophages secrete anti-inflammatory cytokines, including IL-10 and TGF-β, contributing to a tolerogenic milieu [71,85]. Due to the large number of macrophages at the fetomaternal interface but the low number of dendritic cells, macrophages are the predominant antigen-presenting cells at this site [86]. However, decidual macrophages have an altered antigen presentation potential and are more likely to mediate active tolerance [87].
The onset of contractions marks the end of pregnancy. Now the immunological milieu of the endometrium is modulated toward inflammatory processes [88]. Accordingly, macrophages differentiate back to the M1 phenotype and support inflammatory processes by secreting IL-6 and decreasing IL-10 and TGF-β levels [80,89].

4. Other Leukocytes in the Decidua

Dendritic cells (DC) represent appr. 2% of the decidual leukocytes [90]. Defense against infection is the main task for DC in non-pregnant women. Although present in a relatively low abundance, they interact with T cells, NK cells, and macrophages and thus regulate the more abundant cells at the fetomaternal interface. Thereby, DC supports the decidualization and implantation process [91]. Most significantly, during activation of T cells, DC determines the subtype, of which the balance is important for pregnancy success. DC is influenced by hormones, such as progesterone and estrogen, as well as hCG. hCG drives DC-mediated regulatory T cell (Treg) differentiation, whereas progesterone and estradiol rather support DC-mediated Th2 differentiation.
T lymphocytes are as well present at the fetomaternal interface [52]. In the CD4+ Th subset, Th1 lymphocytes can be detrimental to fetal tolerance when activated. In contrast, Tregs help to create a tolerogenic environment, and Th2 cells support the remodeling processes. The largest fraction of decidual T cells is CD8+ T lymphocytes [92]. These cytotoxic T cells have to be tightly controlled to not disturb fetal tolerance. Similar to their innate lymphoid cell pendants (NK cells), the decidual cytotoxic T cells differ from peripheral CD8+ T cells. They interact with inhibitory molecules such as HLA-C expressed by trophoblast cells and express significantly enhanced co-inhibitory molecules such as inhibitory killer cell immunoglobulin-like receptor (KIR), Tim-3 and cytotoxic T lymphocyte-associated protein 4 (CTLA-4) compared to peripheral CD8+ T lymphocytes supporting fetal tolerance [92,93].
Along with the other leukocytes, B cells are present in the decidua as well. They participate in the defense against infection but also support fetal tolerance. A subset of regulatory B cells (Bregs) secret anti-inflammatory cytokines supporting the tolerogenic niche. Moreover, B cells are able to express protective antibodies against paternal antigens to prevent rejection [94]. Antibody-producing B cells are also referred to as plasma cells.
Dendritic cells are also present in the human decidua an represent 1.7% of the leukocytes [90]. They show an immature phenotype [90,95]. In vitro, decidual dendritic cells mediate tolerance towards T cells [95] by secreting anti-inflammatory factors such as IL-10 under the influence of decidualized stromal cells [96].

5. Immune Implications in Adverse Pregnancy Outcomes

Inadequate decidualization can cause subfertility, infertility, and adverse pregnancy outcomes. The decidua creates a receptive environment which is needed for the attachment of the blastocyst, the invasion of trophoblast cells, and the placentation. Thereby, different immune types cooperate, including tolerance as well es tissue remodeling which integrates destructive (type 1 inflammation) and regenerative (type 2 inflammation) mechanisms, which have to be timely coordinated (see Figure 1).
Recurrent implantation failure (RIF) or recurrent pregnancy loss (RPL) can be caused by a disbalance towards type 1 inflammation due to either an increase in type 1 cytokines (i.e., TNFα or IFNγ), inflammatory cells (i.e. Th1) or cytotoxic activity by NK cells or a decrease in tolerogenic (i.e. Tregs, M2c macrophages, IL-10, TGF-β) or type 2 inflammatory mechanisms (i.e. Th2, M2a macrophages, IL-4, IL-13). Reasons for this imbalance can be manifold, including systemic immune alterations, genetic constitution, inflammatory effectors including immune disorders, stress, diet, physical exercises, and obesity, or altered activators including paternal factors, HLA matching, genetics of the embryo, and more. On a cellular basis, not only local immune cells but also trophoblast cells, as well as decidual epithelial and stromal cells, affect the balance by activating and inhibiting soluble and cell-to-cell-contact-mediated factors and receptors [24,42,51,52,97,98,99,100].
Detailed insight has already been provided by several reviews (i.e. in [101,102,103,104,105]).

6. Clinical Significance in Reproductive Medicine

The process of decidualization includes the proliferation and priming of endometrial stroma cells. This includes tissue remodeling and angiogenesis. The influx of leukocytes supports this structural adaption as well as the establishment of a receptive, tolerogenic milieu. General interventions to improve decidualization success are limited (see Table 1) but developing. The further sections aim to provide an overview of diagnostical and therapeutical tools targeting the immune balance during the decidualization process.

6.1. Targeting Regeneration

6.1.1. Endometrial Scratching

Endometrial scratching causes a local injury that is aimed at activating the wound healing processes, which share mechanisms with decidualization [106]. However, the studies came to inconsistent conclusions. Several studies show an enhanced implantation rate after scratching [107,108,109,110,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138], while there are also several studies that could not or barely show significant differences due to scratching [139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155,156,157,158,159,160,161,162,163,164]. Some of these studies even recorded a negative effect or considerable pain for the patient, clearly delivering arguments against endometrial scratching. As the enthusiasm conducting endometrial scratching is declining, the debate concerning the effect of endometrial scratching is still ongoing, where many factors play a potentially decisive role.
The timing of scratching throughout the cycle and the timing of the subsequent embryo transfer might be detrimental. Only a few studies addressed this in detail. A study showed better results when endometrial scratching was conducted in the luteal phase of the previous cycle compared to the follicular phase in the same cycle of the embryo transfer [110]. It was shown that endometrial scratching has a timely limited effect but is not restricted to the actual cycle. Until around 90 days after intervention, an improving effect was observed [111]. However, in a study where scratching in the proliferative (65.6%), periovulatory (69.6%), or secretory (64.3%) phases were compared, no significant differences due to the timing were seen [165]. Moreover, the form and force of the intervention might also affect the outcome. Peeling instead of scratching was shown to improve the pregnancy rate [112]. The aim of scratching is not to cause a vast injury but activate wound healing mechanisms due to a limited injury. Similar approaches are also used in other disciplines. In dermatology, microneedling is used to cause minimal physical trauma, which then activates regeneration due to the release of growth factors and stimulation of stem cells [166]. Whether similar mechanisms are also active in endometrial scratching remains to be elucidated. Nonetheless, molecular studies repeatedly showed an elevation of the expression of receptivity genes after scratching.
In several studies, endometrial scratching induced an elevation of pro- receptivity factors LIF [167], the homeobox proteins HOXA10 and HOXA11, as well as the cytokines IL-6, IL-8, IL-12, IL-13, IFN-𝛾 and monocyte chemotactic protein-(MCP-) 1 [116] as well as pro-angiogenic factors HIF, VEGF and the actual microvessel density [140].
These factors mediate and support receptivity and implantation. However, in patients with RIF LIF [168], HOXA-10 [169], HOXA-11 [170], HIF and the microvessel density [140] are significantly decreased. Restoration of these factors due to endometrial scratching might support implantation and placentation. On a molecular level, endometrial scratching builds a receptive microenvironment. The success of this intervention might, however, be dependent on additional factors. Scratching might support decidualization when there is a temporal or environmental reason, but not genetically or chronically altered decidualization. The identification of a suitable patient group could support the success of endometrial scratching.
For a more detailed insight, many reviews and meta-analytic publications were published concerning this topic, which seems to be declining but is still under a heated debate (reviewed i.a. in [171,172,173,174,175,176,177,178]).

6.1.2. Platelet-Rich Plasma

The infusion with autologous platelet-rich plasma (PRP) is thought to support regeneration processes to improve thin endometrial lining found in patients with RIF [179]. PRP is found to be rich in growth factors, cytokines, and antibacterial peptides—especially after the activation of the platelets. This includes the tolerance-mediating TGF-β, pro-angiogenic VEGF, other growth factors such as platelet-derived growth factors PDGF, fibroblast growth factor FGF, insulin-like growth factor IGF1, IGF2 and epidermal growth factor EGF, inflammatory cytokines IL-8 and regenerative cytokines IL-4, IL-13 and IL-17 [180,181,182].
Thus, it locally facilitates regenerative mechanisms and benefits the whole decidualization process. In patients with thin endometrial lining, RIF, and recurrent miscarriage, the intrauterine infusion improves pregnancy rates and success effectively (reviewed i.a. in [183,184,185,186,187,188]). Comparing intrauterine infusion of hCG, G-CSF, PBMCs, or PRP, the infusion of PRP showed the most effective results in patients with RIF [189].
Activated PRP by thrombin and calcium chloride improves the in vitro behavior, including migration, invasion, and proliferation of endometrial cells [190]. Primary endometrial cells and cell lines show an increased expression of proteases, cytokines including IL-1α, IL-1β, and IL-15, and chemokines including CCL5, CCL7, and CXCL13 after PRP treatment [190]. These interleukins can activate an immune response. Since IL-15 is important for NK cell function, it supports the major decidual leukocyte subset. Proteases are necessary for tissue remodeling, which is essential in implantation. Chemokines recruit further leukocytes to the decidua to support its proper function. Moreover, it affects the hormonal levels supporting implantation success [191].
Although standardization of the method is still lacking, the intrauterine infusion with PRP is a promising tool supporting the regenerative capacity of the endometrial/decidual tissue.

6.2. Targeting Immune Balance

It is speculated that the majority of idiopathic infertility and (recurrent) pregnancy complications are caused by immunological disturbances. Genetic as well as environmental influences affect the immune cell’s ability to create the needed tolerogenic niche. There are diagnostic tools available. However, the therapeutical interventions, which directly target immune components, are still limited, or their application is not sufficiently tested [192,193].

6.2.1. Diagnostic Tools

Several commercial tests are already available which directly or indirectly capture immune-related changes. Endometrial biopsies or pipelle samples can be tested for NK cell, Treg, and plasma cell counts [194,195]. Altered numbers in these immune cells can be an indication of an immune-related cause of infertility.
Elevated plasma cell (antibody-producing B lymphocytes) counts indicate chronic endometritis [196]. An altered endometrial microbiome or chronic infections can create a misregulated inflammatory environment, which impairs fertility. Commercial tests are available to sequence the microbial colonization of the endometrium. However, the treatment options are limited to antibiotics combined with pre- and probiotic support afterward [197]. This might help to establish a healthy microbiome in all body niches, including the uterus. It is thought that besides the occurrence of healthy or unhealthy species also, the quantity plays a critical role in the effects of the upper reproductive tract microbiome [198].
Not only the number but also the function of the immune cells affect the fertility. The activity of immune cells depends on external factors, involving soluble factors creating the local immune milieu and cell-to-cell contacts, as well as internal factors, such as the genetic variants and expression quantity of receptors. Classical MHCI molecules show a broad polymorphism, of which certain haplotypes were correlated with increased pregnancy complications. These can be addressed by the characterization of the HLA and KIR or its receptor KIR genotyping [199]. Certain HLA and their pregnancy-relevant receptor types are found to be associated with poor IVF outcomes, including disturbances in implantation, the formation of the placenta, or the maintenance of the pregnancy [200]. However, the significance is limited, and further research is necessary.
An altered immune milieu can also be caused by autoimmune responses referred to as autoimmune-related infertility. In this case, autoantibodies can be tested. This includes anti-cardiolipin antibodies, lupus anticoagulants, anti-β2-glykoprotein-I antibodies, anti-transglutaminase IgA, and anti-nuclear antibodies, which can be tested [199,201].
A ratio of TNFα+ or IFNγ+ and IL-10+ or IL-4+ T helper cells in the peripheral blood before treatment correlates with IVF success rate [202,203,204,205].

6.2.2. Interventions

Besides the various causes for an enhanced inflammatory/rejecting uterine micromilieu, it can be treated with glucocorticoids, intravenous infusion of phospholipid-stabilized soybean oil (intralipid), anti-TNFα, immunoglobulins [206], tacrolimus or heparin.

Glucocorticoids

Glucocorticoids are steroid hormones which are implicated in various processes. Although the level of glucocorticoids released under stress can compromise fertility, the right dose and timing of glucocorticoid release promote relevant reproductive functions [207]. Glucocorticoids also show immune modulatory effects. In general, glucocorticoids are potent immunosuppressors. Specifically in the uterus, NK cells [208] and macrophages [209,210] express the glucocorticoid receptor. By exposure to glucocorticoids, uterine NK cells decrease in their number [211] and show lowered cytotoxicity [212]. Accordingly, prednisolone decreases NK cell cytotoxicity in vitro [213]. Prednisone also binds TNFα according to in silico analysis inhibiting the inflammatory action of TNFα [214]. These changes create a rather tolerogenic milieu preventing sperm or fetal rejection. Besides the immunological changes, dexamethasone increases the survival and the prolactin secretion [14] as well as the IFGBP-1 secretion [215] of primary endometrial stroma cells in vitro.
However, the success of peri-implantation glucocorticoid administration is still under debate [216]. Although the administration of prednisolone significantly decreases the pathologically elevated numbers of uterine NK cells in patients with RIF, it must not improve the pregnancy rates after treatment [217]. Due to the temporal changes in the immune requirements during decidualization and early pregnancy, the need for general immune suppression must be carefully considered. However, diagnostic tools revealing the immune milieu are limited and not standardly used [218]. More research is needed to develop appropriate guidelines for the administration of glucocorticoids in artificial reproductive techniques (reviewed i.a. in [219]).

Intralipid

Fatty acids show an immune suppressive effect. Thus, soybean oil, which is the active component of intralipid, causes an immune suppressive effect. The exact mechanism of this modulatory capacity is not clearly understood. It inhibits pro-inflammatory Th1 cells and the cytotoxic activity of NK cells [220,221]. In patients with RIF, the perfusion with intralipid decreased the endometrial immune activation [222], supporting a rather tolerogenic milieu. The success of intralipid has been summarized in several reviews (i.a., [223,224,225,226,227,228,229]). However, conflicting studies raise doubts on the effectiveness. In peripheral blood, a rather pro-inflammatory shift towards cytotoxic T cells was observed after intralipid treatment [230]. Other studies did not find an improvement in pregnancy and birth rates [231]. Further research is suggested in order to investigate the success of the administration of intralipid [225,232].

Tumor Necrosis Factor α Inhibition

TNFα is the major effector cytokine of the inflammatory TH1 immune responses. It shows a pleiotropic effect on various cell types and is especially involved in autoimmune responses. Immune as well as non-immune cells express the TNFα receptors and are affected by this cytokine [233]. Blocking or neutralizing antibodies (Adalimumab, Humira; Infliximab, Remicade; Certolizumab pegol, Cimzia; Golimumab, Simponi) or fusion proteins (Etanercept, Enbrel) against TNFα or its receptors prevent its inflammatory effector functions and can support a tolerogenic micromilieu [234].
In patients with RIF, etanercepts improve pregnancy and live birth rate [235].
Especially in subfertile women with elevated TNFα:IL-10 ratios concerning the T helper cells in peripheral blood [236] or increased peripheral NK cell numbers [237], anti-TNFα binding therapy decreased the inflammatory parameters and thereby increased the pregnancy and live birth rate.

Intravenous Immunoglobulin

The action of intravenous immunoglobulin is a result of a variety of mechanisms. Polyclonal immunoglobulin G (IgG) substitutes pathologic autoantibodies. It prevents the activation of antigen-presenting cells and shifts the T cell balance towards regulatory T helper cells. In sum, it downregulates the production of pro-inflammatory cytokines and supports a rather tolerogenic or balanced immune milieu [238,239,240]. Moreover, immunoglobulins suppress NK cell cytotoxicity in vitro [213].
The usage of IVIG in RIF and RPL can support fertility [241], especially in patients with known inflammatory pathologies, including NK cell changes in count or cytotoxicity [242,243,244,245] and Th1:Th2 ratio [240,246] (reviewed in [247,248,249]). In couples with recurrent IVF failure and HLA similarity, IVIG might also increase the chances of pregnancy [250], suggesting a rather immune-balancing than only tolerance-mediating effect of IVIG.

Tacrolimus

Tacrolimus is a calcineurin inhibitor, which is used to prevent organ rejection in transplant patients. Calcineurin inhibitors prevent the production of IL-2. IL-2 is a crucial autocrine signal in T cell development and proliferation. Thus, the treatment with tacrolimus prevents T cell-mediated inflammatory responses and increases anti-inflammatory cytokines [251]. Thus, in RIF patients with elevated Th1:Th2 ratio, tacrolimus improves the pregnancy and live birth rate [252].

Heparin

In addition to the beneficial effects on the decidualization of stromal cells, increasing the secretion of IGFBP-1 and prolactin [253], heparin also favors a regulatory T cell response [254], which might support the tolerogenic immune milieu. Heparin shifts the endometrial cytokines towards and implantation-supporting milieu by increasing the expression of IL-6 and G-CSF [255]. Moreover, heparin inhibits the activity of the inflammatory transcription factor NF-κB in endometrial stroma cells [256]. Thereby, heparin improves the live birth rate [257] in RIF [258] and RPL [259] patients and decrease in adverse pregnancy outcomes [260]. There are also reports which did not find any improvements due to heparin [261,262,263,264]. This could also indicate that the patients for whom this treatment is eligible or the intervention itself needs to be defined more precisely.

Granulocyte Colony Stimulating Factor

Granulocyte colony-stimulating factor (G-CSF; CSF3) is injected either subcutaneously or intrauterine. Locally it might improve endometrial receptivity, implantation processes, and angiogenesis. Thus, G-CSF can increase the live birth rate in patients undergoing IVF [265,266,267]. Although the exact mechanisms remain unclear, it is known that G-CSF is also produced during implantation. Moreover, in the decidua, the expression of its receptor increases pre-ovulatory. G-CSF signaling is involved in proliferation and differentiation and affects the Th2 cytokines and shifts the T helper cell balance towards regulatory responses. G-CSF is a strong inhibitor of cytotoxic NK cell function [268], which is necessary for the uterine receptive milieu. The success of G-CSF in increasing pregnancy rate has been reviewed in detail (i.a., in [267,269,270]). Although not all studies found an improving effect of intrauterine perfusion of G-CSF [271,272,273]. Thus, more research is necessary in order to define the working administration and patient group.

Intrauterine Injection of hCG

The intrauterine injection of hCG before intrauterine insemination (IUI) or embryo transfer (ET) can also shift the local balance towards a receptive, tolerogenic environment. However, several studies showed contradictory results [274]. It is speculated that this intervention only helps a certain group of patients which needs to be specified in further studies. The hCG priming of the leukocytes shifts their immune response to a rather implantation-supporting and tolerogenic phenotype. In patients with RIF, intrauterine administration of hCG increases the percentage of Tregs while improving the live birth rate [275]. Another option is to prime autologous peripheral blood mononuclear cells (PBMCs) with hCG ex vivo and re-inject them into the uterine cavity. This procedure increases the live birth rate in patients with RIF [276]. Despite the promising results, more research is needed [276,277,278].

Seminal Plasma and Paternal Antigens

Independently of fertilization and the fertile window, unprotected sexual intercourse shifts the local immune balance towards tolerance independently of the fertile window. The immunoregulatory components of seminal plasma affect the local cells [279,280].
There are more options to target the optimal balance between tolerance and immune activation against the paternal antigens. The induction of tolerance to paternal antigens is one factor which explains the correlation of the frequency of sexual intercourse with the conceiving rate. Similarly, the induction of mucosal tolerogenic immunity might also explain the increased conceiving rate by the exposure to semen by unprotected oral sex. As a therapeutic tool, active immunization with partner antigens became is suggested. Through active immunization with partner lymphocytes, the maternal immune system is aimed to get familiar and trained with the paternal antigens [281]. The immunological mechanisms are not clearly understood. It is speculated that the immune reaction against the paternal antigens is enough activating in terms of the production of anti-paternal cytotoxic antibodies (APCA). These antibodies, although with cytotoxic potential, are negatively correlated with recurrent spontaneous abortion (RSA) [282]. Their presence after the immunization might explain the elevated pregnancy rates. Probably, as no adjuvants are used, no too inflammatory responses are caused, which would induce rejection and harm fertility.
At the latest, with this example, it is striking that fertility is based on a fine balance between pro- and anti-inflammatory events.
Other conditions and interventions often indirectly affect systemic immune functions. For example, obesity creates a harmful systemic inflammatory milieu, whereas moderate physical exercise supports optimal systemic immune balance.
Besides these available options, there are barely coherently standardized recommendations regarding immunomodulatory therapies currently.
Table 1. Overview of immune-targeting interventions to support sufficient decidualization.
Table 1. Overview of immune-targeting interventions to support sufficient decidualization.
DiagnosticsInterventionsObjectives
NK cell count
Plasma cell count
Treg cell count
Th1:Th2 ratios
[194,195,202,203,204,205]
Glucocorticoids [219], intrauterine application of phospholipid-stabilized soybean oil [225,232], anti-TNFα [235,236,237], hCG infusion [276,277,278], immunoglobulins [247,248,249], tacrolimus [252], heparin [257,258,259], G-CSF [267,269,270]
Immunization with partner
antigens



Balanced tolerogenic
Micromilieu



Balanced inflammatory micromilieu
Microbiome
[197]
Antibiotics, Pre- and
Probiotics
Modify colonizers
Recurrent implantation
failure
Thin endometrial lining
Scratching [171,172,173,174,175,176,177,178],
PRP infusion [183,184,185,186,187,188], G-CSF [267,269,270]
Wound-healing-like
Decidualization,
Regeneration

7. Summary and Outlook

The invasive implantation to build a hemochorial placenta in humans brings the fetal tissue in close contact with maternal tissue and immune cells. Instead of passive immunological ignorance, active tolerance is mediated by about 40–50% of the decidual cells, which are leukocytes. In order to avoid the rejection of the paternal antigens, these tolerance mechanisms actively create a tolerogenic niche. Decidual leukocytes, therefore, differ from their peripheral pendants.
Besides preventing rejection, decidual leukocytes support trophoblast invasion, tissue remodeling, and angiogenesis in order to build a sufficient placenta. These processes require locally and temporally limited inflammatory conditions. These are not comparable to the inflammatory conditions during inflammation which can cause vast destruction and, in the context of pregnancy, the rejection of the foreign structures, including the onset of labor resulting in abortions and pre-term labor. Thus, the decidual leukocytes must be optimal balanced to support pregnancy establishment, development, and maintenance (see Figure 2).
Although the immune components of the decidua and their relevance for pregnancy are known, translational routine implementations are lacking or are expandable. Further research is necessary to examine the actual pathologies, the effects of the interventions, and which diagnostics are necessary to find the suitable intervention for the individual patients. We suggest that attention to immunorelevant therapeutical interventions follow the rise of immunodiagnostics which are already available and find the recognition that it deserves in order to support the success of reproductive medicine.

Author Contributions

Conceptualization, M.B.S. and R.E.; investigation, M.B.S. and R.E.; writing—original draft preparation, M.B.S., A.M., N.S. and R.E.; writing—review and editing, M.B.S., A.M., N.S. and R.E. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Chavan, A.R.; Griffith, O.W.; Wagner, G.P. The inflammation paradox in the evolution of mammalian pregnancy: Turning a foe into a friend. Curr. Opin. Genet. Dev. 2017, 47, 24–32. [Google Scholar] [CrossRef] [PubMed]
  2. Liu, S.; Diao, L.; Huang, C.; Li, Y.; Zeng, Y.; Kwak-Kim, J.Y.H. The role of decidual immune cells on human pregnancy. J. Reprod. Immunol. 2017, 124, 44–53. [Google Scholar] [CrossRef] [PubMed]
  3. Pang, P.C.; Haslam, S.M.; Dell, A.; Clark, G.F. The human fetoembryonic defense system hypothesis: Twenty years on. Mol. Asp. Med. 2016, 51, 71–88. [Google Scholar] [CrossRef] [PubMed]
  4. Dutta, S.; Sengupta, P.; Haque, N. Reproductive immunomodulatory functions of B cells in pregnancy. Int. Rev. Immunol. 2020, 39, 53–66. [Google Scholar] [CrossRef] [PubMed]
  5. Jensen, F.; Muzzio, D.; Soldati, R.; Fest, S.; Zenclussen, A.C. Regulatory B10 cells restore pregnancy tolerance in a mouse model. Biol. Reprod. 2013, 89, 90. [Google Scholar] [CrossRef]
  6. 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]
  7. Ozturk, S.; Demir, R. Particular functions of estrogen and progesterone in establishment of uterine receptivity and embryo implantation. Histol. Histopathol. 2010, 25, 1215–1228. [Google Scholar] [CrossRef]
  8. Liao, H.Q.; Han, M.T.; Cheng, W.; Zhang, C.; Li, H.; Li, M.Q.; Zhu, R. Decidual-derived RANKL facilitates macrophages accumulation and residence at the maternal-fetal interface in human early pregnancy. Am. J. Reprod. Immunol. 2021, 86, e13406. [Google Scholar] [CrossRef]
  9. Kajimura, S.; Aida, K.; Duan, C. Insulin-like growth factor-binding protein-1 (IGFBP-1) mediates hypoxia-induced embryonic growth and developmental retardation. Proc. Natl. Acad. Sci. USA 2005, 102, 1240–1245. [Google Scholar] [CrossRef]
  10. Gordon, S.M. Interleukin-15 in Outcomes of Pregnancy. Int. J. Mol. Sci. 2021, 22, 11094. [Google Scholar] [CrossRef]
  11. Zhang, X.H.; Liang, X.; Liang, X.H.; Wang, T.S.; Qi, Q.R.; Deng, W.B.; Sha, A.G.; Yang, Z.M. The mesenchymal-epithelial transition during in vitro decidualization. Reprod. Sci. 2013, 20, 354–360. [Google Scholar] [CrossRef]
  12. Afshar, Y.; Jeong, J.W.; Roqueiro, D.; DeMayo, F.; Lydon, J.; Radtke, F.; Radnor, R.; Miele, L.; Fazleabas, A. Notch1 mediates uterine stromal differentiation and is critical for complete decidualization in the mouse. FASEB J. 2012, 26, 282–294. [Google Scholar] [CrossRef]
  13. Camden, A.J.; Szwarc, M.M.; Chadchan, S.B.; DeMayo, F.J.; O’Malley, B.W.; Lydon, J.P.; Kommagani, R. Growth regulation by estrogen in breast cancer 1 (GREB1) is a novel progesterone-responsive gene required for human endometrial stromal decidualization. Mol. Hum. Reprod. 2017, 23, 646–653. [Google Scholar] [CrossRef]
  14. Freis, A.; Renke, T.; Kammerer, U.; Jauckus, J.; Strowitzki, T.; Germeyer, A. Effects of a hyperandrogenaemic state on the proliferation and decidualization potential in human endometrial stromal cells. Arch. Gynecol. Obstet. 2017, 295, 1005–1013. [Google Scholar] [CrossRef]
  15. Yang, H.; Zhou, Y.; Edelshain, B.; Schatz, F.; Lockwood, C.J.; Taylor, H.S. FKBP4 is regulated by HOXA10 during decidualization and in endometriosis. Reproduction 2012, 143, 531–538. [Google Scholar] [CrossRef]
  16. Macdonald, L.J.; Sales, K.J.; Grant, V.; Brown, P.; Jabbour, H.N.; Catalano, R.D. Prokineticin 1 induces Dickkopf 1 expression and regulates cell proliferation and decidualization in the human endometrium. Mol. Hum. Reprod. 2011, 17, 626–636. [Google Scholar] [CrossRef]
  17. Adams, N.R.; Vasquez, Y.M.; Mo, Q.; Gibbons, W.; Kovanci, E.; DeMayo, F.J. WNK lysine deficient protein kinase 1 regulates human endometrial stromal cell decidualization, proliferation, and migration in part through mitogen-activated protein kinase 7. Biol. Reprod. 2017, 97, 400–412. [Google Scholar] [CrossRef]
  18. Voorzanger-Rousselot, N.; Goehrig, D.; Journe, F.; Doriath, V.; Body, J.J.; Clezardin, P.; Garnero, P. Increased Dickkopf-1 expression in breast cancer bone metastases. Br. J. Cancer 2007, 97, 964–970. [Google Scholar] [CrossRef]
  19. Abir, R.; Ao, A.; Zhang, X.Y.; Garor, R.; Nitke, S.; Fisch, B. Vascular endothelial growth factor A and its two receptors in human preantral follicles from fetuses, girls, and women. Fertil. Steril. 2010, 93, 2337–2347. [Google Scholar] [CrossRef]
  20. Boutros, T.; Chevet, E.; Metrakos, P. Mitogen-activated protein (MAP) kinase/MAP kinase phosphatase regulation: Roles in cell growth, death, and cancer. Pharmacol. Rev. 2008, 60, 261–310. [Google Scholar] [CrossRef]
  21. Wang, J.; Fu, L.; Gu, F.; Ma, Y. Notch1 is involved in migration and invasion of human breast cancer cells. Oncol. Rep. 2011, 26, 1295–1303. [Google Scholar] [CrossRef] [PubMed]
  22. Chu, M.C.; Selam, F.B.; Taylor, H.S. HOXA10 regulates p53 expression and matrigel invasion in human breast cancer cells. Cancer Biol. Ther. 2004, 3, 568–572. [Google Scholar] [CrossRef] [PubMed]
  23. Petroff, M.G.; Chen, L.; Phillips, T.A.; Azzola, D.; Sedlmayr, P.; Hunt, J.S. B7 family molecules are favorably positioned at the human maternal-fetal interface. Biol. Reprod. 2003, 68, 1496–1504. [Google Scholar] [CrossRef] [PubMed]
  24. Zhang, D.; Yu, Y.; Ding, C.; Zhang, R.; Duan, T.; Zhou, Q. Decreased B7-H3 promotes unexplained recurrent miscarriage via RhoA/ROCK2 signaling pathway and regulates the secretion of decidual NK cellsdagger. Biol. Reprod. 2023, 108, 504–518. [Google Scholar] [CrossRef]
  25. Krstic, J.; Deutsch, A.; Fuchs, J.; Gauster, M.; Gorsek Sparovec, T.; Hiden, U.; Krappinger, J.C.; Moser, G.; Pansy, K.; Szmyra, M.; et al. (Dis)similarities between the Decidual and Tumor Microenvironment. Biomedicines 2022, 10, 1065. [Google Scholar] [CrossRef]
  26. Gonzalez, H.; Hagerling, C.; Werb, Z. Roles of the immune system in cancer: From tumor initiation to metastatic progression. Genes Dev. 2018, 32, 1267–1284. [Google Scholar] [CrossRef]
  27. Zhou, J.; Tang, Z.; Gao, S.; Li, C.; Feng, Y.; Zhou, X. Tumor-Associated Macrophages: Recent Insights and Therapies. Front. Oncol. 2020, 10, 188. [Google Scholar] [CrossRef]
  28. Sethi, B.K.; Chanukya, G.V.; Nagesh, V.S. Prolactin and cancer: Has the orphan finally found a home? Indian J. Endocrinol. Metab. 2012, 16, S195–S198. [Google Scholar] [CrossRef]
  29. Ng, S.W.; Norwitz, G.A.; Pavlicev, M.; Tilburgs, T.; Simon, C.; Norwitz, E.R. Endometrial Decidualization: The Primary Driver of Pregnancy Health. Int. J. Mol. Sci. 2020, 21, 4092. [Google Scholar] [CrossRef]
  30. Erlebacher, A. Immunology of the maternal-fetal interface. Annu. Rev. Immunol. 2013, 31, 387–411. [Google Scholar] [CrossRef]
  31. Pollheimer, J.; Vondra, S.; Baltayeva, J.; Beristain, A.G.; Knofler, M. Regulation of Placental Extravillous Trophoblasts by the Maternal Uterine Environment. Front. Immunol. 2018, 9, 2597. [Google Scholar] [CrossRef]
  32. Helige, C.; Ahammer, H.; Moser, G.; Hammer, A.; Dohr, G.; Huppertz, B.; Sedlmayr, P. Distribution of decidual natural killer cells and macrophages in the neighbourhood of the trophoblast invasion front: A quantitative evaluation. Hum. Reprod. 2014, 29, 8–17. [Google Scholar] [CrossRef]
  33. Jabrane-Ferrat, N. Features of Human Decidual NK Cells in Healthy Pregnancy and during Viral Infection. Front. Immunol. 2019, 10, 1397. [Google Scholar] [CrossRef]
  34. Dengler, V.L.; Galbraith, M.; Espinosa, J.M. Transcriptional regulation by hypoxia inducible factors. Crit. Rev. Biochem. Mol. Biol. 2014, 49, 1–15. [Google Scholar] [CrossRef]
  35. Bulmer, J.N.; Williams, P.J.; Lash, G.E. Immune cells in the placental bed. Int. J. Dev. Biol. 2010, 54, 281–294. [Google Scholar] [CrossRef]
  36. Trundley, A.; Moffett, A. Human uterine leukocytes and pregnancy. Tissue Antigens 2004, 63, 1–12. [Google Scholar] [CrossRef]
  37. Bartmann, C.; Segerer, S.E.; Rieger, L.; Kapp, M.; Sutterlin, M.; Kammerer, U. Quantification of the predominant immune cell populations in decidua throughout human pregnancy. Am. J. Reprod. Immunol. 2014, 71, 109–119. [Google Scholar] [CrossRef]
  38. Miller, D.; Motomura, K.; Garcia-Flores, V.; Romero, R.; Gomez-Lopez, N. Innate Lymphoid Cells in the Maternal and Fetal Compartments. Front. Immunol. 2018, 9, 2396. [Google Scholar] [CrossRef]
  39. Wang, P.; Liang, T.; Zhan, H.; Zhu, M.; Wu, M.; Qian, L.; Zhou, Y.; Ni, F. Unique metabolism and protein expression signature in human decidual NK cells. Front. Immunol. 2023, 14, 1136652. [Google Scholar] [CrossRef]
  40. Keskin, D.B.; Allan, D.S.; Rybalov, B.; Andzelm, M.M.; Stern, J.N.; Kopcow, H.D.; Koopman, L.A.; Strominger, J.L. TGFbeta promotes conversion of CD16+ peripheral blood NK cells into CD16 NK cells with similarities to decidual NK cells. Proc. Natl. Acad. Sci. USA 2007, 104, 3378–3383. [Google Scholar] [CrossRef]
  41. Cerdeira, A.S.; Rajakumar, A.; Royle, C.M.; Lo, A.; Husain, Z.; Thadhani, R.I.; Sukhatme, V.P.; Karumanchi, S.A.; Kopcow, H.D. Conversion of peripheral blood NK cells to a decidual NK-like phenotype by a cocktail of defined factors. J. Immunol. 2013, 190, 3939–3948. [Google Scholar] [CrossRef] [PubMed]
  42. Andreescu, M.; Frincu, F.; Plotogea, M.; Mehedintu, C. Recurrent Abortion and the Involvement of Killer-Cell Immunoglobulin-like Receptor (KIR) Genes, Activated T Cells, NK Abnormalities, and Cytokine Profiles. J. Clin. Med. 2023, 12, 1355. [Google Scholar] [CrossRef] [PubMed]
  43. Ferreira, L.M.R.; Meissner, T.B.; Tilburgs, T.; Strominger, J.L. HLA-G: At the Interface of Maternal-Fetal Tolerance. Trends Immunol. 2017, 38, 272–286. [Google Scholar] [CrossRef] [PubMed]
  44. van der Meer, A.; Lukassen, H.G.; van Lierop, M.J.; Wijnands, F.; Mosselman, S.; Braat, D.D.; Joosten, I. Membrane-bound HLA-G activates proliferation and interferon-gamma production by uterine natural killer cells. Mol. Hum. Reprod. 2004, 10, 189–195. [Google Scholar] [CrossRef] [PubMed]
  45. Li, C.; Houser, B.L.; Nicotra, M.L.; Strominger, J.L. HLA-G homodimer-induced cytokine secretion through HLA-G receptors on human decidual macrophages and natural killer cells. Proc. Natl. Acad. Sci. USA 2009, 106, 5767–5772. [Google Scholar] [CrossRef]
  46. Hanna, J.; Goldman-Wohl, D.; Hamani, Y.; Avraham, I.; Greenfield, C.; Natanson-Yaron, S.; Prus, D.; Cohen-Daniel, L.; Arnon, T.I.; Manaster, I.; et al. Decidual NK cells regulate key developmental processes at the human fetal-maternal interface. Nat. Med. 2006, 12, 1065–1074. [Google Scholar] [CrossRef]
  47. De Oliveira, L.G.; Lash, G.E.; Murray-Dunning, C.; Bulmer, J.N.; Innes, B.A.; Searle, R.F.; Sass, N.; Robson, S.C. Role of interleukin 8 in uterine natural killer cell regulation of extravillous trophoblast cell invasion. Placenta 2010, 31, 595–601. [Google Scholar] [CrossRef]
  48. Robson, A.; Harris, L.K.; Innes, B.A.; Lash, G.E.; Aljunaidy, M.M.; Aplin, J.D.; Baker, P.N.; Robson, S.C.; Bulmer, J.N. Uterine natural killer cells initiate spiral artery remodeling in human pregnancy. FASEB J. 2012, 26, 4876–4885. [Google Scholar] [CrossRef]
  49. Takahashi, H.; Yamamoto, T.; Yamazaki, M.; Murase, T.; Matsuno, T.; Chishima, F. Natural Cytotoxicity Receptors in Decidua Natural Killer Cells of Term Normal Pregnancy. J. Pregnancy 2018, 2018, 4382084. [Google Scholar] [CrossRef]
  50. Du, M.R.; Wang, S.C.; Li, D.J. The integrative roles of chemokines at the maternal-fetal interface in early pregnancy. Cell. Mol. Immunol. 2014, 11, 438–448. [Google Scholar] [CrossRef]
  51. Zhang, X.; Wei, H. Role of Decidual Natural Killer Cells in Human Pregnancy and Related Pregnancy Complications. Front. Immunol. 2021, 12, 728291. [Google Scholar] [CrossRef] [PubMed]
  52. Rao, V.A.; Kurian, N.K.; Rao, K.A. Cytokines, NK cells and regulatory T cell functions in normal pregnancy and reproductive failures. Am. J. Reprod. Immunol. 2023, 89, e13667. [Google Scholar] [CrossRef] [PubMed]
  53. Vacca, P.; Montaldo, E.; Croxatto, D.; Loiacono, F.; Canegallo, F.; Venturini, P.L.; Moretta, L.; Mingari, M.C. Identification of diverse innate lymphoid cells in human decidua. Mucosal Immunol. 2015, 8, 254–264. [Google Scholar] [CrossRef] [PubMed]
  54. Doisne, J.M.; Balmas, E.; Boulenouar, S.; Gaynor, L.M.; Kieckbusch, J.; Gardner, L.; Hawkes, D.A.; Barbara, C.F.; Sharkey, A.M.; Brady, H.J.; et al. Composition, Development, and Function of Uterine Innate Lymphoid Cells. J. Immunol. 2015, 195, 3937–3945. [Google Scholar] [CrossRef] [PubMed]
  55. Einenkel, R.; Ehrhardt, J.; Hartmann, K.; Kruger, D.; Muzzio, D.O.; Zygmunt, M. Hormonally controlled ILC antigen presentation potential is reduced during pregnancy. Reproduction 2020, 160, 155–169. [Google Scholar] [CrossRef]
  56. Xu, Y.; Romero, R.; Miller, D.; Silva, P.; Panaitescu, B.; Theis, K.R.; Arif, A.; Hassan, S.S.; Gomez-Lopez, N. Innate lymphoid cells at the human maternal-fetal interface in spontaneous preterm labor. Am. J. Reprod. Immunol. 2018, 79, e12820. [Google Scholar] [CrossRef]
  57. Vazquez, J.; Chasman, D.A.; Lopez, G.E.; Tyler, C.T.; Ong, I.M.; Stanic, A.K. Transcriptional and Functional Programming of Decidual Innate Lymphoid Cells. Front. Immunol. 2019, 10, 3065. [Google Scholar] [CrossRef]
  58. Vivier, E.; Artis, D.; Colonna, M.; Diefenbach, A.; Di Santo, J.P.; Eberl, G.; Koyasu, S.; Locksley, R.M.; McKenzie, A.N.J.; Mebius, R.E.; et al. Innate Lymphoid Cells: 10 Years on. Cell 2018, 174, 1054–1066. [Google Scholar] [CrossRef]
  59. Vacca, P.; Vitale, C.; Munari, E.; Cassatella, M.A.; Mingari, M.C.; Moretta, L. Human Innate Lymphoid Cells: Their Functional and Cellular Interactions in Decidua. Front. Immunol. 2018, 9, 1897. [Google Scholar] [CrossRef]
  60. Pongcharoen, S.; Niumsup, P.; Sanguansermsri, D.; Supalap, K.; Butkhamchot, P. The effect of interleukin-17 on the proliferation and invasion of JEG-3 human choriocarcinoma cells. Am. J. Reprod. Immunol. 2006, 55, 291–300. [Google Scholar] [CrossRef]
  61. Wang, Y.; Xu, B.; Li, M.Q.; Li, D.J.; Jin, L.P. IL-22 secreted by decidual stromal cells and NK cells promotes the survival of human trophoblasts. Int. J. Clin. Exp. Pathol. 2013, 6, 1781–1790. [Google Scholar]
  62. O’Hern Perfetto, C.; Fan, X.; Dahl, S.; Krieg, S.; Westphal, L.M.; Bunker Lathi, R.; Nayak, N.R. Expression of interleukin-22 in decidua of patients with early pregnancy and unexplained recurrent pregnancy loss. J. Assist. Reprod. Genet. 2015, 32, 977–984. [Google Scholar] [CrossRef]
  63. Perricone, R.; De Carolis, C.; Giacomelli, R.; Guarino, M.D.; De Sanctis, G.; Fontana, L. GM-CSF and pregnancy: Evidence of significantly reduced blood concentrations in unexplained recurrent abortion efficiently reverted by intravenous immunoglobulin treatment. Am. J. Reprod. Immunol. 2003, 50, 232–237. [Google Scholar] [CrossRef]
  64. Robertson, S.A. GM-CSF regulation of embryo development and pregnancy. Cytokine Growth Factor Rev. 2007, 18, 287–298. [Google Scholar] [CrossRef]
  65. Einenkel, R.; Ehrhardt, J.; Zygmunt, M.; Muzzio, D.O. Oxygen regulates ILC3 antigen presentation potential and pregnancy-related hormone actions. Reprod. Biol. Endocrinol. 2022, 20, 109. [Google Scholar] [CrossRef]
  66. Jena, M.K.; Nayak, N.; Chen, K.; Nayak, N.R. Role of Macrophages in Pregnancy and Related Complications. Arch. Immunol. Ther. Exp. 2019, 67, 295–309. [Google Scholar] [CrossRef]
  67. Wynn, T.A.; Chawla, A.; Pollard, J.W. Macrophage biology in development, homeostasis and disease. Nature 2013, 496, 445–455. [Google Scholar] [CrossRef]
  68. Pollard, J.W. Trophic macrophages in development and disease. Nat. Rev. Immunol. 2009, 9, 259–270. [Google Scholar] [CrossRef]
  69. Wei, C.Y.; Li, M.Q.; Zhu, X.Y.; Li, D.J. Immune status of decidual macrophages is dependent on the CCL2/CCR2/JAK2 pathway during early pregnancy. Am. J. Reprod. Immunol. 2021, 86, e13480. [Google Scholar] [CrossRef]
  70. Brown, M.B.; von Chamier, M.; Allam, A.B.; Reyes, L. M1/M2 macrophage polarity in normal and complicated pregnancy. Front. Immunol. 2014, 5, 606. [Google Scholar] [CrossRef]
  71. Zhang, Y.H.; He, M.; Wang, Y.; Liao, A.H. Modulators of the Balance between M1 and M2 Macrophages during Pregnancy. Front. Immunol. 2017, 8, 120. [Google Scholar] [CrossRef] [PubMed]
  72. Jaiswal, M.K.; Mallers, T.M.; Larsen, B.; Kwak-Kim, J.; Chaouat, G.; Gilman-Sachs, A.; Beaman, K.D. V-ATPase upregulation during early pregnancy: A possible link to establishment of an inflammatory response during preimplantation period of pregnancy. Reproduction 2012, 143, 713–725. [Google Scholar] [CrossRef] [PubMed]
  73. Kabawat, S.E.; Mostoufi-Zadeh, M.; Driscoll, S.G.; Bhan, A.K. Implantation site in normal pregnancy. A study with monoclonal antibodies. Am. J. Pathol. 1985, 118, 76–84. [Google Scholar] [PubMed]
  74. Pan, Y.; Yang, L.; Chen, D.; Hou, H.; Zhang, M.; Chen, M.; Ning, F.; Lu, Q.; Zhao, M.; Li, L.; et al. Decidual macrophage derived MMP3 contributes to extracellular matrix breakdown in spiral artery remodeling in early human pregnancy. J. Reprod. Immunol. 2022, 150, 103494. [Google Scholar] [CrossRef]
  75. Sun, F.; Wang, S.; Du, M. Functional regulation of decidual macrophages during pregnancy. J. Reprod. Immunol. 2021, 143, 103264. [Google Scholar] [CrossRef]
  76. Lash, G.E.; Pitman, H.; Morgan, H.L.; Innes, B.A.; Agwu, C.N.; Bulmer, J.N. Decidual macrophages: Key regulators of vascular remodeling in human pregnancy. J. Leukoc. Biol. 2016, 100, 315–325. [Google Scholar] [CrossRef]
  77. Ding, J.; Yang, C.; Zhang, Y.; Wang, J.; Zhang, S.; Guo, D.; Yin, T.; Yang, J. M2 macrophage-derived G-CSF promotes trophoblasts EMT, invasion and migration via activating PI3K/Akt/Erk1/2 pathway to mediate normal pregnancy. J. Cell. Mol. Med. 2021, 25, 2136–2147. [Google Scholar] [CrossRef]
  78. Sunderkotter, C.; Steinbrink, K.; Goebeler, M.; Bhardwaj, R.; Sorg, C. Macrophages and angiogenesis. J. Leukoc. Biol. 1994, 55, 410–422. [Google Scholar] [CrossRef]
  79. Mor, G.; Abrahams, V.M. Potential role of macrophages as immunoregulators of pregnancy. Reprod. Biol. Endocrinol. 2003, 1, 119. [Google Scholar] [CrossRef]
  80. Yao, Y.; Xu, X.H.; Jin, L. Macrophage Polarization in Physiological and Pathological Pregnancy. Front. Immunol. 2019, 10, 792. [Google Scholar] [CrossRef]
  81. Zhang, Y.H.; Aldo, P.; You, Y.; Ding, J.; Kaislasuo, J.; Petersen, J.F.; Lokkegaard, E.; Peng, G.; Paidas, M.J.; Simpson, S.; et al. Trophoblast-secreted soluble-PD-L1 modulates macrophage polarization and function. J. Leukoc. Biol. 2020, 108, 983–998. [Google Scholar] [CrossRef]
  82. Svensson-Arvelund, J.; Mehta, R.B.; Lindau, R.; Mirrasekhian, E.; Rodriguez-Martinez, H.; Berg, G.; Lash, G.E.; Jenmalm, M.C.; Ernerudh, J. The human fetal placenta promotes tolerance against the semiallogeneic fetus by inducing regulatory T cells and homeostatic M2 macrophages. J. Immunol. 2015, 194, 1534–1544. [Google Scholar] [CrossRef]
  83. Aldo, P.B.; Racicot, K.; Craviero, V.; Guller, S.; Romero, R.; Mor, G. Trophoblast induces monocyte differentiation into CD14+/CD16+ macrophages. Am. J. Reprod. Immunol. 2014, 72, 270–284. [Google Scholar] [CrossRef]
  84. Wang, X.Q.; Zhou, W.J.; Hou, X.X.; Fu, Q.; Li, D.J. Correction: Trophoblast-derived CXCL16 induces M2 macrophage polarization that in turn inactivates NK cells at the maternal-fetal interface. Cell. Mol. Immunol. 2019, 16, 313. [Google Scholar] [CrossRef]
  85. Parasar, P.; Guru, N.; Nayak, N.R. Contribution of macrophages to fetomaternal immunological tolerance. Hum. Immunol. 2021, 82, 325–331. [Google Scholar] [CrossRef]
  86. Houser, B.L. Decidual macrophages and their roles at the maternal-fetal interface. Yale J. Biol. Med. 2012, 85, 105–118. [Google Scholar]
  87. Heikkinen, J.; Mottonen, M.; Komi, J.; Alanen, A.; Lassila, O. Phenotypic characterization of human decidual macrophages. Clin. Exp. Immunol. 2003, 131, 498–505. [Google Scholar] [CrossRef]
  88. Unal, E.R.; Cierny, J.T.; Roedner, C.; Newman, R.; Goetzl, L. Maternal inflammation in spontaneous term labor. Am. J. Obstet. Gynecol. 2011, 204, 223.E1–223.E5. [Google Scholar] [CrossRef]
  89. Hamilton, S.; Oomomian, Y.; Stephen, G.; Shynlova, O.; Tower, C.L.; Garrod, A.; Lye, S.J.; Jones, R.L. Macrophages infiltrate the human and rat decidua during term and preterm labor: Evidence that decidual inflammation precedes labor. Biol. Reprod. 2012, 86, 39. [Google Scholar] [CrossRef]
  90. Gardner, L.; Moffett, A. Dendritic cells in the human decidua. Biol. Reprod. 2003, 69, 1438–1446. [Google Scholar] [CrossRef]
  91. Tagliani, E.; Erlebacher, A. Dendritic cell function at the maternal-fetal interface. Expert Rev. Clin. Immunol. 2011, 7, 593–602. [Google Scholar] [CrossRef] [PubMed]
  92. Tilburgs, T.; van der Mast, B.J.; Nagtzaam, N.M.; Roelen, D.L.; Scherjon, S.A.; Claas, F.H. Expression of NK cell receptors on decidual T cells in human pregnancy. J. Reprod. Immunol. 2009, 80, 22–32. [Google Scholar] [CrossRef]
  93. Wang, S.; Sun, F.; Li, M.; Qian, J.; Chen, C.; Wang, M.; Zang, X.; Li, D.; Yu, M.; Du, M. The appropriate frequency and function of decidual Tim-3+CTLA-4+CD8+ T cells are important in maintaining normal pregnancy. Cell Death Dis. 2019, 10, 407. [Google Scholar] [CrossRef] [PubMed]
  94. Muzzio, D.; Zenclussen, A.C.; Jensen, F. The role of B cells in pregnancy: The good and the bad. Am. J. Reprod. Immunol. 2013, 69, 408–412. [Google Scholar] [CrossRef] [PubMed]
  95. Kammerer, U.; Eggert, A.O.; Kapp, M.; McLellan, A.D.; Geijtenbeek, T.B.; Dietl, J.; van Kooyk, Y.; Kampgen, E. Unique appearance of proliferating antigen-presenting cells expressing DC-SIGN (CD209) in the decidua of early human pregnancy. Am. J. Pathol. 2003, 162, 887–896. [Google Scholar] [CrossRef]
  96. Gori, S.; Soczewski, E.; Fernandez, L.; Grasso, E.; Gallino, L.; Merech, F.; Colado, A.; Borge, M.; Perez Leiros, C.; Salamone, G.; et al. Decidualization Process Induces Maternal Monocytes to Tolerogenic IL-10-Producing Dendritic Cells (DC-10). Front. Immunol. 2020, 11, 1571. [Google Scholar] [CrossRef]
  97. Qin, D.; Xu, H.; Chen, Z.; Deng, X.; Jiang, S.; Zhang, X.; Bao, S. The peripheral and decidual immune cell profiles in women with recurrent pregnancy loss. Front. Immunol. 2022, 13, 994240. [Google Scholar] [CrossRef]
  98. Hou, R.; Huang, R.; Zhou, Y.; Lin, D.; Xu, J.; Yang, L.; Wei, X.; Xie, Z.; Zhou, Q. Single-cell profiling of the microenvironment in decidual tissue from women with missed abortions. Fertil. Steril. 2023, 119, 492–503. [Google Scholar] [CrossRef]
  99. Yang, X.; Tian, Y.; Zheng, L.; Luu, T.; Kwak-Kim, J. The Update Immune-Regulatory Role of Pro- and Anti-Inflammatory Cytokines in Recurrent Pregnancy Losses. Int. J. Mol. Sci. 2022, 24, 132. [Google Scholar] [CrossRef]
  100. Lai, N.; Fu, X.; Hei, G.; Song, W.; Wei, R.; Zhu, X.; Guo, Q.; Zhang, Z.; Chu, C.; Xu, K.; et al. The Role of Dendritic Cell Subsets in Recurrent Spontaneous Abortion and the Regulatory Effect of Baicalin on It. J. Immunol. Res. 2022, 2022, 9693064. [Google Scholar] [CrossRef]
  101. Esparvarinha, M.; Madadi, S.; Aslanian-Kalkhoran, L.; Nickho, H.; Dolati, S.; Pia, H.; Danaii, S.; Taghavi, S.; Yousefi, M. Dominant immune cells in pregnancy and pregnancy complications: T helper cells (TH1/TH2, TH17/Treg cells), NK cells, MDSCs, and the immune checkpoints. Cell Biol. Int. 2023, 47, 507–519. [Google Scholar] [CrossRef]
  102. Genest, G.; Banjar, S.; Almasri, W.; Beauchamp, C.; Benoit, J.; Buckett, W.; Dzineku, F.; Gold, P.; Dahan, M.H.; Jamal, W.; et al. Immunomodulation for unexplained recurrent implantation failure: Where are we now? Reproduction 2023, 165, R39–R60. [Google Scholar] [CrossRef]
  103. Mukherjee, N.; Sharma, R.; Modi, D. Immune alterations in recurrent implantation failure. Am. J. Reprod. Immunol. 2023, 89, e13563. [Google Scholar] [CrossRef]
  104. Pantos, K.; Grigoriadis, S.; Maziotis, E.; Pistola, K.; Xystra, P.; Pantou, A.; Kokkali, G.; Pappas, A.; Lambropoulou, M.; Sfakianoudis, K.; et al. The Role of Interleukins in Recurrent Implantation Failure: A Comprehensive Review of the Literature. Int. J. Mol. Sci. 2022, 23, 2198. [Google Scholar] [CrossRef]
  105. Franasiak, J.M.; Alecsandru, D.; Forman, E.J.; Gemmell, L.C.; Goldberg, J.M.; Llarena, N.; Margolis, C.; Laven, J.; Schoenmakers, S.; Seli, E. A review of the pathophysiology of recurrent implantation failure. Fertil. Steril. 2021, 116, 1436–1448. [Google Scholar] [CrossRef]
  106. Loeb, L. Über die experimentelle Erzeugung von Knoten von Deciduagewebe in dem Uterus des Meerschweinchens nach stattgefundener Copulation. Zent. Allg. Pathol. Pathol. Anat. 1907, 18, 563–565. [Google Scholar]
  107. Barash, A.; Dekel, N.; Fieldust, S.; Segal, I.; Schechtman, E.; Granot, I. Local injury to the endometrium doubles the incidence of successful pregnancies in patients undergoing in vitro fertilization. Fertil. Steril. 2003, 79, 1317–1322. [Google Scholar] [CrossRef]
  108. Karow, W.G.; Gentry, W.C.; Skeels, R.F.; Payne, S.A. Endometrial biopsy in the luteal phase of the cycle of conception. Fertil. Steril. 1971, 22, 482–495. [Google Scholar] [CrossRef]
  109. Han, X.; Hu, L. The effect of endometrial scratch on pregnancy outcomes of frozen-thawed embryo transfer: A propensity score-matched study. Gynecol. Endocrinol. 2022, 38, 39–44. [Google Scholar] [CrossRef]
  110. Wang, Y.; Bu, Z.; Hu, L. Comparing the effects of endometrial injury in the luteal phase and follicular phase on in vitro fertilization treatment outcomes. Front. Endocrinol. 2022, 13, 1004265. [Google Scholar] [CrossRef]
  111. Ueno, J.; Salgado, R.M.; Ejzenberg, D.; Carvalho, F.M.H.; Veiga, E.C.A.; Soares Junior, J.M.; Baracat, E.C. Is the length of time between endometrial scratching and embryo transfer important for pregnancy success? An observational study. Rev. Assoc. Med. Bras. 2023, 69, 72–77. [Google Scholar] [CrossRef] [PubMed]
  112. Tamar, A.M.; Martha, L.R.; Carlos, H.N.; Deborah, C.B.; Benjamin, S. Hysteroscopic endometrial peeling as a different approach to endometrial scratching. Case series report. J. Gynecol. Obstet. Hum. Reprod. 2021, 50, 102195. [Google Scholar] [CrossRef] [PubMed]
  113. Mahran, A.; Ibrahim, M.; Bahaa, H. The effect of endometrial injury on first cycle IVF/ICSI outcome: A randomized controlled trial. Int. J. Reprod. Biomed. 2016, 14, 193–198. [Google Scholar] [CrossRef] [PubMed]
  114. Maged, A.M.; Rashwan, H.; AbdelAziz, S.; Ramadan, W.; Mostafa, W.A.I.; Metwally, A.A.; Katta, M. Randomized controlled trial of the effect of endometrial injury on implantation and clinical pregnancy rates during the first ICSI cycle. Int. J. Gynaecol. Obstet. 2018, 140, 211–216. [Google Scholar] [CrossRef]
  115. Madhuri, M.S.; Thyagaraju, C.; Naidu, A.; Dasari, P. The effect of endometrial scratching on pregnancy rate after failed intrauterine insemination: A Randomised Controlled Trail. Eur. J. Obstet. Gynecol. Reprod. Biol. 2022, 268, 37–42. [Google Scholar] [CrossRef]
  116. Liang, Y.; Han, J.; Jia, C.; Ma, Y.; Lan, Y.; Li, Y.; Wang, S. Effect of Endometrial Injury on Secretion of Endometrial Cytokines and IVF Outcomes in Women with Unexplained Subfertility. Mediat. Inflamm. 2015, 2015, 757184. [Google Scholar] [CrossRef]
  117. Guven, S.; Kart, C.; Unsal, M.A.; Yildirim, O.; Odaci, E.; Yulug, E. Endometrial injury may increase the clinical pregnancy rate in normoresponders undergoing long agonist protocol ICSI cycles with single embryo transfer. Eur. J. Obstet. Gynecol. Reprod. Biol. 2014, 173, 58–62. [Google Scholar] [CrossRef]
  118. van Hoogenhuijze, N.E.; van Eekelen, R.; Mol, F.; Schipper, I.; Groenewoud, E.R.; Traas, M.A.F.; Janssen, C.A.H.; Teklenburg, G.; de Bruin, J.P.; van Oppenraaij, R.H.F.; et al. Economic evaluation of endometrial scratching before the second IVF/ICSI treatment: A cost-effectiveness analysis of a randomized controlled trial (SCRaTCH trial). Hum. Reprod. 2022, 37, 254–263. [Google Scholar] [CrossRef]
  119. Huang, S.Y.; Wang, C.J.; Soong, Y.K.; Wang, H.S.; Wang, M.L.; Lin, C.Y.; Chang, C.L. Site-specific endometrial injury improves implantation and pregnancy in patients with repeated implantation failures. Reprod. Biol. Endocrinol. 2011, 9, 140. [Google Scholar] [CrossRef]
  120. Gibreel, A.; Badawy, A.; El-Refai, W.; El-Adawi, N. Endometrial scratching to improve pregnancy rate in couples with unexplained subfertility: A randomized controlled trial. J. Obstet. Gynaecol. Res. 2013, 39, 680–684. [Google Scholar] [CrossRef]
  121. Kara, M.; Aydin, T.; Turktekin, N.; Karacavus, S. Efficacy of the local endometrial injury in patients who had previous failed IVF-ICSI outcome. Iran. J. Reprod. Med. 2012, 10, 567–570. [Google Scholar]
  122. Nastri, C.O.; Ferriani, R.A.; Raine-Fenning, N.; Martins, W.P. Endometrial scratching performed in the non-transfer cycle and outcome of assisted reproduction: A randomized controlled trial. Ultrasound Obstet. Gynecol. 2013, 42, 375–382. [Google Scholar] [CrossRef]
  123. Parsanezhad, M.E.; Dadras, N.; Maharlouei, N.; Neghahban, L.; Keramati, P.; Amini, M. Pregnancy rate after endometrial injury in couples with unexplained infertility: A randomized clinical trial. Iran. J. Reprod. Med. 2013, 11, 869–874. [Google Scholar]
  124. Singh, N.; Toshyan, V.; Kumar, S.; Vanamail, P.; Madhu, M. Does endometrial injury enhances implantation in recurrent in-vitro fertilization failures? A prospective randomized control study from tertiary care center. J. Hum. Reprod. Sci. 2015, 8, 218–223. [Google Scholar] [CrossRef]
  125. Kitaya, K.; Matsubayashi, H.; Takaya, Y.; Nishiyama, R.; Yamaguchi, K.; Ishikawa, T. Clinical background affecting pregnancy outcome following local endometrial injury in infertile patients with repeated implantation failure. Gynecol. Endocrinol. 2016, 32, 587–590. [Google Scholar] [CrossRef]
  126. Kanazawa, E.; Nakashima, A.; Yonemoto, K.; Otsuka, M.; Yoshioka, N.; Kuramoto, T.; Mitao, H.; Imaishi, H.; Komai, K.; Ushijima, K. Injury to the endometrium prior to the frozen-thawed embryo transfer cycle improves pregnancy rates in patients with repeated implantation failure. J. Obstet. Gynaecol. Res. 2017, 43, 128–134. [Google Scholar] [CrossRef]
  127. Siristatidis, C.; Kreatsa, M.; Koutlaki, N.; Galazios, G.; Pergialiotis, V.; Papantoniou, N. Endometrial injury for RIF patients undergoing IVF/ICSI: A prospective nonrandomized controlled trial. Gynecol. Endocrinol. 2017, 33, 297–300. [Google Scholar] [CrossRef]
  128. Reljic, M.; Knez, J.; Kovac, V.; Kovacic, B. Endometrial injury, the quality of embryos, and blastocyst transfer are the most important prognostic factors for in vitro fertilization success after previous repeated unsuccessful attempts. J. Assist. Reprod. Genet. 2017, 34, 775–779. [Google Scholar] [CrossRef]
  129. Helmy, M.E.E.; Maher, M.A.; Elkhouly, N.I.; Ramzy, M. A randomized trial of local endometrial injury during ovulation induction cycles. Int. J. Gynaecol. Obstet. 2017, 138, 47–52. [Google Scholar] [CrossRef]
  130. Olesen, M.S.; Hauge, B.; Ohrt, L.; Olesen, T.N.; Roskaer, J.; Baek, V.; Elbaek, H.O.; Nohr, B.; Nyegaard, M.; Overgaard, M.T.; et al. Therapeutic endometrial scratching and implantation after in vitro fertilization: A multicenter randomized controlled trial. Fertil. Steril. 2019, 112, 1015–1021. [Google Scholar] [CrossRef]
  131. Tang, Z.; Hong, M.; He, F.; Huang, D.; Dai, Z.; Xuan, H.; Zhang, H.; Zhu, W. Effect of endometrial injury during menstruation on clinical outcomes in frozen-thawed embryo transfer cycles: A randomized control trial. J. Obstet. Gynaecol. Res. 2020, 46, 451–458. [Google Scholar] [CrossRef] [PubMed]
  132. Chen, T.; Shi, H.; Fang, L.L.; Su, Y.C. The effect of endometrial injury on reproductive outcomes of frozen-thawed embryo transfer cycles in women with one implantation failure. J. Int. Med. Res. 2020, 48, 300060520913130. [Google Scholar] [CrossRef] [PubMed]
  133. Acet, F.; Sahin, G.; Goker, E.N.T.; Tavmergen, E. The effect of hysteroscopy and conventional curretage versus no hysteroscopy on live birth rates in recurrent in vitro fertilisation failure: A retrospective cohort study from a single referral centre experience. J. Obstet. Gynaecol. 2022, 42, 2134–2138. [Google Scholar] [CrossRef] [PubMed]
  134. Turktekin, N.; Karakus, C.; Ozyurt, R. Comparing the effects of endometrial injury with hysteroscopy or Pipelle cannula on fertility outcome. Eur. Rev. Med. Pharmacol. Sci. 2022, 26, 4693–4697. [Google Scholar] [CrossRef] [PubMed]
  135. Coughlan, C.; Yuan, X.; Demirol, A.; Ledger, W.; Li, T.C. Factors affecting the outcome of “endometrial scratch” in women with recurrent implantation failure. J. Reprod. Med. 2014, 59, 39–43. [Google Scholar]
  136. Maged, A.M.; Al-Inany, H.; Salama, K.M.; Souidan, I.I.; Abo Ragab, H.M.; Elnassery, N. Endometrial Scratch Injury Induces Higher Pregnancy Rate for Women with Unexplained Infertility Undergoing IUI with Ovarian Stimulation: A Randomized Controlled Trial. Reprod. Sci. 2016, 23, 239–243. [Google Scholar] [CrossRef]
  137. Bahaa Eldin, A.M.; Abdelmaabud, K.H.; Laban, M.; Hassanin, A.S.; Tharwat, A.A.; Aly, T.R.; Elbohoty, A.E.; Elsayed, H.M.; Ibrahim, A.M.; Ibrahim, M.E.; et al. Endometrial Injury May Increase the Pregnancy Rate in Patients Undergoing Intrauterine Insemination: An Interventional Randomized Clinical Trial. Reprod. Sci. 2016, 23, 1326–1331. [Google Scholar] [CrossRef]
  138. Taneja, J.; Ogutu, D.; Ah-Moye, M. Rare successful pregnancy in a patient with Swyer Syndrome. Case Rep. Women’s Health 2016, 12, 1–2. [Google Scholar] [CrossRef]
  139. van Hoogenhuijze, N.E.; Mol, F.; Laven, J.S.E.; Groenewoud, E.R.; Traas, M.A.F.; Janssen, C.A.H.; Teklenburg, G.; de Bruin, J.P.; van Oppenraaij, R.H.F.; Maas, J.W.M.; et al. Endometrial scratching in women with one failed IVF/ICSI cycle-outcomes of a randomised controlled trial (SCRaTCH). Hum. Reprod. 2021, 36, 87–98. [Google Scholar] [CrossRef]
  140. Yu, X.; Gao, C.; Dai, C.; Yang, F.; Deng, X. Endometrial injury increases expression of hypoxia-inducible factor and angiogenesis in the endometrium of women with recurrent implantation failure. Reprod. Biomed. Online 2019, 38, 761–767. [Google Scholar] [CrossRef]
  141. Metwally, M.; Chatters, R.; Pye, C.; Dimairo, M.; White, D.; Walters, S.; Cohen, J.; Young, T.; Cheong, Y.; Laird, S.; et al. Endometrial scratch to increase live birth rates in women undergoing first-time in vitro fertilisation: RCT and systematic review. Health Technol. Assess. 2022, 26, 1–212. [Google Scholar] [CrossRef]
  142. Safdarian, L.; Movahedi, S.; Aleyasine, A.; Aghahosaini, M.; Fallah, P.; Rezaiian, Z. Local injury to the endometrium does not improve the implantation rate in good responder patients undergoing in-vitro fertilization. Iran. J. Reprod. Med. 2011, 9, 285–288. [Google Scholar]
  143. Melnick, A.P.; Murphy, E.M.; Masbou, A.K.; Sapra, K.J.; Rosenwaks, Z.; Spandorfer, S.D. Autologous endometrial coculture biopsy: Is timing everything? Fertil. Steril. 2015, 104, 104–109.e1. [Google Scholar] [CrossRef]
  144. Shokeir, T.; Ebrahim, M.; El-Mogy, H. Hysteroscopic-guided local endometrial injury does not improve natural cycle pregnancy rate in women with unexplained infertility: Randomized controlled trial. J. Obstet. Gynaecol. Res. 2016, 42, 1553–1557. [Google Scholar] [CrossRef]
  145. Shahrokh-Tehraninejad, E.; Dashti, M.; Hossein-Rashidi, B.; Azimi-Nekoo, E.; Haghollahi, F.; Kalantari, V. A Randomized Trial to Evaluate the Effect of Local Endometrial Injury on the Clinical Pregnancy Rate of Frozen Embryo Transfer Cycles in Patients with Repeated Implantation Failure. J. Fam. Reprod. Health 2016, 10, 108–114. [Google Scholar]
  146. Levin, D.; Hasson, J.; Cohen, A.; Or, Y.; Ata, B.; Barzilay, L.; Almog, B. The effect of endometrial injury on implantation and clinical pregnancy rates. Gynecol. Endocrinol. 2017, 33, 779–782. [Google Scholar] [CrossRef]
  147. Tk, A.; Singhal, H.; Premkumar, S.P.; Acharya, M.; Kamath, M.S.; George, K. Local endometrial injury in women with failed IVF undergoing a repeat cycle: A randomized controlled trial. Eur. J. Obstet. Gynecol. Reprod. Biol. 2017, 214, 109–114. [Google Scholar] [CrossRef]
  148. Liu, W.; Tal, R.; Chao, H.; Liu, M.; Liu, Y. Effect of local endometrial injury in proliferative vs. luteal phase on IVF outcomes in unselected subfertile women undergoing in vitro fertilization. Reprod. Biol. Endocrinol. 2017, 15, 75. [Google Scholar] [CrossRef]
  149. Mackens, S.; Racca, A.; Van de Velde, H.; Drakopoulos, P.; Tournaye, H.; Stoop, D.; Blockeel, C.; Santos-Ribeiro, S. Follicular-phase endometrial scratching: A truncated randomized controlled trial. Hum. Reprod. 2020, 35, 1090–1098. [Google Scholar] [CrossRef]
  150. Kalyoncu, S.; Yazicioglu, A.; Demir, M. Endometrial scratching for poor responders based on the Bologna criteria in ICSI fresh embryo transfer cycles: A preliminary retrospective cohort study. J. Turk. Ger. Gynecol. Assoc. 2021, 22, 47–52. [Google Scholar] [CrossRef]
  151. Rigos, I.; Athanasiou, V.; Vlahos, N.; Papantoniou, N.; Profer, D.; Siristatidis, C. The Addition of Endometrial Injury to Freeze-All Strategy in Women with Repeated Implantation Failures. J. Clin. Med. 2021, 10, 2162. [Google Scholar] [CrossRef] [PubMed]
  152. Noori, N.; Ghaemdoust, F.; Ghasemi, M.; Liavaly, M.; Keikha, N.; Dehghan Haghighi, J. The effect of endometrial scratching on reproductive outcomes in infertile women undergoing IVF treatment cycles. J. Obstet. Gynaecol. 2022, 42, 3611–3615. [Google Scholar] [CrossRef] [PubMed]
  153. Dain, L.; Ojha, K.; Bider, D.; Levron, J.; Zinchenko, V.; Walster, S.; Dirnfeld, M. Effect of local endometrial injury on pregnancy outcomes in ovum donation cycles. Fertil. Steril. 2014, 102, 1048–1054. [Google Scholar] [CrossRef] [PubMed]
  154. Mak, J.S.M.; Chung, C.H.S.; Chung, J.P.W.; Kong, G.W.S.; Saravelos, S.H.; Cheung, L.P.; Li, T.C. The effect of endometrial scratch on natural-cycle cryopreserved embryo transfer outcomes: A randomized controlled study. Reprod. Biomed. Online 2017, 35, 28–36. [Google Scholar] [CrossRef] [PubMed]
  155. Ashrafi, M.; Tehraninejad, E.S.; Haghiri, M.; Masomi, M.; Sadatmahalleh, S.J.; Arabipoor, A. The effect of endometrial scratch injury on pregnancy outcome in women with previous intrauterine insemination failure: A randomized clinical trial. J. Obstet. Gynaecol. Res. 2017, 43, 1421–1427. [Google Scholar] [CrossRef]
  156. Eskew, A.M.; Reschke, L.D.; Woolfolk, C.; Schulte, M.B.; Boots, C.E.; Broughton, D.E.; Jimenez, P.T.; Omurtag, K.R.; Keller, S.L.; Ratts, V.S.; et al. Effect of endometrial mechanical stimulation in an unselected population undergoing in vitro fertilization: Futility analysis of a double-blind randomized controlled trial. J. Assist. Reprod. Genet. 2019, 36, 299–305. [Google Scholar] [CrossRef]
  157. Frantz, S.; Parinaud, J.; Kret, M.; Rocher-Escriva, G.; Papaxanthos-Roche, A.; Creux, H.; Chansel-Debordeaux, L.; Benard, A.; Hocke, C. Decrease in pregnancy rate after endometrial scratch in women undergoing a first or second in vitro fertilization. A multicenter randomized controlled trial. Hum. Reprod. 2019, 34, 92–99. [Google Scholar] [CrossRef]
  158. Lensen, S.; Osavlyuk, D.; Armstrong, S.; Stadelmann, C.; Hennes, A.; Napier, E.; Wilkinson, J.; Sadler, L.; Gupta, D.; Strandell, A.; et al. A Randomized Trial of Endometrial Scratching before In Vitro Fertilization. N. Engl. J. Med. 2019, 380, 325–334. [Google Scholar] [CrossRef]
  159. Crosby, D.A.; Glover, L.E.; Downey, P.; Mooney, E.E.; McAuliffe, F.M.; O’Farrelly, C.; Brennan, D.J.; Wingfield, M. The impact of accurately timed mid-luteal endometrial injury in nulligravid women undergoing their first or second embryo transfer. Ir. J. Med. Sci. 2021, 190, 1071–1077. [Google Scholar] [CrossRef]
  160. Metwally, M.; Chatters, R.; Dimairo, M.; Walters, S.; Pye, C.; White, D.; Bhide, P.; Chater, T.; Cheong, Y.; Choudhary, M.; et al. A randomised controlled trial to assess the clinical effectiveness and safety of the endometrial scratch procedure prior to first-time IVF, with or without ICSI. Hum. Reprod. 2021, 36, 1841–1853. [Google Scholar] [CrossRef]
  161. Yavangi, M.; Varmaghani, N.; Pirdehghan, A.; Varmaghani, M.; Faryadras, M. Comparison of pregnancy outcome in intrauterine insemination-candidate women with and without endometrial scratch injury: An RCT. Int. J. Reprod. Biomed. 2021, 19, 457–464. [Google Scholar] [CrossRef]
  162. Farzaneh, F.; Khastehfekr, F. The effect of topical endometrial scratching on pregnancy outcome in women with previous failure of intrauterine insemination: A non-randomized clinical trial. Int. J. Reprod. Biomed. 2021, 19, 465–470. [Google Scholar] [CrossRef]
  163. Glanville, E.J.; Wilkinson, J.; Sadler, L.; Wong, T.Y.; Acharya, S.; Aziz, N.; Clarke, F.; Das, S.; Dawson, J.; Hammond, B.; et al. A randomized trial of endometrial scratching in women with PCOS undergoing ovulation induction cycles. Reprod. Biomed. Online 2022, 44, 316–323. [Google Scholar] [CrossRef]
  164. Wong, T.Y.; Lensen, S.; Wilkinson, J.; Glanville, E.J.; Acharya, S.; Clarke, F.; Das, S.; Dawson, J.; Hammond, B.; Jayaprakasan, K.; et al. Effect of endometrial scratching on unassisted conception for unexplained infertility: A randomized controlled trial. Fertil. Steril. 2022, 117, 612–619. [Google Scholar] [CrossRef]
  165. Bernard, A.; Schumacher, K.; Marsh, C. Endometrial Scratch (Injury): Does Timing Matter? J. Fam. Reprod. Health 2019, 13, 85–88. [Google Scholar] [CrossRef]
  166. Iriarte, C.; Awosika, O.; Rengifo-Pardo, M.; Ehrlich, A. Review of applications of microneedling in dermatology. Clin. Cosmet. Investig. Dermatol. 2017, 10, 289–298. [Google Scholar] [CrossRef]
  167. Ersahin, S.S.; Ersahin, A. Endometrial injury concurrent with hysteroscopy increases the expression of Leukaemia inhibitory factor: A preliminary study. Reprod. Biol. Endocrinol. 2022, 20, 11. [Google Scholar] [CrossRef]
  168. Mrozikiewicz, A.E.; Ozarowski, M.; Jedrzejczak, P. Biomolecular Markers of Recurrent Implantation Failure—A Review. Int. J. Mol. Sci. 2021, 22, 10082. [Google Scholar] [CrossRef]
  169. Yang, Y.; Chen, X.; Saravelos, S.H.; Liu, Y.; Huang, J.; Zhang, J.; Li, T.C. HOXA-10 and E-cadherin expression in the endometrium of women with recurrent implantation failure and recurrent miscarriage. Fertil. Steril. 2017, 107, 136–143.e2. [Google Scholar] [CrossRef]
  170. 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]
  171. Santamaria, X.; Katzorke, N.; Simon, C. Endometrial ‘scratching’: What the data show. Curr. Opin. Obstet. Gynecol. 2016, 28, 242–249. [Google Scholar] [CrossRef]
  172. Maged, A.M.; Ogila, A.I.; Mohsen, R.A.; Mahmoud, S.I.; Fouad, M.A.; El Komy, R.O.; Lasheen, Y.; El-Nassery, N.; Dahab, S.; Hussein, E.A. Endometrial scratch injury in infertile women seeking conception through natural or intrauterine insemination cycles: A systematic review and meta-analysis. Int. J. Gynaecol. Obstet. 2021. online ahead of print. [Google Scholar] [CrossRef]
  173. Kang, Y.; Wang, Z.; Yang, Y.; Liang, H.; Duan, X.; Gao, Q.; Yin, Z. Impact of endometrial scratching on reproductive outcome in patients: A systematic review and meta-analysis. Medicine 2022, 101, e30150. [Google Scholar] [CrossRef] [PubMed]
  174. Baradwan, S.; Alshahrani, M.S.; AlSghan, R.; Alkhamis, W.H.; Alsharif, S.A.; Alanazi, G.A.; Abdelwahed, R.M.; Alkholy, E.A.; Fouad, M.; Saleh, M.; et al. The Effect of Endometrial Scratch on Pregnancy Rate in Women with Previous Intrauterine Insemination Failure: A Systematic Review and Meta-analysis of Randomized Controlled Trials. Reprod. Sci. 2022. online ahead of print. [Google Scholar] [CrossRef] [PubMed]
  175. Palomba, S.; Vitagliano, A.; Marci, R.; Caserta, D. Endometrial Scratching for Improving Endometrial Receptivity: A Critical Review of Old and New Clinical Evidence. Reprod. Sci. 2022. [Google Scholar] [CrossRef] [PubMed]
  176. Gui, J.; Xu, W.; Yang, J.; Feng, L.; Jia, J. Impact of local endometrial injury on in vitro fertilization/intracytoplasmic sperm injection outcomes: A systematic review and meta-analysis. J. Obstet. Gynaecol. Res. 2019, 45, 57–68. [Google Scholar] [CrossRef]
  177. El-Toukhy, T.; Sunkara, S.; Khalaf, Y. Local endometrial injury and IVF outcome: A systematic review and meta-analysis. Reprod. Biomed. Online 2012, 25, 345–354. [Google Scholar] [CrossRef]
  178. Lensen, S.F.; Armstrong, S.; Gibreel, A.; Nastri, C.O.; Raine-Fenning, N.; Martins, W.P. Endometrial injury in women undergoing in vitro fertilisation (IVF). Cochrane Database Syst. Rev. 2021, 6, CD009517. [Google Scholar] [CrossRef]
  179. Aghajanzadeh, F.; Esmaeilzadeh, S.; Basirat, Z.; Mahouti, T.; Heidari, F.N.; Golsorkhtabaramiri, M. Using autologous intrauterine platelet-rich plasma to improve the reproductive outcomes of women with recurrent implantation failure. JBRA Assist. Reprod. 2020, 24, 30–33. [Google Scholar] [CrossRef]
  180. Hajipour, H.; Farzadi, L.; Latifi, Z.; Keyhanvar, N.; Navali, N.; Fattahi, A.; Nouri, M.; Dittrich, R. An update on platelet-rich plasma (PRP) therapy in endometrium and ovary related infertilities: Clinical and molecular aspects. Syst. Biol. Reprod. Med. 2021, 67, 177–188. [Google Scholar] [CrossRef]
  181. Sharara, F.I.; Lelea, L.L.; Rahman, S.; Klebanoff, J.S.; Moawad, G.N. A narrative review of platelet-rich plasma (PRP) in reproductive medicine. J. Assist. Reprod. Genet. 2021, 38, 1003–1012. [Google Scholar] [CrossRef]
  182. Amable, P.R.; Carias, R.B.; Teixeira, M.V.; da Cruz Pacheco, I.; Correa do Amaral, R.J.; Granjeiro, J.M.; Borojevic, R. Platelet-rich plasma preparation for regenerative medicine: Optimization and quantification of cytokines and growth factors. Stem Cell Res. Ther. 2013, 4, 67. [Google Scholar] [CrossRef]
  183. Maged, A.M.; El-Mazny, A.; Kamal, N.; Mahmoud, S.I.; Fouad, M.; El-Nassery, N.; Kotb, A.; Ragab, W.S.; Ogila, A.I.; Metwally, A.A.; et al. The value of platelet-rich plasma in women with previous implantation failure: A systematic review and meta-analysis. J. Assist. Reprod. Genet. 2023. [Google Scholar] [CrossRef]
  184. Maleki-Hajiagha, A.; Razavi, M.; Rouholamin, S.; Rezaeinejad, M.; Maroufizadeh, S.; Sepidarkish, M. Intrauterine infusion of autologous platelet-rich plasma in women undergoing assisted reproduction: A systematic review and meta-analysis. J. Reprod. Immunol. 2020, 137, 103078. [Google Scholar] [CrossRef]
  185. Anitua, E.; Allende, M.; de la Fuente, M.; Del Fabbro, M.; Alkhraisat, M.H. Efficacy of Platelet-Rich Plasma in Women with a History of Embryo Transfer Failure: A Systematic Review and Meta-Analysis with Trial Sequential Analysis. Bioengineering 2023, 10, 303. [Google Scholar] [CrossRef]
  186. Li, M.; Kang, Y.; Wang, Q.; Yan, L. Efficacy of Autologous Intrauterine Infusion of Platelet-Rich Plasma in Patients with Unexplained Repeated Implantation Failures in Embryo Transfer: A Systematic Review and Meta-Analysis. J. Clin. Med. 2022, 11, 6753. [Google Scholar] [CrossRef]
  187. Deng, H.; Wang, S.; Li, Z.; Xiao, L.; Ma, L. Effect of intrauterine infusion of platelet-rich plasma for women with recurrent implantation failure: A systematic review and meta-analysis. J. Obstet. Gynaecol. 2023, 43, 2144177. [Google Scholar] [CrossRef]
  188. 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]
  189. Kong, X.; Tang, G.; Liu, Y.; Zheng, Z.; Li, Y.; Yan, F. Efficacy of intrauterine infusion therapy before embryo transfer in recurrent implantation failure: A systematic review and network meta-analysis. J. Reprod. Immunol. 2023, 156, 103819. [Google Scholar] [CrossRef]
  190. Aghajanova, L.; Houshdaran, S.; Balayan, S.; Manvelyan, E.; Irwin, J.C.; Huddleston, H.G.; Giudice, L.C. In vitro evidence that platelet-rich plasma stimulates cellular processes involved in endometrial regeneration. J. Assist. Reprod. Genet. 2018, 35, 757–770. [Google Scholar] [CrossRef] [PubMed]
  191. Yuan, B.; Luo, S.; Mao, J.; Luo, B.; Wang, J. Effects of intrauterine infusion of platelet-rich plasma on hormone levels and endometrial receptivity in patients with repeated embryo implantation failure. Am. J. Transl. Res. 2022, 14, 5651–5659. [Google Scholar]
  192. Kieu, V.; Lantsberg, D.; Mizrachi, Y.; Stern, C.; Polyakov, A.; Teh, W.T. A survey study of endometrial receptivity tests and immunological treatments in in vitro fertilisation (IVF). Aust. N. Z. J. Obstet. Gynaecol. 2022, 62, 306–311. [Google Scholar] [CrossRef] [PubMed]
  193. Woon, E.V.; Day, A.; Bracewell-Milnes, T.; Male, V.; Johnson, M. Immunotherapy to improve pregnancy outcome in women with abnormal natural killer cell levels/activity and recurrent miscarriage or implantation failure: A systematic review and meta-analysis. J. Reprod. Immunol. 2020, 142, 103189. [Google Scholar] [CrossRef] [PubMed]
  194. Toth, B.; Vomstein, K.; Togawa, R.; Bottcher, B.; Hudalla, H.; Strowitzki, T.; Daniel, V.; Kuon, R.J. The impact of previous live births on peripheral and uterine natural killer cells in patients with recurrent miscarriage. Reprod. Biol. Endocrinol. 2019, 17, 72. [Google Scholar] [CrossRef] [PubMed]
  195. Lapides, L.; Varga, I.; Klein, M.; Rybanska, L.; Belusakova, V.; Babal, P. When Less Is More—Pipelle Endometrial Sampling for Quantification of Uterine Natural Killer Cells in Patients with Recurrent Implantation Failure or Habitual Abortion. Physiol. Res. 2022, 71, S65–S73. [Google Scholar] [CrossRef]
  196. Hartman, S.K.; Symons, W.A.; Yeh, I.-T. Chronic endometritis: How many plasma cells does it take to make the diagnosis? FASEB J. 2011, 25, 1002.13. [Google Scholar] [CrossRef]
  197. Park, H.J.; Kim, Y.S.; Yoon, T.K.; Lee, W.S. Chronic endometritis and infertility. Clin. Exp. Reprod. Med. 2016, 43, 185–192. [Google Scholar] [CrossRef]
  198. Einenkel, R.; Zygmunt, M.; Muzzio, D.O. Microorganisms in the healthy upper reproductive tract: From denial to beneficial assignments for reproductive biology. Reprod. Biol. 2019, 19, 113–118. [Google Scholar] [CrossRef]
  199. Vomstein, K.; Feil, K.; Strobel, L.; Aulitzky, A.; Hofer-Tollinger, S.; Kuon, R.-J.; Toth, B. Immunological Risk Factors in Recurrent Pregnancy Loss: Guidelines Versus Current State of the Art. J. Clin. Med. 2021, 10, 869. [Google Scholar] [CrossRef]
  200. Salih, S.M.; Havemann, L.; Lindheim, S.R. Human Leukocyte Antigen (HLA) Typing in Medically Assisted Reproduction. In Textbook of Assisted Reproduction; Allahbadia, G.N., Ata, B., Lindheim, S.R., Woodward, B.J., Bhagavath, B., Eds.; Springer: Singapore, 2020; pp. 299–306. [Google Scholar]
  201. Wu, L.; Fang, X.; Lu, F.; Zhang, Y.; Wang, Y.; Kwak-Kim, J. Anticardiolipin and/or anti-beta2-glycoprotein-I antibodies are associated with adverse IVF outcomes. Front. Immunol. 2022, 13, 986893. [Google Scholar] [CrossRef]
  202. Kwak-Kim, J.Y.; Chung-Bang, H.S.; Ng, S.C.; Ntrivalas, E.I.; Mangubat, C.P.; Beaman, K.D.; Beer, A.E.; Gilman-Sachs, A. Increased T helper 1 cytokine responses by circulating T cells are present in women with recurrent pregnancy losses and in infertile women with multiple implantation failures after IVF. Hum. Reprod. 2003, 18, 767–773. [Google Scholar] [CrossRef]
  203. Winger, E.E.; Reed, J.L.; Ashoush, S.; El-Toukhy, T.; Ahuja, S.; Taranissi, M. Degree of TNF-alpha/IL-10 cytokine elevation correlates with IVF success rates in women undergoing treatment with Adalimumab (Humira) and IVIG. Am. J. Reprod. Immunol. 2011, 65, 610–618. [Google Scholar] [CrossRef] [PubMed]
  204. Winger, E.E.; Reed, J.L.; Ashoush, S.; El-Toukhy, T.; Ahuja, S.; Taranissi, M. Elevated preconception CD56+16+ and/or Th1:Th2 levels predict benefit from IVIG therapy in subfertile women undergoing IVF. Am. J. Reprod. Immunol. 2011, 66, 394–403. [Google Scholar] [CrossRef] [PubMed]
  205. Nakagawa, K.; Kwak-Kim, J.; Kuroda, K.; Sugiyama, R.; Yamaguchi, K. Immunosuppressive treatment using tacrolimus promotes pregnancy outcome in infertile women with repeated implantation failures. Am. J. Reprod. Immunol. 2017, 78, e12682. [Google Scholar] [CrossRef] [PubMed]
  206. Nardo, L.; Chouliaras, S. Adjuvants in IVF-evidence for what works and what does not work. Ups. J. Med. Sci. 2020, 125, 144–151. [Google Scholar] [CrossRef]
  207. Whirledge, S.; Cidlowski, J.A. Glucocorticoids and Reproduction: Traffic Control on the Road to Reproduction. Trends Endocrinol. Metab. 2017, 28, 399–415. [Google Scholar] [CrossRef]
  208. Henderson, T.A.; Saunders, P.T.K.; Moffett-King, A.; Groome, N.P.; Critchley, H.O.D. Steroid receptor expression in uterine natural killer cells. J. Clin. Endocr. Metab. 2003, 88, 440–449. [Google Scholar] [CrossRef]
  209. Thiruchelvam, U.; Maybin, J.A.; Armstrong, G.M.; Greaves, E.; Saunders, P.T.K.; Critchley, H.O.D. Cortisol regulates the paracrine action of macrophages by inducing vasoactive gene expression in endometrial cells. J. Leukoc. Biol. 2016, 99, 1165–1171. [Google Scholar] [CrossRef]
  210. Horton, J.S.; Yamamoto, S.Y.; Bryant-Greenwood, G.D. Relaxin Modulates Proinflammatory Cytokine Secretion from Human Decidual Macrophages. Biol. Reprod. 2011, 85, 788–797. [Google Scholar] [CrossRef]
  211. Quenby, S.; Kalumbi, C.; Bates, M.; Farquharson, R.; Vince, G. Prednisolone reduces preconceptual endometrial natural killer cells in women with recurrent miscarriage. Fertil. Steril. 2005, 84, 980–984. [Google Scholar] [CrossRef]
  212. Chen, Y.Z.; Wang, Y.; Zhuang, Y.L.; Zhou, F.L.; Huang, L.L. Mifepristone Increases the Cytotoxicity of Uterine Natural Killer Cells by Acting as a Glucocorticoid Antagonist via ERK Activation. PLoS ONE 2012, 7, e36413. [Google Scholar] [CrossRef]
  213. Thum, M.Y.; Bhaskaran, S.; Abdalla, H.I.; Ford, B.; Sumar, N.; Bansal, A. Prednisolone suppresses NK cell cytotoxicity in vitro in women with a history of infertility and elevated NK cell cytotoxicity. Am. J. Reprod. Immunol. 2008, 59, 259–265. [Google Scholar] [CrossRef]
  214. Mahdian, S.; Zarrabi, M.; Moini, A.; Shahhoseini, M.; Movahedi, M. In silico evidence for prednisone and progesterone efficacy in recurrent implantation failure treatment. J. Mol. Model. 2022, 28, 105. [Google Scholar] [CrossRef]
  215. Kuroda, K.; Venkatakrishnan, R.; Salker, M.S.; Lucas, E.S.; Shaheen, F.; Kuroda, M.; Blanks, A.; Christian, M.; Quenby, S.; Brosens, J.J. Induction of 11beta-HSD 1 and activation of distinct mineralocorticoid receptor- and glucocorticoid receptor-dependent gene networks in decidualizing human endometrial stromal cells. Mol. Endocrinol. 2013, 27, 192–202. [Google Scholar] [CrossRef]
  216. Boomsma, C.M.; Kamath, M.S.; Keay, S.D.; Macklon, N.S. Peri-implantation glucocorticoid administration for assisted reproductive technology cycles. Cochrane Database Syst. Rev. 2022, 6, CD005996. [Google Scholar] [CrossRef]
  217. Cooper, S.; Laird, S.M.; Mariee, N.; Li, T.C.; Metwally, M. The effect of prednisolone on endometrial uterine NK cell concentrations and pregnancy outcome in women with reproductive failure. A retrospective cohort study. J. Reprod. Immunol. 2019, 131, 1–6. [Google Scholar] [CrossRef]
  218. Robertson, S.A.; Jin, M.; Yu, D.; Moldenhauer, L.M.; Davies, M.J.; Hull, M.L.; Norman, R.J. Corticosteroid therapy in assisted reproduction—Immune suppression is a faulty premise. Hum. Reprod. 2016, 31, 2164–2173. [Google Scholar] [CrossRef]
  219. Michael, A.E.; Papageorghiou, A.T. Potential significance of physiological and pharmacological glucocorticoids in early pregnancy. Hum. Reprod. Update 2008, 14, 497–517. [Google Scholar] [CrossRef]
  220. Mayer, K.; Meyer, S.; Reinholz-Muhly, M.; Maus, U.; Merfels, M.; Lohmeyer, J.; Grimminger, F.; Seeger, W. Short-time infusion of fish oil-based lipid emulsions, approved for parenteral nutrition, reduces monocyte proinflammatory cytokine generation and adhesive interaction with endothelium in humans. J. Immunol. 2003, 171, 4837–4843. [Google Scholar] [CrossRef]
  221. Roussev, R.G.; Acacio, B.; Ng, S.C.; Coulam, C.B. Duration of intralipid’s suppressive effect on NK cell’s functional activity. Am. J. Reprod. Immunol. 2008, 60, 258–263. [Google Scholar] [CrossRef]
  222. Ledee, N.; Vasseur, C.; Petitbarat, M.; Chevrier, L.; Vezmar, K.; Dray, G.; Cheniere, S.; Lobersztajn, A.; Vitoux, D.; Cassuto, G.N.; et al. Intralipid (R) may represent a new hope for patients with reproductive failures and simultaneously an over-immune endometrial activation. J. Reprod. Immunol. 2018, 130, 18–22. [Google Scholar] [CrossRef]
  223. Kumar, P.; Marron, K.; Harrity, C. Intralipid therapy and adverse reproductive outcome: Is there any evidence? Reprod. Fertil. 2021, 2, 173–186. [Google Scholar] [CrossRef] [PubMed]
  224. Han, E.J.; Lee, H.N.; Kim, M.K.; Lyu, S.W.; Lee, W.S. Efficacy of intralipid administration to improve in vitro fertilization outcomes: A systematic review and meta-analysis. Clin. Exp. Reprod. Med. 2021, 48, 203–210. [Google Scholar] [CrossRef] [PubMed]
  225. Rimmer, M.P.; Black, N.; Keay, S.; Quenby, S.; Al Wattar, B.H. Intralipid infusion at time of embryo transfer in women with history of recurrent implantation failure: A systematic review and meta-analysis. J. Obstet. Gynaecol. Res. 2021, 47, 2149–2156. [Google Scholar] [CrossRef] [PubMed]
  226. Placais, L.; Kolanska, K.; Kraiem, Y.B.; Cohen, J.; Suner, L.; Bornes, M.; Sedille, L.; Rosefort, A.; D’Argent, E.M.; Selleret, L.; et al. Intralipid therapy for unexplained recurrent miscarriage and implantation failure: Case-series and literature review. Eur. J. Obstet. Gynecol. Reprod. Biol. 2020, 252, 100–104. [Google Scholar] [CrossRef] [PubMed]
  227. Zhou, P.; Wu, H.; Lin, X.; Wang, S.; Zhang, S. The effect of intralipid on pregnancy outcomes in women with previous implantation failure in in vitro fertilization/intracytoplasmic sperm injection cycles: A systematic review and meta-analysis. Eur. J. Obstet. Gynecol. Reprod. Biol. 2020, 252, 187–192. [Google Scholar] [CrossRef]
  228. Coulam, C.B. Intralipid treatment for women with reproductive failures. Am. J. Reprod. Immunol. 2021, 85, e13290. [Google Scholar] [CrossRef]
  229. Shreeve, N.; Sadek, K. Intralipid therapy for recurrent implantation failure: New hope or false dawn? J. Reprod. Immunol. 2012, 93, 38–40. [Google Scholar] [CrossRef]
  230. Foyle, K.L.; Sharkey, D.J.; Moldenhauer, L.M.; Green, E.S.; Wilson, J.J.; Roccisano, C.J.; Hull, M.L.; Tremellen, K.P.; Robertson, S.A. Effect of Intralipid infusion on peripheral blood T cells and plasma cytokines in women undergoing assisted reproduction treatment. Clin. Transl. Immunol. 2021, 10, e1328. [Google Scholar] [CrossRef]
  231. Martini, A.E.; Jasulaitis, S.; Fogg, L.F.; Uhler, M.L.; Hirshfeld-Cytron, J.E. Evaluating the Utility of Intralipid Infusion to Improve Live Birth Rates in Patients with Recurrent Pregnancy Loss or Recurrent Implantation Failure. J. Hum. Reprod. Sci. 2018, 11, 261–268. [Google Scholar] [CrossRef]
  232. Canella, P.; Barini, R.; Carvalho, P.O.; Razolli, D.S. Lipid emulsion therapy in women with recurrent pregnancy loss and repeated implantation failure: The role of abnormal natural killer cell activity. J. Cell. Mol. Med. 2021, 25, 2290–2296. [Google Scholar] [CrossRef]
  233. Jang, D.I.; Lee, A.H.; Shin, H.Y.; Song, H.R.; Park, J.H.; Kang, T.B.; Lee, S.R.; Yang, S.H. The Role of Tumor Necrosis Factor Alpha (TNF-alpha) in Autoimmune Disease and Current TNF-alpha Inhibitors in Therapeutics. Int. J. Mol. Sci. 2021, 22, 2719. [Google Scholar] [CrossRef]
  234. Clark, D.A. Anti-TNFalpha therapy in immune-mediated subfertility: State of the art. J. Reprod. Immunol. 2010, 85, 15–24. [Google Scholar] [CrossRef]
  235. Santiago, K.Y.; Porchia, L.M.; Lopez-Bayghen, E. Endometrial preparation with etanercept increased embryo implantation and live birth rates in women suffering from recurrent implantation failure during IVF. Reprod. Biol. 2021, 21, 100480. [Google Scholar] [CrossRef]
  236. Winger, E.E.; Reed, J.L.; Ashoush, S.; El-Toukhy, T.; Taranissi, M. Die-off ratio correlates with increased TNF-alpha:IL-10 ratio and decreased IVF success rates correctable with humira. Am. J. Reprod. Immunol. 2012, 68, 428–437. [Google Scholar] [CrossRef]
  237. Jerzak, M.; Ohams, M.; Gorski, A.; Baranowski, W. Etanercept immunotherapy in women with a history of recurrent reproductive failure. Ginekol. Pol. 2012, 83, 260–264. [Google Scholar]
  238. Gilardin, L.; Bayry, J.; Kaveri, S.V. Intravenous immunoglobulin as clinical immune-modulating therapy. Can. Med. Assoc. J. 2015, 187, 257–264. [Google Scholar] [CrossRef]
  239. Kaufman, G.N.; Massoud, A.H.; Dembele, M.; Yona, M.; Piccirillo, C.A.; Mazer, B.D. Induction of regulatory T cells by intravenous immunoglobulin: A bridge between adaptive and innate immunity. Front. Immunol. 2015, 6, 469. [Google Scholar] [CrossRef]
  240. Ahmadi, M.; Abdolmohammadi-Vahid, S.; Ghaebi, M.; Aghebati-Maleki, L.; Dolati, S.; Farzadi, L.; Ghasemzadeh, A.; Hamdi, K.; Younesi, V.; Nouri, M.; et al. Regulatory T cells improve pregnancy rate in RIF patients after additional IVIG treatment. Syst. Biol. Reprod. Med. 2017, 63, 350–359. [Google Scholar] [CrossRef]
  241. Coulam, C.B.; Krysa, L.W.; Bustillo, M. Intravenous immunoglobulin for in-vitro fertilization failure. Hum. Reprod. 1994, 9, 2265–2269. [Google Scholar] [CrossRef]
  242. Ho, Y.K.; Chen, H.H.; Huang, C.C.; Lee, C.I.; Lin, P.Y.; Lee, M.S.; Lee, T.H. Peripheral CD56+CD16+ NK Cell Populations in the Early Follicular Phase Are Associated with Successful Clinical Outcomes of Intravenous Immunoglobulin Treatment in Women with Repeated Implantation Failure. Front. Endocrinol. 2019, 10, 937. [Google Scholar] [CrossRef]
  243. Ramos-Medina, R.; Garcia-Segovia, A.; Gil, J.; Carbone, J.; Aguaron de la Cruz, A.; Seyfferth, A.; Alonso, B.; Alonso, J.; Leon, J.A.; Alecsandru, D.; et al. Experience in IVIg therapy for selected women with recurrent reproductive failure and NK cell expansion. Am. J. Reprod. Immunol. 2014, 71, 458–466. [Google Scholar] [CrossRef] [PubMed]
  244. Heilmann, L.; Schorsch, M.; Hahn, T. CD3 CD56+ CD16+ natural killer cells and improvement of pregnancy outcome in IVF/ICSI failure after additional IVIG-treatment. Am. J. Reprod. Immunol. 2010, 63, 263–265. [Google Scholar] [CrossRef] [PubMed]
  245. Coulam, C.B.; Goodman, C. Increased pregnancy rates after IVF/ET with intravenous immunoglobulin treatment in women with elevated circulating C56+ cells. Early Pregnancy 2000, 4, 90–98. [Google Scholar] [PubMed]
  246. Han, A.R.; Lee, S.K. Immune modulation of i.v. immunoglobulin in women with reproductive failure. Reprod. Med. Biol. 2018, 17, 115–124. [Google Scholar] [CrossRef]
  247. Saab, W.; Seshadri, S.; Huang, C.; Alsubki, L.; Sung, N.; Kwak-Kim, J. A systemic review of intravenous immunoglobulin G treatment in women with recurrent implantation failures and recurrent pregnancy losses. Am. J. Reprod. Immunol. 2021, 85, e13395. [Google Scholar] [CrossRef]
  248. Abdolmohammadi-Vahid, S.; Pashazadeh, F.; Pourmoghaddam, Z.; Aghebati-Maleki, L.; Abdollahi-Fard, S.; Yousefi, M. The effectiveness of IVIG therapy in pregnancy and live birth rate of women with recurrent implantation failure (RIF): A systematic review and meta-analysis. J. Reprod. Immunol. 2019, 134–135, 28–33. [Google Scholar] [CrossRef]
  249. Sapir, T.; Carp, H.; Shoenfeld, Y. Intravenous immunoglobulin (IVIG) as treatment for recurrent pregnancy loss (RPL). Harefuah 2005, 144, 415–420, 453, 454. [Google Scholar]
  250. Elram, T.; Simon, A.; Israel, S.; Revel, A.; Shveiky, D.; Laufer, N. Treatment of recurrent IVF failure and human leukocyte antigen similarity by intravenous immunoglobulin. Reprod. Biomed. Online 2005, 11, 745–749. [Google Scholar] [CrossRef]
  251. Bahrami-Asl, Z.; Farzadi, L.; Fattahi, A.; Yousefi, M.; Quinonero, A.; Hakimi, P.; Latifi, Z.; Nejabati, H.R.; Ghasemnejad, T.; Sadigh, A.R.; et al. Tacrolimus Improves the Implantation Rate in Patients with Elevated Th1/2 Helper Cell Ratio and Repeated Implantation Failure (RIF). Geburtshilfe Frauenheilkd 2020, 80, 851–862. [Google Scholar] [CrossRef]
  252. Nakagawa, K.; Kwak-Kim, J.; Ota, K.; Kuroda, K.; Hisano, M.; Sugiyama, R.; Yamaguchi, K. Immunosuppression with tacrolimus improved reproductive outcome of women with repeated implantation failure and elevated peripheral blood TH1/TH2 cell ratios. Am. J. Reprod. Immunol. 2015, 73, 353–361. [Google Scholar] [CrossRef]
  253. Fluhr, H.; Spratte, J.; Heidrich, S.; Ehrhardt, J.; Greinacher, A.; Zygmunt, M. The molecular charge and size of heparins determine their impact on the decidualization of human endometrial stromal cells. Mol. Hum. Reprod. 2011, 17, 354–359. [Google Scholar] [CrossRef]
  254. Kashiwakura, Y.; Kojima, H.; Kanno, Y.; Hashiguchi, M.; Kobata, T. Heparin affects the induction of regulatory T cells independent of anti-coagulant activity and suppresses allogeneic immune responses. Clin. Exp. Immunol. 2020, 202, 119–135. [Google Scholar] [CrossRef]
  255. Niu, Z.; Zhou, M.; Xia, L.; Zhao, S.; Zhang, A. Uterine cytokine profiles after low-molecular-weight heparin administration are associated with pregnancy outcomes of patients with repeated implantation failure. Front. Endocrinol. 2022, 13, 1008923. [Google Scholar] [CrossRef]
  256. Spratte, J.; Meyer zu Schwabedissen, H.; Endlich, N.; Zygmunt, M.; Fluhr, H. Heparin inhibits TNF-α signaling in human endometrial stromal cells by interaction with NF-κB. Mol. Hum. Reprod. 2013, 19, 227–236. [Google Scholar] [CrossRef]
  257. Grandone, E.; Villani, M.; Dentali, F.; Tiscia, G.L.; Colaizzo, D.; Cappucci, F.; Fischetti, L.; Ageno, W.; Margaglione, M. Low-molecular -weight heparin in pregnancies after ART—A retrospective study. Thromb. Res. 2014, 134, 336–339. [Google Scholar] [CrossRef]
  258. Potdar, N.; Gelbaya, T.A.; Konje, J.C.; Nardo, L.G. Adjunct low-molecular-weight heparin to improve live birth rate after recurrent implantation failure: A systematic review and meta-analysis. Hum. Reprod. Update 2013, 19, 674–684. [Google Scholar] [CrossRef]
  259. Elmahashi, M.O.; Elbareg, A.M.; Essadi, F.M.; Ashur, B.M.; Adam, I. Low dose aspirin and low-molecular-weight heparin in the treatment of pregnant Libyan women with recurrent miscarriage. BMC Res. Notes 2014, 7, 23. [Google Scholar] [CrossRef]
  260. Dias, A.T.B.; Modesto, T.B.; Oliveira, S.A. Effectiveness of the use of Low Molecular Heparin in patients with repetition abortion history: Systematic review and meta-analysis. JBRA Assist. Reprod. 2021, 25, 10–27. [Google Scholar] [CrossRef]
  261. Siristatidis, C.; Dafopoulos, K.; Salamalekis, G.; Galazios, G.; Christoforidis, N.; Moustakarias, T.; Koutlaki, N.; Bouschanetzis, C.; Loutradis, D.; Drakakis, P. Administration of low-molecular-weight heparin in patients with two or more unsuccessful IVF/ICSI cycles: A multicenter cohort study. Gynecol. Endocrinol. 2018, 34, 747–751. [Google Scholar] [CrossRef]
  262. Kamel, A.M.; El-Faissal, Y.; Aboulghar, M.; Mansour, R.; Serour, G.I.; Aboulghar, M. Does intrauterine injection of low-molecular-weight heparin improve the clinical pregnancy rate in intracytoplasmic sperm injection? Clin. Exp. Reprod. Med. 2016, 43, 247–252. [Google Scholar] [CrossRef]
  263. Hamdi, K.; Danaii, S.; Farzadi, L.; Abdollahi, S.; Chalabizadeh, A.; Abdollahi Sabet, S. The Role of Heparin in Embryo Implantation in Women with Recurrent Implantation Failure in the Cycles of Assisted Reproductive Techniques (without History of Thrombophilia). J. Fam. Reprod. Health 2015, 9, 59–64. [Google Scholar]
  264. Akhtar, M.A.; Sur, S.; Raine-Fenning, N.; Jayaprakasan, K.; Thornton, J.; Quenby, S.; Marjoribanks, J. Heparin for assisted reproduction: Summary of a Cochrane review. Fertil. Steril. 2015, 103, 33–34. [Google Scholar] [CrossRef] [PubMed]
  265. Huang, P.; Yao, C.; Wei, L.; Lin, Z. The intrauterine perfusion of granulocyte-colony stimulating factor (G-CSF) before frozen-thawed embryo transfer in patients with two or more implantation failures. Hum. Fertil. 2020, 25, 301–305. [Google Scholar] [CrossRef] [PubMed]
  266. Jiang, Y.; Zhao, Q.; Zhang, Y.; Zhou, L.; Lin, J.; Chen, Y.; Qian, X. Treatment of G-CSF in unexplained, repeated implantation failure: A systematic review and meta-analysis. J. Gynecol. Obstet. Hum. Reprod. 2020, 49, 101866. [Google Scholar] [CrossRef] [PubMed]
  267. Rocha, M.N.C.; Florencio, R.S.; Alves, R.R.F. The role played by granulocyte colony stimulating factor (G-CSF) on women submitted to in vitro fertilization associated with thin endometrium: Systematic review. JBRA Assist. Reprod. 2020, 24, 278–282. [Google Scholar] [CrossRef]
  268. Schlahsa, L.; Jaimes, Y.; Blasczyk, R.; Figueiredo, C. Granulocyte-colony-stimulatory factor: A strong inhibitor of natural killer cell function. Transfusion 2011, 51, 293–305. [Google Scholar] [CrossRef]
  269. Fu, L.L.; Xu, Y.; Yan, J.; Zhang, X.Y.; Li, D.D.; Zheng, L.W. Efficacy of granulocyte colony-stimulating factor for infertility undergoing IVF: A systematic review and meta-analysis. Reprod. Biol. Endocrinol. 2023, 21, 34. [Google Scholar] [CrossRef]
  270. Hou, Z.; Jiang, F.; Yang, J.; Liu, Y.; Zha, H.; Yang, X.; Bie, J.; Meng, Y. What is the impact of granulocyte colony-stimulating factor (G-CSF) in subcutaneous injection or intrauterine infusion and during both the fresh and frozen embryo transfer cycles on recurrent implantation failure: A systematic review and meta-analysis? Reprod. Biol. Endocrinol. 2021, 19, 125. [Google Scholar] [CrossRef]
  271. Zhu, Y.C.; Sun, Y.X.; Shen, X.Y.; Jiang, Y.; Liu, J.Y. Effect of intrauterine perfusion of granular leukocyte-colony stimulating factor on the outcome of frozen embryo transfer. World J. Clin. Cases 2021, 9, 9038–9049. [Google Scholar] [CrossRef]
  272. Kalem, Z.; Namli Kalem, M.; Bakirarar, B.; Kent, E.; Makrigiannakis, A.; Gurgan, T. Intrauterine G-CSF Administration in Recurrent Implantation Failure (RIF): An Rct. Sci. Rep. 2020, 10, 5139. [Google Scholar] [CrossRef]
  273. Kamath, M.S.; Kirubakaran, R.; Sunkara, S.K. Granulocyte-colony stimulating factor administration for subfertile women undergoing assisted reproduction. Cochrane Database Syst. Rev. 2020, 1, CD013226. [Google Scholar] [CrossRef]
  274. Craciunas, L.; Tsampras, N.; Raine-Fenning, N.; Coomarasamy, A. Intrauterine administration of human chorionic gonadotropin (hCG) for subfertile women undergoing assisted reproduction. Cochrane Database Syst. Rev. 2018, 10, CD011537. [Google Scholar] [CrossRef]
  275. Liu, X.; Ma, D.; Wang, W.; Qu, Q.; Zhang, N.; Wang, X.; Fang, J.; Ma, Z.; Hao, C. Intrauterine administration of human chorionic gonadotropin improves the live birth rates of patients with repeated implantation failure in frozen-thawed blastocyst transfer cycles by increasing the percentage of peripheral regulatory T cells. Arch. Gynecol. Obstet. 2019, 299, 1165–1172. [Google Scholar] [CrossRef]
  276. Pourmoghadam, Z.; Soltani-Zangbar, M.S.; Sheikhansari, G.; Azizi, R.; Eghbal-Fard, S.; Mohammadi, H.; Siahmansouri, H.; Aghebati-Maleki, L.; Danaii, S.; Mehdizadeh, A.; et al. Intrauterine administration of autologous hCG- activated peripheral blood mononuclear cells improves pregnancy outcomes in patients with recurrent implantation failure; A double-blind, randomized control trial study. J. Reprod. Immunol. 2020, 142, 103182. [Google Scholar] [CrossRef]
  277. Li, S.; Wang, J.; Cheng, Y.; Zhou, D.; Yin, T.; Xu, W.; Yu, N.; Yang, J. Intrauterine administration of hCG-activated autologous human peripheral blood mononuclear cells (PBMC) promotes live birth rates in frozen/thawed embryo transfer cycles of patients with repeated implantation failure. J. Reprod. Immunol. 2017, 119, 15–22. [Google Scholar] [CrossRef]
  278. Conforti, A.; Longobardi, S.; Carbone, L.; Iorio, G.G.; Cariati, F.; Campitiello, M.R.; Strina, I.; Palese, M.; D’Hooghe, T.; Alviggi, C. Does Intrauterine Injection of hCG Improve IVF Outcome? A Systematic Review and a Meta-Analysis. Int. J. Mol. Sci. 2022, 23, 12193. [Google Scholar] [CrossRef]
  279. Robertson, S.A.; Guerin, L.R.; Bromfield, J.J.; Branson, K.M.; Ahlstrom, A.C.; Care, A.S. Seminal fluid drives expansion of the CD4+CD25+ T regulatory cell pool and induces tolerance to paternal alloantigens in mice. Biol. Reprod. 2009, 80, 1036–1045. [Google Scholar] [CrossRef]
  280. Sharkey, D.J.; Macpherson, A.M.; Tremellen, K.P.; Robertson, S.A. Seminal plasma differentially regulates inflammatory cytokine gene expression in human cervical and vaginal epithelial cells. Mol. Hum. Reprod. 2007, 13, 491–501. [Google Scholar] [CrossRef]
  281. Gunther, V.; Alkatout, I.; Meyerholz, L.; Maass, N.; Gorg, S.; von Otte, S.; Ziemann, M. Live Birth Rates after Active Immunization with Partner Lymphocytes. Biomedicines 2021, 9, 1350. [Google Scholar] [CrossRef]
  282. Agrawal, S.; Pandey, M.K.; Mandal, S.; Mishra, L.; Agarwal, S. Humoral immune response to an allogenic foetus in normal fertile women and recurrent aborters. BMC Pregnancy Childbirth 2002, 2, 6. [Google Scholar] [CrossRef]
Figure 1. Schematic immune changes during decidualization and early pregnancy. During the menstrual cycle, the decidualization starts in the second half of the secretory. Exposure to semen and seminal plasma activates both inflammatory as well as anti-inflammatory mechanisms. Anti-inflammatory cells and factors (green) contribute to the tolerance towards semen and the fetus. This includes tolerogenic macrophages (M2c), regulatory T cells (Treg), interleukin (IL)-10, transforming growth factor (TGF)-β, and human leukocyte antigen (HLA)-G. Type 1 inflammatory processes (red) support mild tissue destruction, which is necessary for tissue remodeling during implantation and placentation. This includes T helper cells (Th)1, inflammatory macrophages (M1), and several factors, such as IL-1β, -6, -8, tumor necrosis factor (TNF)-α, granulocyte-macrophage colony-stimulating factor (GM-CSF) and the chemokines CCL5, CXCL10, -16. Together with rather type 2 inflammatory effects (yellow), tissue remodeling and angiogenesis are induced. This is mediated by “wound healing”-like macrophages (M2a/d), Th2, and factors such as IL-4, -17, -22, and human chorionic gonadotropin (hCG). In the decidual environment, otherwise, cytotoxic cells, such as natural killer (NK) cells and cytotoxic T lymphocytes (CTL), are less cytotoxic and rather secrete angiogenic factors. Moreover, cells such as innate lymphoid cells type 3 (ILC3) support both a microenvironment that favors tissue remodeling as well as mediating tolerance. All three immunological branches need to be tightly controlled in order to support the optimal development of early pregnancy. A shift towards tolerance or away from inflammation can cause shallow placentation and an insufficient remodeling of spiral arteries. A shift away from tolerance towards inflammation can cause the rejection of the conceptus. After completion of placentation, the immune balance shifts towards tolerance in order to maintain the pregnancy. Decidual tissue remodeling and, thus, inflammatory processes are barely needed anymore until the induction of labor.
Figure 1. Schematic immune changes during decidualization and early pregnancy. During the menstrual cycle, the decidualization starts in the second half of the secretory. Exposure to semen and seminal plasma activates both inflammatory as well as anti-inflammatory mechanisms. Anti-inflammatory cells and factors (green) contribute to the tolerance towards semen and the fetus. This includes tolerogenic macrophages (M2c), regulatory T cells (Treg), interleukin (IL)-10, transforming growth factor (TGF)-β, and human leukocyte antigen (HLA)-G. Type 1 inflammatory processes (red) support mild tissue destruction, which is necessary for tissue remodeling during implantation and placentation. This includes T helper cells (Th)1, inflammatory macrophages (M1), and several factors, such as IL-1β, -6, -8, tumor necrosis factor (TNF)-α, granulocyte-macrophage colony-stimulating factor (GM-CSF) and the chemokines CCL5, CXCL10, -16. Together with rather type 2 inflammatory effects (yellow), tissue remodeling and angiogenesis are induced. This is mediated by “wound healing”-like macrophages (M2a/d), Th2, and factors such as IL-4, -17, -22, and human chorionic gonadotropin (hCG). In the decidual environment, otherwise, cytotoxic cells, such as natural killer (NK) cells and cytotoxic T lymphocytes (CTL), are less cytotoxic and rather secrete angiogenic factors. Moreover, cells such as innate lymphoid cells type 3 (ILC3) support both a microenvironment that favors tissue remodeling as well as mediating tolerance. All three immunological branches need to be tightly controlled in order to support the optimal development of early pregnancy. A shift towards tolerance or away from inflammation can cause shallow placentation and an insufficient remodeling of spiral arteries. A shift away from tolerance towards inflammation can cause the rejection of the conceptus. After completion of placentation, the immune balance shifts towards tolerance in order to maintain the pregnancy. Decidual tissue remodeling and, thus, inflammatory processes are barely needed anymore until the induction of labor.
Life 13 01097 g001
Figure 2. Comparison of peripheral vs. decidual leukocyte tasks. During decidualization, immune cells are recruited to the decidua and locally adjusted towards rather tolerogenic and implantation-supporting functions. Since they belong to the group of innate lymphoid cells (ILCs), NK cells are the most abundant cells in the decidua. The second most abundant cells are macrophages. Due to their vast secretory capacity, all the shown cells are not only adjusted by the decidual environment but also participate in the creation of an implantation-supporting milieu as well. Frequencies in the periphery and in the decidua in early pregnancy and phenotypic characteristics are shown.
Figure 2. Comparison of peripheral vs. decidual leukocyte tasks. During decidualization, immune cells are recruited to the decidua and locally adjusted towards rather tolerogenic and implantation-supporting functions. Since they belong to the group of innate lymphoid cells (ILCs), NK cells are the most abundant cells in the decidua. The second most abundant cells are macrophages. Due to their vast secretory capacity, all the shown cells are not only adjusted by the decidual environment but also participate in the creation of an implantation-supporting milieu as well. Frequencies in the periphery and in the decidua in early pregnancy and phenotypic characteristics are shown.
Life 13 01097 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Stope, M.B.; Mustea, A.; Sänger, N.; Einenkel, R. Immune Cell Functionality during Decidualization and Potential Clinical Application. Life 2023, 13, 1097. https://doi.org/10.3390/life13051097

AMA Style

Stope MB, Mustea A, Sänger N, Einenkel R. Immune Cell Functionality during Decidualization and Potential Clinical Application. Life. 2023; 13(5):1097. https://doi.org/10.3390/life13051097

Chicago/Turabian Style

Stope, Matthias B., Alexander Mustea, Nicole Sänger, and Rebekka Einenkel. 2023. "Immune Cell Functionality during Decidualization and Potential Clinical Application" Life 13, no. 5: 1097. https://doi.org/10.3390/life13051097

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