Next Article in Journal
Potential Anti-Inflammatory and Chondroprotective Effect of Luzula sylvatica
Previous Article in Journal
NLRP3 Inflammasome: From Pathophysiology to Therapeutic Target in Major Depressive Disorder
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Update Immune-Regulatory Role of Pro- and Anti-Inflammatory Cytokines in Recurrent Pregnancy Losses

1
Department of Obstetrics, The First Hospital of China Medical University, Shenyang 110001, China
2
The First Hospital of China Medical University, Shenyang 110001, China
3
Reproductive Medicine and Immunology, Obstetrics and Gynecology, Clinical Sciences Department, Chicago Medical School, Rosalind Franklin University of Medicine and Science, Vernon Hills, IL 60061, USA
4
Clinical Immunology Laboratory, Foundational Sciences and Humanities, Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(1), 132; https://doi.org/10.3390/ijms24010132
Submission received: 14 October 2022 / Revised: 12 December 2022 / Accepted: 15 December 2022 / Published: 21 December 2022
(This article belongs to the Section Molecular Immunology)

Abstract

:
Recurrent pregnancy losses (RPL) is a common reproductive disorder with various underlying etiologies. In recent years, rapid progress has been made in exploring the immunological mechanisms for RPL. A propensity toward Th2 over Th1 and regulatory T (Treg) over Th17 immune responses may be advantageous for reproductive success. In women with RPL and animals prone to abortion, an inordinate expression of cytokines associated with implantation and early embryo development is present in the endometrium or decidua secreted from immune and non-immune cells. Hence, an adverse cytokine milieu at the maternal-fetal interface assaults immunological tolerance, leading to fetal rejection. Similar to T cells, NK cells can be categorized based on the characteristics of cytokines they secrete. Decidual NK (dNK) cells of RPL patients exhibited an increased NK1/NK2 ratio (IFN-γ/IL-4 producing NK cell ratios), leading to pro-inflammatory cytokine milieu and increased NK cell cytotoxicity. Genetic polymorphism may be the underlying etiologies for Th1 and Th17 propensity since it alters cytokine production. In addition, various hormones participate in cytokine regulations, including progesterone and estrogen, controlling cytokine balance in favor of the Th2 type. Consequently, the intricate regulation of cytokines and hormones may prevent the RPL of immune etiologies. Local or systemic administration of cytokines or their antagonists might help maintain adequate cytokine milieu, favoring Th2 over Th1 response or Treg over Th17 immune response in women with RPL. Herein, we provided an updated comprehensive review regarding the immune-regulatory role of pro- and anti-inflammatory cytokines in RPL. Understanding the roles of cytokines involved in RPL might significantly advance the early diagnosis, monitoring, and treatment of RPL.

1. Introduction

Recurrent pregnancy losses (RPL) is a common reproductive disorder affecting 2–5% of reproductive-age women [1], which is defined as two or more pregnancy losses before 20 weeks of pregnancy by the American Society for Reproductive Medicine [2]. RPL can be caused by embryonic chromosomal, immunological, anatomical, environmental, and thrombotic abnormalities [3,4]. Approximately 50% of RPL patients have unknown causes, called unexplained RPL (uRPL) [5]. Rapid progress has been made in understanding the immunological mechanisms for RPL [6]. Maternal immune abnormalities are often present in women with uRPL [7,8], such as an increased natural killer (NK) cell count and/or activity [9,10] and a tilted T helper (Th)1 over Th2 immune response, which is thought to be harmful to pregnancy [11]. The classification of T cell subsets has extended beyond Th1 and Th2 cells to more complex regulatory T cell (Treg) and Th17 cell subsets [12]. Women with RPL have the propensity to Th17 over Treg cell immunity [13]. Th17 cells produce interleukin (IL)-17, a pro-inflammatory cytokine that promotes inflammation and maternal-fetal rejection and interacts with Th1 cells, further contributing to the immunopathology of RPL [14]. Contrarily, Treg cells mediate maternal-fetal tolerance [15], allowing an embryo, a semi-allograft, to live without rejection in the uterus [7].
Regulatory uterine NK (uNK) cells refer to highly granulated NK cells located in the endometrium with the phenotype of CD56++/CD16 [16]. They have the ability to secrete a range of cytokines [16]. In normal pregnancy, increased regulatory uNK cells are pivotal for maintaining reproductive success since they play a fundamental role in trophoblast invasion, spiral artery remodeling, and appropriate placentation [17,18,19]. Decreased number or dysfunction of regulatory uNK cells is closely related to RPL [20].
The interaction between tolerogenic dendritic cells (DCs) and Treg cells in pregnancy is pivotal for sustaining maternal-fetal immune tolerance. Tolerogenic DCs promote the proliferation and differentiation of Treg cells by increasing the expression of IL-10 [21]. These special Treg subsets play a crucial role in avoiding maternal immune response against embryo-paternal derived antigens. Furthermore, tolerogenic DCs interacting with trophoblasts could elevate the proportion of Tregs FoxP3+ [22]. However, this phenomenon does not exist in DCs that do not interact with trophoblast cells [22]. Similarly, NK cells express killer cell immunoglobulin-like receptors (KIRs), which interact with HLA-C derived from extravillous trophoblast cells (EVTs). The improper combination between KIR and HLA-C affects the ability of NK cells to secrete cytokines, including granulocyte-macrophage colony-stimulating factor (GM-CSF), thus participating in the occurrence of RPL [4].
Various cytokines play significant roles in the maintenance of pregnancy. The ability of embryos to avoid immune rejection is partially facilitated by the presence of fundamental cytokines in the peripheral blood and/or at the maternal-fetal interface. Therefore, cytokine gene polymorphisms, which modify cytokine production levels and functions, may contribute to RPL. Cytokines are classified as pro-inflammatory (Th1) or anti-inflammatory (Th2), depending on their functions. Th1 immunity induces a cell-mediated cytotoxic response to intracellular pathogens [7], whereas Th2 immunity primarily generates humoral immunity and immune tolerance [23]. Th1 pro-inflammatory cytokines are mostly tumor necrosis factor (TNF)-α, interferon (IFN)-γ, and IL-1β; Th2 cytokines are predominantly IL-4, IL-5, IL-10, and IL-13 [24,25]. During the implantation window, pro-inflammatory cytokines promote the invasion of trophoblast cells and endometrial neovascularization. However, prolonged or overexposure to pro-inflammatory cytokines may harm the pregnancy, resulting in miscarriage.
The endometrium secretes copious amounts of cytokines throughout the follicular and luteal phases of the menstrual cycle [26], contributing to a favorable uterine milieu for the preparation of implantation and placental development. The CD4+ T lymphocytes are the primary source of cytokines. Additionally, NK cells, macrophages, epithelial cells, mesenchymal cells, and other leucocytes produce cytokines at the maternal-fetal interface [27]. During trophoblast implantation, pregnancy hormones and direct stimulation by trophoblast cells improve the endometrial receptivity while the endometrium is gradually transformed into the decidua. Cytokines and receptors on decidual cells are primarily responsible for interacting with trophoblast cells. Cytokines have multiple functions, including regulation of embryo implantation, placental development, cytotrophoblast proliferation, vascular remodeling, trophoblast invasion, cell death, and the induction of embryo tolerance in the uterus [28,29].
Although the contribution of cytokines to RPL has been widely studied [30,31,32], their precise roles need further investigation. This review analyzes the updated immune-regulatory role of pro- and anti-inflammatory cytokines in maternal peripheral blood and endometrium, including Th1/Th2, Treg/Th17, and NK cell-related cytokines. Although other cells also produce cytokines at the maternal-fetal interface, such as trophoblasts, stromal cells, and macrophages, they are not within the scope of this review. Understanding the roles of cytokines involved in RPL might significantly advance the early diagnosis, monitoring, and treatment of RPL. This review highlights relevant studies with significant discoveries that merit additional investigation. Two independent scholars conducted a comprehensive publication search to collect the articles with the following keywords: “recurrent miscarriage,” “recurrent pregnancy losses,” “recurrent spontaneous abortion,” and “cytokine” from PubMed, Web of Science, and Google Scholar database. All articles were published in English from January 1985 to September 2022. Letters were not included.

2. Pro- and Anti-Inflammatory Cytokines in Maternal Peripheral Blood

According to a prior animal study exploring the Th1 immune response associated with RPL, trophoblast antigen could activate lymphocytes from abortion-prone female mice to produce pro-inflammatory cytokines, such as TNF-α [33]. The ratios of Th1/Th2 cytokine-producing T helper cells from RPL patients (IFN-γ/IL-4, IFN-γ/IL-10, TNF-α/IL-4 and TNF-α/IL-10 producing Th cell ratios) were considerably higher than normal pregnant women after in vitro stimulation of peripheral blood mononuclear cells (PBMCs) [30]. Another study investigated the Th1/Th2 cytokine ratios in the peripheral blood of uRPL women (n = 44) and normal pregnant women (n = 42). They reported that uRPL women had a higher IFN-γ/IL-4 cytokine ratio [34]. According to this study, the IFN-γ/IL-4 cytokine ratio had a sensitivity of 84.09% and a specificity of 69.05% for diagnosing RPL [34]. The area under the curve (AUC) of IFN-γ/IL-4 was 0.821 (p < 0.05) [34]. Hence, Th1/Th2 ratios may be utilized to predict pregnancy outcomes, closely monitor pregnancy, and swiftly treat patients at risk of miscarriage. Contrarily, a Th1-to-Th2 switch was reported in RPL patients compared to healthy pregnant women of comparable gestational weeks [35], which was different from other studies [36,37,38]. Differences in these studies may be related to the selection of the control group and the timing of the investigation. In the study of Bates et al. [35], 25 pregnant women undergoing voluntary termination of the pregnancy were included but obstetrical and infertility histories of the control group were not reported. Th1/Th2 ratios vary during each developmental phase of pregnancy [39,40], but in the study of Makhseed et al., the study and control groups were investigated in different gestational weeks [38]. It is noteworthy that several animal studies failed to document that a single aberrant cytokine, either Th1 or Th2 cytokine, can affect pregnancy outcome. IL-10 knock-out (KO) mice did not have altered pregnancy outcomes. Additionally, IL-4, IL-5, IL-9, and IL-13 KO mice did not have decreased live birth rate or offspring weight [41]. Therefore, a complicated cytokine network may determine the balance between Th1/Th2 immune responses during pregnancy with a propensity toward Th2 immune response after implantation is over, which in turn, again biased to Th1 immune response at the time of parturition [7].

2.1. TNF-α

TNF-α is an important cytokine produced by Th1 cells and is known to have diverse immune-modulatory functions during various stages of human reproduction. TNF-α promotes trophoblast invasion by regulating the expression and secretion of inflammatory factors derived from endometrial stromal cells, such as IL-17 [42] (Figure 1). Moreover, TNF-α induces vascular endothelial growth factor (VEGF) secretion from trophoblasts, which aids in embryo implantation in the uterus (Table 1) [43]. By coordinating angiogenesis, VEGF is crucial for embryo implantation and placental development [43]. In addition, TNF-α stimulates the production of IL-10, which decreases inflammatory activity [44]. After in-vitro fertilization and embryo transfer (IVF-ET), TNF-α producing CD3+/CD4+ T cell numbers were raised in RPL patients [45]. However, despite having elevated serum TNF-α expression, these individuals had low IL-10 levels [45]. Neuroendocrine factors such as glucocorticoids may affect the serum TNF-α expression but do not produce a secondary IL-10 increase [46]. Therefore, women with RPL may lack neuro-endocrine regulators, resulting in a TNF-α/IL-10 ratio imbalance [45].
Most researchers believe that the serum level of TNF-α in RPL women is greater than in normal pregnant women [37,57,58] (Table 2). In addition, plasma TNF-α levels increase in RPL patients during the first trimester regardless of the outcome of the pregnancy. Its level in secondary RPL was higher than in primary RPL, possibly due to genetic factors [59]. Patients with secondary RPL may have been sensitized by fetal or trophoblast antigen in a prior pregnancy and generated a humoral or cytotoxic reaction to the trophoblast antigen in the subsequent pregnancy, resulting in inflammatory responses in early pregnancy. In contrast, mitogen-activated PBMCs from RPL women with successful pregnancy outcomes exhibited comparable TNF-α levels to those with miscarriages but lower than normal pregnant females [35]. These findings raised concerns about whether TNF-α is detrimental to pregnancy outcomes. TNF-α induces RPL mostly by increasing the expression of pro-apoptotic genes on the embryonic membrane surface [60], which leads to membrane degradation [61]. Additionally, TNF-α may induce antiphospholipid antibody-related placental injury, leading to miscarriage [62].

2.2. IFN-γ

IFN-γ is found in the human endometrium [72]. Both inflammatory cells in the decidua and developing embryos release IFN-γ, which maintains the pregnancy by regulating the expression of IL-6, monocyte chemotactic protein (MCP)-1, and macrophage colony-stimulating factor (M-CSF) in early pregnancy [73]. In the rodent model, IFN-γ facilitates implantation through vessel recasting, vascular development at the implantation site, and maintaining decidual tissues during placental development [28]. However, an excessive level of IFN-γ is harmful to the embryo. Most literature has reported significantly increased IFN-γ expression in spontaneous abortion or RPL patients [37,64,74,75], although Bates et al. discovered no difference between RPL women and normal controls [35]. IFN-γ inhibits the production of GM-CSF, which is indispensable for a successful pregnancy [76]. Moreover, IFN-γ influences the expression of proteins involved in the endometrial adhesion of embryos [77].

2.3. Transforming Growth Factor (TGF)-β

TGF-β is a multifunctional cytokine with various regulatory roles [78]. It may stimulate the development of Th1 immune cells [79] (Figure 2). At the same time, it controls the cytokine network and maintains maternal immune tolerance [80]. Controversies surround the expression levels of TGF-β in RPL patients. Plasma TGF-β levels in RPL patients have been reported to be significantly higher than in healthy pregnant women [63,81], although contradictory reports were also reported [64,65]. The increased TGF-β1 could inhibit trophoblast invasion by upregulating kisspeptin expression [82].

2.4. IL-1β

IL-1β promotes inflammation, induces B cell development and proliferation, and activates NK and T cells [83]. It impacts endometrial growth during embryo implantation [52] and regulates placentation [84]. In addition, IL-1β can promote the expression of matrix metalloproteinase (MMP), thus participating in the regulation of trophoblast invasion [85]. During pregnancy, β-HCG induces the production of IL-1β from decidual T cells [86]. In a retrospective study of 56 RPL patients and 56 normal pregnant women with antinuclear antibody (ANA), IL-1β predicted RPL with a sensitivity of 76.8% and a specificity of 91.1% [87]. This study indicates that IL-1β plays a crucial role in developing RPL and may predict RPL in ANA-positive women [87]. In animal experiments, the pregnancy rate was decreased when IL-1β receptor antagonist was administered prior to embryo transfer [88].
In addition to its primary function in pregnancy, IL-1β contributes to blood clotting. IL-1β increases the amounts of proteins such as ICAM-1, aυβ3, and MCP-1, which increase platelet stability and the risk of thrombosis [89]. The pro-thrombotic condition generated by these proteins causes the production of microthrombi during early pregnancy, ultimately resulting in miscarriage. Therefore, anticoagulant therapy may be useful for RPL patients with elevated IL-1β expression.

2.5. IL-33

IL-33 belongs to the IL-1 family [90]. It enhances Th1 and Th2 immune responses [91] and contributes to the Th17 immune response [92]. Additionally, it activates NK cells and promotes IFN-γ production from NK cells [93,94]. During a normal pregnancy, IL-33 enhances Th2 immunity and maintains maternal immune tolerance to the fetus [95].
Serum IL-33 level was considerably lower in RPL women compared to those with normal pregnancies (p < 0.05) [66], as validated by another investigation [96]. However, others discovered that the serum IL-33 levels in pregnant women who were going to miscarry were much higher than those of normal pregnancies (>6 weeks of gestation) [97]. The elevated IL-33 may function during pregnancy as a compensating strategy to save the fetus. However, the mechanism by which decreasing IL-33 expression and leading to RPL remains unclear. A successful pregnancy needs the activation of the IL-33/ST2 pathway, and IL-33 is required for embryo implantation in the mice model [56]. Furthermore, IL-33 played a vital role in the placental formation and maintenance of healthy NF-κB and ERK1/2 pathways [98]. These activated pathways upregulate the CCL2/CCR2 axis, subsequently enhancing the proliferation and invasion of decidual stromal cells [98]. By boosting Th2 immunity at the maternal-fetal interface, CCL2 creates a favorable environment for the fetus [98]. The reduced serum IL-33 level in RPL may be attributable to the down-regulated CCL2.

2.6. IL-4

IL-4 is a Th2 cytokine, and its level in the culture supernatants of PBMCs from RPL was lower than that of normal pregnant women [35], although a contradictory study was reported [64]. However, the IL-4 level in the peripheral blood of RPL patients was not linked with pregnancy outcomes [35].

2.7. IL-6

IL-6 is essential for embryo implantation and placentation [99]. During parturition, IL-6 levels were reported to be higher in maternal serum [100], amniotic fluid [101], and placental tissues [102] than without labor. IL-6 has multiple functions, such as promoting inflammatory responses and suppressing TNF-α expression [103]. IL-6 level was significantly higher in the culture supernatant of PBMCs from normal pregnancies than RPL [37]. Additionally, RPL women with low IL-6 levels during early pregnancy had poor pregnancy outcomes [57]. When PBMCs were reconstituted and stimulated with autologous trophoblast or choriocarcinoma cell antigens, leukocytes from RPL patients secreted persistently lower levels of IL-6 than those of healthy controls [36].

2.8. IL-10

IL-10 is a Th2 cytokine that suppresses the Th1 immune response by reducing the expressions of TNF-α, IFN-γ, and IL-1 [104]. IL-10 is produced by activated Th2 cells and promotes embryo development by maintaining immunological tolerance [105]. Administration of IL-10 to the abortion-prone mice reduced embryo loss [106], whereas IL-10-deficient mice were more likely to experience miscarriages due to inflammatory changes compared to wild-type mice [107]. Normal pregnant women had higher serum levels of IL-10 compared to RPL patients [57]. The IL-10 level in the peripheral blood of uRPL women was significantly lower, and a higher level was related to healthy pregnancies [36,37,64,67,108]. However, there was a markedly higher amount of IL-10 in the supernatant of cultured PBMCs from pregnant RPL women compared to those of normal pregnant controls, supporting a notion that Th2 immunity was enhanced rather than repressed in RPL [35]. Moreover, the peripheral blood IL-10 level in RPL patients did not predict pregnancy outcomes [35]. IL-10 has a significant role in linking immune responses and angiogenesis and suppresses endoplasmic reticulum (ER) pressure, promoting protein composition and energy stabilization [109]. Increased ER stress leads to the activation of pro-inflammatory responses and placental abnormalities [110].

3. Treg and Th17-Related Cytokines in Maternal Peripheral Blood

A Treg/Th17 cell imbalance was identified in RPL women [13]. Compared to healthy pregnant women, RPL patients have fewer Treg cells [111] and more Th17 cells in the peripheral blood and decidua [14,32,69,112,113,114]. Due to their pro-inflammatory activities, Th17-related cytokines, including IL-17, can cause embryo rejection, whereas Treg cell-regulated cytokines such as IL-10 and TGF-β may enhance immunological tolerance and improve pregnancy outcomes [112,115].

3.1. IL-6

IL-6 is a crucial cytokine that represses the proliferation of Treg cells and promotes the differentiation of Th17 cells (Figure 3). Serum IL-6 and soluble IL-6 receptor levels were elevated in RPL women, while the inhibitor of IL-6/IL-6R signal transduction, soluble glycoprotein (gp)130, was reduced [116]. Following paternal lymphocyte immunotherapy, the expression of soluble IL-6R was reduced, whereas the soluble gp130 level and Treg cell numbers were increased [116]. These data imply that IL-6 signaling is crucial in regulating the Treg to Th17 cell ratio. Moreover, constant Toll-like receptor (TLR) activation or the suppression of Treg function by IL-6 has been documented [117], indicating that the decline of Treg cell function in RPL patients was associated with the IL-6-related inflammatory response.

3.2. IL-7

IL-7 modulates Th17 cell development from naïve T cells [48]. IL-7 was revealed to promote the differentiation of Th17 cells via inhibiting forkhead/winged helix transcription factor (FOXP3) expression in Treg cells and subsequently inhibit the differentiation of Treg cells induced by TGF-β [7,118]. Then, IL-7 disturbs the Treg/Th17 equilibrium in favor of Th17 immunity, ultimately resulting in RPL [119]. Significantly elevated expressions of IL-7 in the decidua of women with spontaneous abortion and RPL indicate that IL-7 likely plays a significant role in promoting pro-inflammatory immune response at the embryo-maternal interface [119]. However, the study was limited by the lack of individual IL-17 investigation, which might be necessary for clinical work [119]. The underlying mechanism of the IL-7 signaling pathway was investigated in RPL by evaluating the IL-7 and IL-7R expression levels in the decidua tissues of RPL patients [32]. By administering an IL-7R inhibitor to the abortion-prone mice, FOXP3+ Treg cells (%) were dramatically increased, and Th17 cells (%) were significantly reduced [32]. When IL-7R expression was decreased in endometrial stromal cells, the IL-7 signaling pathway was blocked, and the ability of trophoblast cells to invade was diminished [32]. RPL is partially mediated by the diminished invasion of trophoblast cells [120]. In autoimmune instances, IL-7 expression is increased to compensate for decreased IL-7R [121,122]. Based on research advancements in autoimmune diseases, drugs targeting the IL-7 signaling pathway could be evaluated for the treatment of RPL [119].

3.3. IL-17

The IL-17 family consists of multiple related cytokines ranging from IL-17A to IL-17F. Among those, IL-17A and IL-17F are the most widely studied cytokines. Due to its pro-inflammatory function, the possible role of IL-17 in spontaneous abortion has received increasing interest in recent years [69,123,124,125]. However, a recent study revealed that stromal cell-derived IL-17 aggregated Th17 cells, thereby accelerating trophoblast invasion and reducing trophoblast death, indicating a favorable effect on pregnancy maintenance [126]. This result revealed a novel role for Th17 in trophoblast implantation and placental development. Similarly, another study confirmed that the peripheral blood IL-17 levels in normal pregnant women were higher than those of patients with abortion, showing that IL-17 is a protective regulatory factor for a healthy pregnancy [127]. However, the abortion patients in this study were in the first trimester, while the normal pregnant women were in the second or third trimester [127]. In contrast, the morbidity of uRPL is associated with an increased level of IL-17 in peripheral blood [48]. In the mice model, intraperitoneal injection of IL-17 into normal pregnant mice triggered miscarriages, whereas the anti-IL-17 antibody reduced the prevalence of miscarriage in abortion-prone mice [128]. Therefore, the serum level of IL-17 may predict pregnancy outcomes, and further clinical studies are warranted.

3.4. IL-22

Previous studies have indicated that IL-22 is advantageous for pregnancy. IL-22 has a role in the healing process of damaged trophoblast cells at the maternal-fetal interface because it participates in epithelial cell regeneration and tissue repair by binding to IL-22R1 on trophoblast cells [129,130]. Significantly fewer IL-22-derived CD4+ T cells were demonstrated in the decidua of RPL women who miscarried genetically normal embryos than in normal pregnant women [55]. IL-22 mRNA and its transcription factor are at the implantation site [55]. Therefore, producing IL-22 in the implantation zone is likely essential for a healthy pregnancy. Contrary to these studies, it has been reported that serum IL-22 level was elevated in RPL patients [68]. Further studies are needed to explore the immunopathogenic mechanism of IL-22 in RPL.

3.5. IL-23

When the IL-23 level is increased above the initial levels, Th17 cells expand dramatically, resulting in an imbalance of Treg/Th17 cells and consequential embryo rejection [131]. Both peripheral blood and decidual levels of IL-23 were considerably higher in RPL women than in normal pregnancies, indicating that IL-23 may be involved in the pathophysiology of RPL [67,69].

3.6. IL-27

Th17 cell expression is inhibited by IL-27, which is present at the maternal-fetal interface in both human and mouse studies [125,132,133,134]. In an experiment using mice, decreased IL-27 expression in decidua was associated with an increased incidence of miscarriage [133]. Decidual IL-27 expression in RPL patients was lower than in women with spontaneous abortion or healthy pregnancies [125]. IL-27 inhibited the level of IL-17 and raised the expression of IL-10 in a dose-dependent manner but had no effect on the expression of TGF-β in decidua [125]. Additionally, IL-27 affects IL-17 and IL-10 levels in women with RPL, which is secreted mostly by Th17 and Treg cells [125]. These findings suggest that IL-27 can influence specific immune cells in the decidua during pregnancy.

3.7. IL-35

CD4+ Foxp3+ Treg cells produce the majority of IL-35 [135], and trophoblast cells also secrete IL-35 during early pregnancy [136]. IL-35 is critical for maintaining the inhibitory action of Treg cells [135]. The expression of IL-35 in the serum of RPL patients was considerably lower than that of normal pregnant women, indicating that IL-35 has a role in maintaining a pregnancy [70,71].
According to the studies mentioned above, the expressions of Th1 cytokines in the peripheral blood of RPL patients are significantly increased, including TNF-α and IFN-γ. On the other hand, the circulating levels of Th2 cytokines, such as IL-6 and IL-10, were remarkably lower in RPL females compared to normal pregnant women. IL-10 seems to be a core Th2 cytokine since it inhibits the activity of Th1 cytokines and has the function of immune regulation. Nowadays, maternal-fetal immune tolerance has gradually expanded from the Th1/Th2 pattern to Th1/Th2/Treg/Th17 pattern. Therefore, Th17 cytokines IL-17 and IL-22 may play central roles in the etiology of RPL, which needs further research.

4. Endometrial Cytokine Imbalance in RPL

The imbalance of cytokine expression in the endometrium or decidua is relevant for RPL (Table 3). In both women with RPL and abortion-prone animal models, an inordinate expression of a few cytokines associated with implantation and early embryo development was present in immune and non-immune cells in the endometrium or decidua. This creates an adverse cytokine milieu, which assaults immunological tolerance and causes fetal rejection. Due to the intricacy of cytokines in the endometrium, which undergo dramatic changes during pregnancy, it is difficult to define the precise function of each cytokine in RPL. In addition, it is hard to identify the source of a specific cytokine because numerous immune and non-immune cells can produce the same cytokine. Despite the limitations highlighted, some cytokines have been reported to play a significant role in this illness.
Before the blastocyst reaches the endometrium, stromal cells produce TNF-α and IL-1β to trigger an inflammatory response during the implantation window [49]. Human endometrial cells can express TNF-α mRNA [142,143]. The expression of TNF-α varies throughout the menstrual cycle and gradually increases in the late luteal phase [144]. In addition, inhibiting TNF-α reduces stress-induced miscarriage in mice [145]. RPL patients had elevated levels of IFN-γ mRNA and protein in their endometrium and decidua [138]. TNF-α and IFN-γ can impede embryo growth by inducing apoptosis of trophoblast cells [146].
In the first trimester, IL-1 family cytokines are predominant at the maternal-fetal interface [76]. IL-1β contributes to decidualization by increasing the production of cyclooxygenase-2 (COX2) and prostaglandin E2 (PGE2) [85]. In a study of placental tissues (6–13 weeks of gestation) taken from 15 normal pregnancies and 15 RPL patients, significantly elevated levels of IL-1β were demonstrated in the decidua of the RPL group [31]. These results illuminated the complicated function of IL-1β at the maternal-fetal interface and its effect on abortion [31]. Furthermore, the protein expression of IL-18 in decidual tissues of RPL patients was much higher than that of normal pregnant women, indicating that precise regulation of IL-18 was necessary for a healthy pregnancy [141].
IL-6 plays a constructive role in decidualization and increases endometrial receptivity, thus facilitating the regulation of maternal-fetal interaction [85]. Patients with RPL have lower IL-6 mRNA and protein expressions in the mid-luteal endometrial tissues [139,147,148]. However, another study showed that IL-6 mRNA and protein expressions in the decidual tissues of RPL patients were elevated compared to normal pregnant controls [137]. The disparities in these studies may be attributable to the heterogeneity of the different populations and the various underlying disorders.
IL-10 expression at the maternal-fetal interface was documented [149,150]. It is a potent cytokine that induces the production of tolerogenic DCs [151], which are essential for sustaining maternal-fetal immunological tolerance [152]. Patients with RPL whose decidual tissues express low levels of IL-10 have an impaired immune protection system [138,153,154,155]. Decreased production of IL-10 with the increased synthesis of inflammatory factors, may provide a condition that induces early miscarriage [156]. In addition, IL-10 and other cytokines, such as TGF-β, can help coordinate the development of Treg cells in the decidua [157]. Furthermore, the expression of IL-4 was remarkably lower in the decidua of RPL patients compared with normal pregnancies [138,158]. However, the IL-17A expression in the decidua tissue of RPL patients was considerably higher than in healthy pregnant women [112].
A recently published report indicates that IL-35 is expressed in primary early trophoblast cells and the HTR-8 trophoblast cell line. Trophoblasts could transform naive conventional T cells into iTR 35 cells in the presence of IL-35. Murine spontaneous abortion models have decreased expressions of both IL-35 and iTR 35 cells at the fetomaternal interface, suggesting that IL-35 is crucial for sustaining fetomaternal tolerance [159].
IL-11 is a crucial cytokine associated with decidualization [160] and placentation [161]. The reduced expression of IL-11 in the endometrial tissues of RPL shows that it plays a significant role in preventing abortion [140]. Therefore, RPL might be potentially addressed by correcting IL-11 signaling deficiencies in the endometrium [162]. Defective decidualization and reduced proliferation of uterine stromal cells were observed in IL-11R KO animals [163,164]. However, these in vivo intervention experiments conducted in mice can not fully reflect changes in humans. Defective implantation and placentation are major concerns in human pregnancy since spontaneous miscarriage occurs in 30% of human pregnancies, most happening before a clinical pregnancy test [165]. In addition, IL-11 is expressed in the human endometrium [166]. Thus, it is necessary to conduct a relevant investigation to identify whether the changes in IL-11 and its receptors are related to RPL.
It is known that RPL is related to a reduction in decidual TGF-β compared to healthy pregnant women due to a decreased production of TGF-β from decidual DCs [153], a decreased mRNA expression of TGF-β [137], or a smaller proportion of TGF-β+ Tregs in the decidua [154]. Furthermore, when trophoblast cells obtained after uterine curettage were investigated, those from RPL women (n = 11) had substantially decreased expression of TGF-β3 compared to those from normal pregnant women (n = 20) [167]. Since TGF-β is involved in stimulating implantation and early embryo development, a reduction in TGF-β may be causally related to the immunological etiology of RPL. Furthermore, a recent study showed that the TGF-β signaling pathway promotes the differentiation of placental EVTs into decidual EVTs, playing a vital role in decidualization [168].
According to a previous report [169], Th1 cytokines stimulate pro-coagulant factors on vascular endothelial cells, leading to the production of thrombus and inflammation at the maternal-fetal interface. When fibrinogen-like protein 2 (fgl2), one of the pro-coagulants, is blocked by anti-fgl2, spontaneous abortions were prevented in the mice model. In human pregnancies, enhanced fgl2 expression was demonstrated in the trophoblast cells of failed human pregnancies [170]. Fgl2 plays a role in the conversion of prothrombin to thrombin, the accumulation of fibrin, and the activation of polymorphonuclear leukocytes, which in turn impairs placental blood flow. Th2 cytokines suppress and counteract the Th1 response. In addition, Th1 cytokines can promote apoptosis and destroy the trophoblast barrier between a semi-autogenous fetus and the maternal immune system, leading to miscarriage. Th1 cytokines can also exert their effects by increasing the proliferation of NK cells, lymphokine-activated killer (LAK) cells, and cytotoxic T lymphocyte (CTL) cells, which destroy trophoblast cells and result in fetal absorption in murine pregnancies [171].

5. Cytokines Produced by NK Cells

Similar to T cells, NK cells can be categorized based on the cytokines they secrete [172,173,174,175]. In the peripheral blood of non-pregnant women, IFN-γ or TNF-α-producing and IL-4, IL-5, IL-13, TGF-β or IL-10-non-producing CD56bright NK cells and CD56dim NK cells (NK1) are the most abundant cells (60%) [176]. However, the proportion of IL-4, IL-5, and IL-13-producing, IFN-γ, TNF-α, IL-10, or TGF-β-non-producing NK2 cell populations is extremely low [176]. NK3 cells refer to the TGF-β-producing, IFN-γ, TNF-α, IL-4, IL-5 or IL-13-non-producing cells, and the IL-10-producing cells might be NKr1 cells [176]. In a normal pregnancy, NK cells switch from type 1 to type 2 immune response [177,178]. The numbers of circulating NKr1 and decidual NK3 cells are reduced in spontaneous abortion patients, suggesting that these cells are key players in maintaining pregnancy by regulating maternal immune response [176].
Elevated NK cell cytotoxicity has been reported in the peripheral blood of RPL patients [179]. In RPL patients, NK cytotoxicity may be utilized as a biomarker to predict future abortions [180,181]. However, a contradictory study was reported [182], which could be attributed to the different experimental methodologies and study designs.
RPL patients had a considerably greater number or proportion of circulating NK cells than normal pregnant women [183,184,185]. In women with RPL, endometrial NK cell counts were significantly higher during the mid-luteal phase compared to the normal fertile controls [186]. dNK cells exhibited increased NK1/NK2 ratios (IFN-γ/IL-4 producing NK cell ratios) in women with RPL [187]. However, TNF-α and IFN-γ producing uNK cells in RPL patients were lower than in normal pregnant women [188]. The proportion of IL-4 and IL-10-producing CD56bright NK cells in the peripheral blood was lower in non-pregnant women with a history of RPL than in normal fertile women [189]. In contrast, the proportion of TNF-α and IFN-γ producing NK cells increased significantly [189]. These findings indicate the presence of increased NK1/NK2 ratios in women with RPL [189].
Moreover, RPL patients have higher absolute numbers of CD56+ NK cells with intracellular IFN-γ, TGF-β, and IL-4 expressions, demonstrating that the immune system is activated in RPL women compared to normal controls [190]. Decidual NK22 cells are the primary cells to produce IL-22. IL-22 participates in host defense, mucosal homeostasis, and trophoblast cell invasion [191]. CD56bright/IL-22+ cells were negatively correlated with CD56bright NK cells producing IFN-γ or TNF-α both in peripheral blood and endometrium [191]. The mRNA and protein expressions of IL-22 in the decidua of RPL were considerably reduced [192], and diminished IL-22 levels in the decidual tissues of RPL may compromise decidual homeostasis and ultimately result in embryo rejection [192].

5.1. Cytokine in the Micro Milieu of NK Cells

The maternal-fetal interface contains IL-15 and IL-18, activating NK cells to secrete various cytokines. IL-15 is also present in the female reproductive tract [193] and stimulates the proliferation of uNK cells in both mice and humans [194,195]. By activating progesterone receptors, stromal cells released IL-15, and the level of IL-15 was positively correlated with the number of uNK cells [196]. In animal studies, uNK cells cannot differentiate in the uterus without IL-15 [197]. Similarly, IL-15 KO mice lacked uNK cells. However, a lack of uNK cells did not impact on embryo implantation and development [197]. Endometrial stromal cells produce more IL-15 during decidualization [198]. In humans, higher endometrial IL-15 expression was closely associated with RPL [199]. Similarly, IL-15 mRNA and protein expressions were significantly increased in the placental tissues of RPL patients [200]. Over-expression of IL-15 in the decidua of RPL patients may lead to the failure of implantation and angiogenesis, followed by placental injury and fetal loss [200].
IL-18 can stimulate the production of IFN-γ from uNK cells with the presence of Th1 cytokine IL-12 [201]. It may augment the level of pro-inflammatory cytokines released by macrophages at the embryo-maternal interface, followed by the activation of uNK cells. Additionally, IL-18 increases NK cell cytotoxicity [202] and stimulates Th2 cytokine production from NK cells, such as IL-4 and IL-13 [203]. IL-18 expressions in peripheral blood and endometrium of RPL patients were considerably greater than those of healthy fertile females [204]. Briefly, cytokine variations in the micro milieu of NK cells may influence NK cell activity and contribute to the pathogenesis of RPL.

5.2. uNK Cells and Angiogenic Cytokines

RPL is associated with increased endometrial vascular density, suggesting that RPL is implicated in the premature maturation of the vascular network [205]. It has been reported that uNK cells produce extensive angiogenic factors, such as VEGF-C, angiopoietin (Ang), and platelet-derived growth factor (PLGF), which are vital for angiogenesis in the endometrium [29]. During the implantation window, CD56+ uNK cells are one of the major sources of angiogenic cytokines in the endometrium [206]. Angiogenic cytokine gene array study demonstrated that the expressions of VEGF-A, Ang, and basic fibroblast growth factor (bFGF) in CD56+ uNK cells from RPL patients were considerably higher than those of normal pregnant women [206]. Differences in results may be attributed to the test method (angiogenic cytokine array vs. ELISA), experiment sensitivity, and study participants.

6. Cytokine Gene Polymorphisms in RPL

The increased expressions of pro-inflammatory cytokines may be caused partly by gene polymorphisms, and women with cytokine gene polymorphisms might be genetically prone to RPL. In addition, gene polymorphisms may affect the expression levels of cytokines, as well as their functions, which play an important role in the pathogenesis of RPL [207]. Therefore, cytokine gene polymorphisms might be utilized to predict abortion, stratify pregnancy management, and improve maternal and fetal pregnancy outcomes by increasing the surveillance of the high-risk abortion group.

6.1. TNF-α Gene Polymorphisms in RPL

In RPL patients, polymorphisms in the TNF-α gene have been extensively investigated (Table 4). Previous meta-analyses indicated that TNF-α -308G/A and -238G/A polymorphisms had no relationship with the risk of RPL [208,209]. However, subsequent meta-analyses demonstrated that the TNF-α -308G/A polymorphism was related to an increased risk of RPL [210,211] but not for TNF-α -238G/A [210,211].

6.2. IFN-γ Gene Polymorphisms in RPL

IFN-γ 874A > T gene polymorphism affects the level of IFN-γ, and the T allele increases the cytokine expression [223]. Genotype frequency of IFN-γ 874A > T polymorphism was significantly different in Argentine women with RPL from that of healthy pregnant women [221]. The frequency of AT genotype was significantly higher, and that of the AA genotype was considerably lower in patients with RPL compared with those of normal fertile women [221]. It has been observed that the TT genotype that increases IFN-γ production conspicuously boosts the risk of RPL [219]. However, other studies reported a lack of association between IFN-γ 874A > T polymorphism and RPL [207,208,222].

6.3. IL-10 Gene Polymorphisms in RPL

Different alleles at sites −1082, −592, and −819 regulate IL-10 expression [224,225]. The IL-10 −1082 GG, −592 AA, and −819 TT genotypes may enhance IL-10 expression [224,225]. It is hypothesized that phenotypes related to the underproduction of IL-10 may induce pregnancy complications. Certain IL-10 genotypes increase the chance of RPL, whereas others protect against abortion (Table 5). A recent meta-analysis of 2047 RPL patients and 2055 control pregnant women revealed an association between −1082 A/G and the incidence of RPL [226]. However, no significant association was seen between the −819C/T, 592C/A genotypes and RPL [208,226]. Four meta-analyses produced contradictory results about the association between the most extensively investigated −1082 A/G genotype and RPL. The first meta-analysis comprised six studies with 635 RPL patients and concluded that −1082 A/G1082 was unrelated to RPL [208]. The second meta-analysis comprised six studies and reached the same conclusion [227]. The third meta-analysis, which included the previous six studies [208,227] and their own data from India, indicated that the wild-type allele of −1082 A/G was protective for RPL [228]. A recent publication of the fourth meta-analysis, including 13 studies, suggested that the GG genotype increased the risk of RPL [229]. The conflicting results may be related to the diverse study populations.

6.4. Other Cytokines Gene Polymorphisms in RPL

Gene polymorphisms in other cytokines are summarized in Table 6 (IL-18), Table 7 (IL-6), and Table 8 (IL-1β and IL-1 RN). Furthermore, no significant correlation was present between the two IL-4 polymorphisms and the probability of RPL [222,233]. Additionally, women who carry the IL-2RN*2 allele are less likely to have clinical pregnancies after IVF [234].
Inconsistent results in expression levels and gene polymorphisms of various cytokines in women with RPL could be due to other factors affecting the expression levels of cytokines, differences in study design, study population, and lifestyle. Moreover, it is vital to investigate whether the reported genotype frequency of particular cytokine polymorphism can be applicable to RPL of other ethnic backgrounds, as some of the studies mentioned above only applied to a certain ethnic population and not others [250]. Multiple cytokine polymorphisms may contribute to RPL rather than a single gene polymorphism. Moreover, altered cytokine production by gene polymorphisms may not represent the immune and inflammatory change at the maternal-fetal interface since cytokines exert their effect in both autocrine and paracrine manners. Finally, the association between cytokine gene polymorphisms and RPL warrants further studies with larger study populations. Meta-analysis may solve the decreased statistical significance caused by the small sample size and obtain more accurate results.

7. Hormonal Regulation of Cytokines and RPL

Various hormones regulate the expression of pregnancy-maintaining Th2 cytokines. The decrease of estrogen (E2) and progesterone (P4) induced the production of Th1 pro-inflammatory cytokines [251]. In addition, E2 and P4 enhance Th2 anti-inflammatory response and reduce the activity of pro-inflammatory macrophages and NK cells [252]. P4 levels are similar at the maternal-fetal interface throughout implantation and early pregnancy. In humans, P4 promotes the cytokines production by T cells, such as IL-4, IL-5, leukemia inhibitory factor, and M-CSF in decidua [253], while suppressing the production of IFN-γ and TNF-α [254,255] and impeding the production of Th17 related cytokines, such as IL-17A and IL-23 in peripheral blood [256]. A few studies investigated the indications for the use of P4 in treating RPL. A large sample study involving 826 subjects revealed that compared with the placebo group, vaginal micronized progesterone plays a weak or no difference in improving the living birth rate of RPL patients [257]. Similarly, the proof of dydrogesterone for treating RPL is low certainty compared to placebo [257]. There is no sufficient data to analyze the effect of 17-α-hydroxyprogesterone or oral micronized progesterone on increasing the living birth rate of RPL patients [257]. In another study, the subgroup analysis of three experiments contained patients with three or more recurrent miscarriages [258]. P4 significantly reduced the abortion rate compared with placebo or no therapy [258]. It seems that P4 has a significant therapeutic effect on RPL. Therefore, they pointed out that P4 could be considered for treating RPL women since the abortion rate was lower and there was no augment in side effects of mothers and neonates [258].
E2 also has immunomodulatory effects. E2 levels during pregnancy have been shown to affect the Th1/Th2 cytokine balance, maintaining maternal-fetal tolerance [259]. E2 regulates cytokine expression in a dose-dependent manner. It increases IFN-γ and IL-12 levels in the peripheral blood before ovulation. However, it increases IL-10 levels throughout pregnancy, reducing the IFN-γ/IL-10 ratio during human pregnancy [260]. In the human PBMC culture study, E2 decreased the secretion of IL-2 and IFN-γ but did not affect the level of TNF-α, IL-17A, and IL-23 [256]. However, E2 administration significantly reduced the circulating IL-17 expression in allergic encephalitis mice [261]. These discrepant results imply that it is necessary to explore the effect of E2 on Th17 cytokines in pregnant women. Like P4, E2 increases peripheral blood IL-6 levels in humans [256]. Since E2 can suppress the serum levels of Th1 cytokines, such as IL-2 and IFN-γ, and increase the serum level of Th2 cytokine IL-6, it can regulate the cytokine balance in favor of the Th2-type immune response.

8. Conclusions

RPL is a disorder with numerous potential causes. Dysregulation of cytokine expression may be one of the underlying pathologies, but the mechanisms need to be explored further. Cytokines rarely exhibit activity independently; rather, they comprise a regulatory network that maintains equilibrium. Moreover, the ultimate effect of an altered cytokine level is determined by the overall modulation of the immune response rather than a mechanism by a specific cytokine. The gaps between the highest and lowest levels of a particular cytokine can be as much as tenfold; hence, one mechanism by a single cytokine cannot account for RPL. Consequently, understanding the intricate mechanisms of various cytokines may aid in establishing therapeutic modalities for RPL of immune etiologies and achieving a successful pregnancy.
Currently, available data do not support a routine check for cytokine or cytokine gene polymorphisms in women with RPL, and most obstetrical guidelines do not recommend testing cytokine levels [250]. It may be unnecessary to do cytokine testing in RPL patients who clearly have an endocrinologic or infectious etiology. Noteworthily, Th1/Th2 cell ratios have been utilized to diagnose and monitor women with RPL. Even though a variety of immunomodulatory treatments, including corticosteroids, paternal lymphocyte immunization, progesterone, intralipid infusion, TNF-α inhibitors, granulocyte colony-stimulating factor (G-CSF), low molecular weight heparin (LMWH), and intravenous immunoglobulin (IVIg) therapy, have been utilized in the treatment of RPL women, their efficacy is still the subject of contention [262,263,264]. Hence, not a single gene or a cytokine but a panel of cytokines and cytokine genes is likely to be necessary to assess women with RPL, considering the complexity of the immune network and the multifactorial nature of treatment responses. In women with RPL and dysregulated cytokine profiles, local or systemic administration of cytokines or their antagonists may be a potential therapeutic approach for maintaining immunological balance and improving maternal and fetal outcomes. Well-designed large-scale clinical trials are needed in the future.

Author Contributions

X.Y., Y.T. and L.Z. contributed to search the articles. X.Y., Y.T. and L.Z. wrote the manuscript. T.L. and J.K.-K. revised the manuscript. 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. El Hachem, H.; Crepaux, V.; May-Panloup, P.; Descamps, P.; Legendre, G.; Bouet, P.E. Recurrent pregnancy loss: Current perspectives. Int. J. Women’s Health 2017, 9, 331–345. [Google Scholar] [CrossRef] [Green Version]
  2. Youssef, A.; Vermeulen, N.; Lashley, E.; Goddijn, M.; van der Hoorn, M.L.P. Comparison and appraisal of (inter)national recurrent pregnancy loss guidelines. Reprod. Biomed. Online 2019, 39, 497–503. [Google Scholar] [CrossRef] [Green Version]
  3. Bagkou Dimakou, D.; Lissauer, D.; Tamblyn, J.; Coomarasamy, A.; Richter, A. Understanding human immunity in idiopathic recurrent pregnancy loss. Eur. J. Obstet. Gynecol. Reprod. Biol. 2022, 270, 17–29. [Google Scholar] [CrossRef]
  4. Dambaeva, S.V.; Lee, D.H.; Sung, N.; Chen, C.Y.; Bao, S.; Gilman-Sachs, A.; Kwak-Kim, J.; Beaman, K.D. Recurrent Pregnancy Loss in Women with Killer Cell Immunoglobulin-Like Receptor KIR2DS1 is Associated with an Increased HLA-C2 Allelic Frequency. Am. J. Reprod. Immunol. 2016, 75, 94–103. [Google Scholar] [CrossRef]
  5. Ewington, L.J.; Tewary, S.; Brosens, J.J. New insights into the mechanisms underlying recurrent pregnancy loss. J. Obstet. Gynaecol. Res. 2019, 45, 258–265. [Google Scholar] [CrossRef]
  6. Nair, R.R.; Verma, P.; Singh, K. Immune-endocrine crosstalk during pregnancy. Gen. Comp. Endocrinol. 2017, 242, 18–23. [Google Scholar] [CrossRef]
  7. Saini, V.; Arora, S.; Yadav, A.; Bhattacharjee, J. Cytokines in recurrent pregnancy loss. Clin. Chim. Acta 2011, 412, 702–708. [Google Scholar] [CrossRef]
  8. Motedayyen, H.; Rezaei, A.; Zarnani, A.H.; Tajik, N. Human amniotic epithelial cells inhibit activation and pro-inflammatory cytokines production of naive CD4+ T cells from women with unexplained recurrent spontaneous abortion. Reprod. Biol. 2018, 18, 182–188. [Google Scholar] [CrossRef]
  9. Zhu, L.Y.; Chen, X.; Xu, Z.Z.; Xu, L.; Mao, T.; Zhang, H. Changes and clinical significance of peripheral blood helper T lymphocyte and natural killer (NK) cells in unexplained recurrent spontaneous abortion (URSA) patients after abortion and successful pregnancy. Clin. Exp. Obstet. Gynecol. 2015, 42, 62–66. [Google Scholar] [CrossRef]
  10. Fu, B.; Li, X.; Sun, R.; Tong, X.; Ling, B.; Tian, Z.; Wei, H. Natural killer cells promote immune tolerance by regulating inflammatory TH17 cells at the human maternal-fetal interface. Proc. Natl. Acad. Sci. USA 2013, 110, E231–E240. [Google Scholar] [CrossRef]
  11. Saito, S.; Nakashima, A.; Shima, T.; Ito, M. Th1/Th2/Th17 and regulatory T-cell paradigm in pregnancy. Am. J. Reprod. Immunol. 2010, 63, 601–610. [Google Scholar] [CrossRef] [PubMed]
  12. Lash, G.E.; Ernerudh, J. Decidual cytokines and pregnancy complications: Focus on spontaneous miscarriage. J. Reprod. Immunol. 2015, 108, 83–89. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Zhu, L.; Chen, H.; Liu, M.; Yuan, Y.; Wang, Z.; Chen, Y.; Wei, J.; Su, F.; Zhang, J. Treg/Th17 Cell Imbalance and IL-6 Profile in Patients With Unexplained Recurrent Spontaneous Abortion. Reprod. Sci. 2017, 24, 882–890. [Google Scholar] [CrossRef] [PubMed]
  14. Lee, S.K.; Kim, J.Y.; Lee, M.; Gilman-Sachs, A.; Kwak-Kim, J. Th17 and regulatory T cells in women with recurrent pregnancy loss. Am. J. Reprod. Immunol. 2012, 67, 311–318. [Google Scholar] [CrossRef]
  15. Keller, C.C.; Eikmans, M.; van der Hoorn, M.P.; Lashley, L. Recurrent miscarriages and the association with regulatory T cells; A systematic review. J. Reprod. Immunol. 2020, 139, 103105. [Google Scholar] [CrossRef]
  16. Vivier, E.; Tomasello, E.; Baratin, M.; Walzer, T.; Ugolini, S. Functions of natural killer cells. Nat. Immunol. 2008, 9, 503–510. [Google Scholar] [CrossRef]
  17. Vacca, P.; Moretta, L.; Moretta, A.; Mingari, M.C. Origin, phenotype and function of human natural killer cells in pregnancy. Trends Immunol. 2011, 32, 517–523. [Google Scholar] [CrossRef]
  18. Chazara, O.; Xiong, S.; Moffett, A. Maternal KIR and fetal HLA-C: A fine balance. J. Leukoc. Biol. 2011, 90, 703–716. [Google Scholar] [CrossRef]
  19. Saito, S.; Nakashima, A.; Myojo-Higuma, S.; Shiozaki, A. The balance between cytotoxic NK cells and regulatory NK cells in human pregnancy. J. Reprod. Immunol. 2008, 77, 14–22. [Google Scholar] [CrossRef]
  20. Fu, Y.Y.; Ren, C.E.; Qiao, P.Y.; Meng, Y.H. Uterine natural killer cells and recurrent spontaneous abortion. Am. J. Reprod. Immunol. 2021, 86, e13433. [Google Scholar] [CrossRef]
  21. Maldonado, R.A.; von Andrian, U.H. How tolerogenic dendritic cells induce regulatory T cells. Adv. Immunol. 2010, 108, 111–165. [Google Scholar] [CrossRef] [Green Version]
  22. Salamone, G.; Fraccaroli, L.; Gori, S.; Grasso, E.; Paparini, D.; Geffner, J.; Pérez Leirós, C.; Ramhorst, R. Trophoblast cells induce a tolerogenic profile in dendritic cells. Hum. Reprod. 2012, 27, 2598–2606. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Wilczyński, J.R. Cancer and pregnancy share similar mechanisms of immunological escape. Chemotherapy 2006, 52, 107–110. [Google Scholar] [CrossRef]
  24. Bansal, A.S. Joining the immunological dots in recurrent miscarriage. Am. J. Reprod. Immunol. 2010, 64, 307–315. [Google Scholar] [CrossRef]
  25. Chen, Q.; Carroll, H.P.; Gadina, M. The newest interleukins: Recent additions to the ever-growing cytokine family. Vitam. Horm. 2006, 74, 207–228. [Google Scholar] [CrossRef]
  26. Ali, S.; Majid, S.; Ali, M.N.; Taing, S.; Rehman, M.U.; Arafah, A. Cytokine imbalance at materno-embryonic interface as a potential immune mechanism for recurrent pregnancy loss. Int. Immunopharmacol. 2021, 90, 107118. [Google Scholar] [CrossRef]
  27. Vignali, D. How many mechanisms do regulatory T cells need? Eur. J. Immunol. 2008, 38, 908–911. [Google Scholar] [CrossRef]
  28. Murphy, S.P.; Tayade, C.; Ashkar, A.A.; Hatta, K.; Zhang, J.; Croy, B.A. Interferon gamma in successful pregnancies. Biol. Reprod. 2009, 80, 848–859. [Google Scholar] [CrossRef] [Green Version]
  29. Lash, G.E.; Schiessl, B.; Kirkley, M.; Innes, B.A.; Cooper, A.; Searle, R.F.; Robson, S.C.; Bulmer, J.N. Expression of angiogenic growth factors by uterine natural killer cells during early pregnancy. J. Leukoc. Biol. 2006, 80, 572–580. [Google Scholar] [CrossRef] [Green Version]
  30. 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]
  31. Löb, S.; Amann, N.; Kuhn, C.; Schmoeckel, E.; Wöckel, A.; Zati Zehni, A.; Kaltofen, T.; Keckstein, S.; Mumm, J.N.; Meister, S.; et al. Interleukin-1 beta is significantly upregulated in the decidua of spontaneous and recurrent miscarriage placentas. J. Reprod. Immunol. 2021, 144, 103283. [Google Scholar] [CrossRef] [PubMed]
  32. Wu, L.; Li, J.; Xu, H.L.; Xu, B.; Tong, X.H.; Kwak-Kim, J.; Liu, Y.S. IL-7/IL-7R signaling pathway might play a role in recurrent pregnancy losses by increasing inflammatory Th17 cells and decreasing Treg cells. Am. J. Reprod. Immunol. 2016, 76, 454–464. [Google Scholar] [CrossRef] [PubMed]
  33. Hill, J.A.; Polgar, K.; Anderson, D.J. T-helper 1-type immunity to trophoblast in women with recurrent spontaneous abortion. JAMA 1995, 273, 1933–1936. [Google Scholar] [CrossRef] [PubMed]
  34. Peng, Y.; Yin, S.; Wang, M. Significance of the ratio interferon-γ/interleukin-4 in early diagnosis and immune mechanism of unexplained recurrent spontaneous abortion. Int. J. Gynaecol. Obstet. 2021, 154, 39–43. [Google Scholar] [CrossRef]
  35. Bates, M.D.; Quenby, S.; Takakuwa, K.; Johnson, P.M.; Vince, G.S. Aberrant cytokine production by peripheral blood mononuclear cells in recurrent pregnancy loss? Hum. Reprod. 2002, 17, 2439–2444. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Raghupathy, R.; Makhseed, M.; Azizieh, F.; Hassan, N.; Al-Azemi, M.; Al-Shamali, E. Maternal Th1- and Th2-type reactivity to placental antigens in normal human pregnancy and unexplained recurrent spontaneous abortions. Cell. Immunol. 1999, 196, 122–130. [Google Scholar] [CrossRef]
  37. Raghupathy, R.; Makhseed, M.; Azizieh, F.; Omu, A.; Gupta, M.; Farhat, R. Cytokine production by maternal lymphocytes during normal human pregnancy and in unexplained recurrent spontaneous abortion. Hum. Reprod. 2000, 15, 713–718. [Google Scholar] [CrossRef] [Green Version]
  38. Makhseed, M.; Raghupathy, R.; Azizieh, F.; Al-Azemi, M.M.; Hassan, N.A.; Bandar, A. Mitogen-induced cytokine responses of maternal peripheral blood lymphocytes indicate a differential Th-type bias in normal pregnancy and pregnancy failure. Am. J. Reprod. Immunol. 1999, 42, 273–281. [Google Scholar] [CrossRef]
  39. Marzi, M.; Vigano, A.; Trabattoni, D.; Villa, M.L.; Salvaggio, A.; Clerici, E.; Clerici, M. Characterization of type 1 and type 2 cytokine production profile in physiologic and pathologic human pregnancy. Clin. Exp. Immunol. 1996, 106, 127–133. [Google Scholar] [CrossRef]
  40. Tranchot-Diallo, J.; Gras, G.; Parnet-Mathieu, F.; Benveniste, O.; Marcé, D.; Roques, P.; Milliez, J.; Chaouat, G.; Dormont, D. Modulations of cytokine expression in pregnant women. Am. J. Reprod. Immunol. 1997, 37, 215–226. [Google Scholar] [CrossRef]
  41. Fallon, P.G.; Jolin, H.E.; Smith, P.; Emson, C.L.; Townsend, M.J.; Fallon, R.; Smith, P.; McKenzie, A.N. IL-4 induces characteristic Th2 responses even in the combined absence of IL-5, IL-9, and IL-13. Immunity 2002, 17, 7–17. [Google Scholar] [CrossRef] [Green Version]
  42. You, Y.; Stelzl, P.; Joseph, D.N.; Aldo, P.B.; Maxwell, A.J.; Dekel, N.; Liao, A.; Whirledge, S.; Mor, G. TNF-α Regulated Endometrial Stroma Secretome Promotes Trophoblast Invasion. Front. Immunol. 2021, 12, 737401. [Google Scholar] [CrossRef]
  43. Chung, I.B.; Yelian, F.D.; Zaher, F.M.; Gonik, B.; Evans, M.I.; Diamond, M.P.; Svinarich, D.M. Expression and regulation of vascular endothelial growth factor in a first trimester trophoblast cell line. Placenta 2000, 21, 320–324. [Google Scholar] [CrossRef]
  44. Lio, D.; Candore, G.; Colombo, A.; Colonna Romano, G.; Gervasi, F.; Marino, V.; Scola, L.; Caruso, C. A genetically determined high setting of TNF-alpha influences immunologic parameters of HLA-B8,DR3 positive subjects: Implications for autoimmunity. Hum. Immunol. 2001, 62, 705–713. [Google Scholar] [CrossRef]
  45. Ng, S.C.; Gilman-Sachs, A.; Thaker, P.; Beaman, K.D.; Beer, A.E.; Kwak-Kim, J. Expression of intracellular Th1 and Th2 cytokines in women with recurrent spontaneous abortion, implantation failures after IVF/ET or normal pregnancy. Am. J. Reprod. Immunol. 2002, 48, 77–86. [Google Scholar] [CrossRef]
  46. Elenkov, I.J.; Chrousos, G.P.; Wilder, R.L. Neuroendocrine regulation of IL-12 and TNF-alpha/IL-10 balance. Clinical implications. Ann. N. Y. Acad. Sci. 2000, 917, 94–105. [Google Scholar] [CrossRef]
  47. Dominguez, F.; Gadea, B.; Mercader, A.; Esteban, F.J.; Pellicer, A.; Simón, C. Embryologic outcome and secretome profile of implanted blastocysts obtained after coculture in human endometrial epithelial cells versus the sequential system. Fertil. Steril. 2010, 93, 774–782.e1. [Google Scholar] [CrossRef]
  48. Saifi, B.; Rezaee, S.A.; Tajik, N.; Ahmadpour, M.E.; Ashrafi, M.; Vakili, R.; SoleimaniAsl, S.; Aflatoonian, R.; Mehdizadeh, M. Th17 cells and related cytokines in unexplained recurrent spontaneous miscarriage at the implantation window. Reprod. Biomed. Online 2014, 29, 481–489. [Google Scholar] [CrossRef] [Green Version]
  49. Salama, K.M.; Alloush, M.K.; Al Hussini, R.M. Are the cytokines TNF alpha and IL 1Beta early predictors of embryo implantation? Cross sectional study. J. Reprod. Immunol. 2020, 137, 102618. [Google Scholar] [CrossRef]
  50. Guo, F.; Si, C.; Zhou, M.; Wang, J.; Zhang, D.; Leung, P.C.K.; Xu, B.; Zhang, A. Decreased PECAM1-mediated TGF-β1 expression in the mid-secretory endometrium in women with recurrent implantation failure. Hum. Reprod. 2018, 33, 832–843. [Google Scholar] [CrossRef]
  51. Bahrami-Asl, Z.; Hajipour, H.; Rastgar Rezaei, Y.; Novinbahador, T.; Latifi, Z.; Nejabati, H.R.; Farzadi, L.; Fattahi, A.; Nouri, M.; Dominguez, F. Cytokines in embryonic secretome as potential markers for embryo selection. Am. J. Reprod. Immunol. 2021, 85, e13385. [Google Scholar] [CrossRef]
  52. Hirota, Y.; Osuga, Y.; Hasegawa, A.; Kodama, A.; Tajima, T.; Hamasaki, K.; Koga, K.; Yoshino, O.; Hirata, T.; Harada, M.; et al. Interleukin (IL)-1beta stimulates migration and survival of first-trimester villous cytotrophoblast cells through endometrial epithelial cell-derived IL-8. Endocrinology 2009, 150, 350–356. [Google Scholar] [CrossRef] [Green Version]
  53. Wu, M.Y.; Chen, H.F.; Chen, S.U.; Chao, K.H.; Yang, Y.S.; Ho, H.N. Increase in the production of interleukin-10 early after implantation is related to the success of pregnancy. Am. J. Reprod. Immunol. 2001, 46, 386–392. [Google Scholar] [CrossRef]
  54. Dimitriadis, E.; Robb, L.; Liu, Y.X.; Enders, A.C.; Martin, H.; Stoikos, C.; Wallace, E.; Salamonsen, L.A. IL-11 and IL-11Ralpha immunolocalisation at primate implantation sites supports a role for IL-11 in placentation and fetal development. Reprod. Biol. Endocrinol. 2003, 1, 34. [Google Scholar] [CrossRef]
  55. Logiodice, F.; Lombardelli, L.; Kullolli, O.; Haller, H.; Maggi, E.; Rukavina, D.; Piccinni, M.P. Decidual Interleukin-22-Producing CD4+ T Cells (Th17/Th0/IL-22+ and Th17/Th2/IL-22+, Th2/IL-22+, Th0/IL-22+), Which Also Produce IL-4, Are Involved in the Success of Pregnancy. Int. J. Mol. Sci. 2019, 20, 428. [Google Scholar] [CrossRef] [Green Version]
  56. Salker, M.S.; Nautiyal, J.; Steel, J.H.; Webster, Z.; Sućurović, S.; Nicou, M.; Singh, Y.; Lucas, E.S.; Murakami, K.; Chan, Y.W.; et al. Disordered IL-33/ST2 activation in decidualizing stromal cells prolongs uterine receptivity in women with recurrent pregnancy loss. PLoS ONE 2012, 7, e52252. [Google Scholar] [CrossRef] [Green Version]
  57. Makhseed, M.; Raghupathy, R.; Azizieh, F.; Farhat, R.; Hassan, N.; Bandar, A. Circulating cytokines and CD30 in normal human pregnancy and recurrent spontaneous abortions. Hum. Reprod. 2000, 15, 2011–2017. [Google Scholar] [CrossRef] [Green Version]
  58. Arslan, E.; Colakoğlu, M.; Celik, C.; Gezginç, K.; Acar, A.; Capar, M.; Aköz, M.; Akyürek, C. Serum TNF-alpha, IL-6, lupus anticoagulant and anticardiolipin antibody in women with and without a past history of recurrent miscarriage. Arch. Gynecol. Obstet. 2004, 270, 227–229. [Google Scholar] [CrossRef]
  59. Piosik, Z.M.; Goegebeur, Y.; Klitkou, L.; Steffensen, R.; Christiansen, O.B. Plasma TNF-α levels are higher in early pregnancy in patients with secondary compared with primary recurrent miscarriage. Am. J. Reprod. Immunol. 2013, 70, 347–358. [Google Scholar] [CrossRef]
  60. Garcia-Lloret, M.I.; Winkler-Lowen, B.; Guilbert, L.J. Monocytes adhering by LFA-1 to placental syncytiotrophoblasts induce local apoptosis via release of TNF-alpha. A model for hematogenous initiation of placental inflammations. J. Leukoc. Biol. 2000, 68, 903–908. [Google Scholar] [CrossRef]
  61. Fortunato, S.J.; Menon, R.; Lombardi, S.J. Support for an infection-induced apoptotic pathway in human fetal membranes. Am. J. Obstet. Gynecol. 2001, 184, 1392–1397; discussion 1397–1398. [Google Scholar] [CrossRef]
  62. Berman, J.; Girardi, G.; Salmon, J.E. TNF-alpha is a critical effector and a target for therapy in antiphospholipid antibody-induced pregnancy loss. J. Immunol. 2005, 174, 485–490. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Banerjee, P.; Jana, S.K.; Pasricha, P.; Ghosh, S.; Chakravarty, B.; Chaudhury, K. Proinflammatory cytokines induced altered expression of cyclooxygenase-2 gene results in unreceptive endometrium in women with idiopathic recurrent spontaneous miscarriage. Fertil. Steril. 2013, 99, 179–187.e2. [Google Scholar] [CrossRef] [PubMed]
  64. Hossein, H.; Mahroo, M.; Abbas, A.; Firouzeh, A.; Nadia, H. Cytokine production by peripheral blood mononuclear cells in recurrent miscarriage. Cytokine 2004, 28, 83–86. [Google Scholar] [CrossRef] [PubMed]
  65. Daher, S.; de Arruda Geraldes Denardi, K.; Blotta, M.H.; Mamoni, R.L.; Reck, A.P.; Camano, L.; Mattar, R. Cytokines in recurrent pregnancy loss. J. Reprod. Immunol. 2004, 62, 151–157. [Google Scholar] [CrossRef] [PubMed]
  66. Zidan, H.E.; Abdul-Maksoud, R.S.; Mowafy, H.E.; Elsayed, W.S.H. The association of IL-33 and Foxp3 gene polymorphisms with recurrent pregnancy loss in Egyptian women. Cytokine 2018, 108, 115–119. [Google Scholar] [CrossRef]
  67. Cai, J.; Li, M.; Huang, Q.; Fu, X.; Wu, H. Differences in Cytokine Expression and STAT3 Activation between Healthy Controls and Patients of Unexplained Recurrent Spontaneous Abortion (URSA) during Early Pregnancy. PLoS ONE 2016, 11, e0163252. [Google Scholar] [CrossRef] [Green Version]
  68. Roomandeh, N.; Saremi, A.; Arasteh, J.; Pak, F.; Mirmohammadkhani, M.; Kokhaei, P.; Zare, A. Comparing Serum Levels of Th17 and Treg Cytokines in Women with Unexplained Recurrent Spontaneous Abortion and Fertile Women. Iran. J. Immunol. 2018, 15, 59–67. [Google Scholar]
  69. Wang, W.J.; Hao, C.F.; Yi, L.; Yin, G.J.; Bao, S.H.; Qiu, L.H.; Lin, Q.D. Increased prevalence of T helper 17 (Th17) cells in peripheral blood and decidua in unexplained recurrent spontaneous abortion patients. J. Reprod. Immunol. 2010, 84, 164–170. [Google Scholar] [CrossRef]
  70. Ozkan, Z.S.; Deveci, D.; Simsek, M.; Ilhan, F.; Risvanli, A.; Sapmaz, E. What is the impact of SOCS3, IL-35 and IL17 in immune pathogenesis of recurrent pregnancy loss? J. Matern. Fetal Neonatal Med. 2015, 28, 324–328. [Google Scholar] [CrossRef]
  71. Yue, C.Y.; Zhang, B.; Ying, C.M. Elevated Serum Level of IL-35 Associated with the Maintenance of Maternal-Fetal Immune Tolerance in Normal Pregnancy. PLoS ONE 2015, 10, e0128219. [Google Scholar] [CrossRef] [PubMed]
  72. Yeaman, G.R.; Collins, J.E.; Currie, J.K.; Guyre, P.M.; Wira, C.R.; Fanger, M.W. IFN-gamma is produced by polymorphonuclear neutrophils in human uterine endometrium and by cultured peripheral blood polymorphonuclear neutrophils. J. Immunol. 1998, 160, 5145–5153. [Google Scholar] [PubMed]
  73. Nasu, K.; Matsui, N.; Narahara, H.; Tanaka, Y.; Miyakawa, I. Effects of interferon-gamma on cytokine production by endometrial stromal cells. Hum. Reprod. 1998, 13, 2598–2601. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Calleja-Agius, J.; Jauniaux, E.; Pizzey, A.R.; Muttukrishna, S. Investigation of systemic inflammatory response in first trimester pregnancy failure. Hum. Reprod. 2012, 27, 349–357. [Google Scholar] [CrossRef] [Green Version]
  75. Palfi, M.; Jablonowska, B.; Matthiesen, L.; Ernerudh, J. Circulating interferon-gamma- and interleukin-4-secreting cells in recurrent spontaneous abortions. Am. J. Reprod. Immunol. 1999, 41, 257–263. [Google Scholar] [CrossRef]
  76. Robertson, S.A.; Seamark, R.F.; Guilbert, L.J.; Wegmann, T.G. The role of cytokines in gestation. Crit. Rev. Immunol. 1994, 14, 239–292. [Google Scholar] [CrossRef]
  77. Fontana, V.; Choren, V.; Vauthay, L.; Calvo, J.C.; Calvo, L.; Cameo, M. Exogenous interferon-gamma alters murine inner cell mass and trophoblast development. Effect on the expression of ErbB1, ErbB4 and heparan sulfate proteoglycan (perlecan). Reproduction 2004, 128, 717–725. [Google Scholar] [CrossRef] [Green Version]
  78. Matsumura, T.; Suzuki, T.; Aizawa, K.; Sawaki, D.; Munemasa, Y.; Ishida, J.; Nagai, R. Regulation of transforming growth factor-beta-dependent cyclooxygenase-2 expression in fibroblasts. J. Biol. Chem. 2009, 284, 35861–35871. [Google Scholar] [CrossRef] [Green Version]
  79. Swain, S.L.; Huston, G.; Tonkonogy, S.; Weinberg, A. Transforming growth factor-beta and IL-4 cause helper T cell precursors to develop into distinct effector helper cells that differ in lymphokine secretion pattern and cell surface phenotype. J. Immunol. 1991, 147, 2991–3000. [Google Scholar]
  80. Freis, A.; Schlegel, J.; Daniel, V.; Jauckus, J.; Strowitzki, T.; Germeyer, A. Cytokines in relation to hCG are significantly altered in asymptomatic women with miscarriage—A pilot study. Reprod. Biol. Endocrinol. 2018, 16, 93. [Google Scholar] [CrossRef] [Green Version]
  81. Ogasawara, M.S.; Aoki, K.; Aoyama, T.; Katano, K.; Iinuma, Y.; Ozaki, Y.; Suzumori, K. Elevation of transforming growth factor-beta1 is associated with recurrent miscarriage. J. Clin. Immunol. 2000, 20, 453–457. [Google Scholar] [CrossRef]
  82. Fang, L.; Yan, Y.; Gao, Y.; Wu, Z.; Wang, Z.; Yang, S.; Cheng, J.C.; Sun, Y.P. TGF-β1 inhibits human trophoblast cell invasion by upregulating kisspeptin expression through ERK1/2 but not SMAD signaling pathway. Reprod. Biol. Endocrinol. 2022, 20, 22. [Google Scholar] [CrossRef] [PubMed]
  83. Hefler, L.A.; Tempfer, C.B.; Unfried, G.; Schneeberger, C.; Lessl, K.; Nagele, F.; Huber, J.C. A polymorphism of the interleukin-1beta gene and idiopathic recurrent miscarriage. Fertil. Steril. 2001, 76, 377–379. [Google Scholar] [CrossRef] [PubMed]
  84. Nilkaeo, A.; Bhuvanath, S. Interleukin-1 modulation of human placental trophoblast proliferation. Mediat. Inflamm. 2006, 2006, 79359. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. 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] [PubMed]
  86. Furcron, A.E.; Romero, R.; Mial, T.N.; Balancio, A.; Panaitescu, B.; Hassan, S.S.; Sahi, A.; Nord, C.; Gomez-Lopez, N. Human Chorionic Gonadotropin Has Anti-Inflammatory Effects at the Maternal-Fetal Interface and Prevents Endotoxin-Induced Preterm Birth, but Causes Dystocia and Fetal Compromise in Mice. Biol. Reprod. 2016, 94, 136. [Google Scholar] [CrossRef] [Green Version]
  87. Gao, R.; Kong, L.; Qing, P.; Cheng, K.; Chen, H.; Zhang, S.; Hu, X.; Hu, Z.; Yu, F.; Qin, L. Interleukin-1β as clinically predictive risk marker for recurrent pregnancy loss in women positive for antinuclear antibody. Int. J. Clin. Pract. 2021, 75, e14887. [Google Scholar] [CrossRef]
  88. Spandorfer, S.D.; Neuer, A.; Liu, H.C.; Rosenwaks, Z.; Witkin, S.S. Involvement of interleukin-1 and the interleukin-1 receptor antagonist in in vitro embryo development among women undergoing in vitro fertilization-embryo transfer. J. Assist. Reprod. Genet. 2003, 20, 502–505. [Google Scholar] [CrossRef] [Green Version]
  89. Jackson, S.P. Arterial thrombosis--insidious, unpredictable and deadly. Nat. Med. 2011, 17, 1423–1436. [Google Scholar] [CrossRef]
  90. Haraldsen, G.; Balogh, J.; Pollheimer, J.; Sponheim, J.; Küchler, A.M. Interleukin-33—Cytokine of dual function or novel alarmin? Trends Immunol. 2009, 30, 227–233. [Google Scholar] [CrossRef]
  91. Smithgall, M.D.; Comeau, M.R.; Yoon, B.R.; Kaufman, D.; Armitage, R.; Smith, D.E. IL-33 amplifies both Th1- and Th2-type responses through its activity on human basophils, allergen-reactive Th2 cells, iNKT and NK cells. Int. Immunol. 2008, 20, 1019–1030. [Google Scholar] [CrossRef] [PubMed]
  92. Xu, D.; Jiang, H.R.; Kewin, P.; Li, Y.; Mu, R.; Fraser, A.R.; Pitman, N.; Kurowska-Stolarska, M.; McKenzie, A.N.; McInnes, I.B.; et al. IL-33 exacerbates antigen-induced arthritis by activating mast cells. Proc. Natl. Acad. Sci. USA 2008, 105, 10913–10918. [Google Scholar] [CrossRef] [PubMed]
  93. Nakae, S.; Morita, H.; Ohno, T.; Arae, K.; Matsumoto, K.; Saito, H. Role of interleukin-33 in innate-type immune cells in allergy. Allergol. Int. 2013, 62, 13–20. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Bourgeois, E.; Van, L.P.; Samson, M.; Diem, S.; Barra, A.; Roga, S.; Gombert, J.M.; Schneider, E.; Dy, M.; Gourdy, P.; et al. The pro-Th2 cytokine IL-33 directly interacts with invariant NKT and NK cells to induce IFN-gamma production. Eur. J. Immunol. 2009, 39, 1046–1055. [Google Scholar] [CrossRef] [PubMed]
  95. Gajardo Carrasco, T.; Morales, R.A.; Pérez, F.; Terraza, C.; Yáñez, L.; Campos-Mora, M.; Pino-Lagos, K. Alarmin’ Immunologists: IL-33 as a Putative Target for Modulating T Cell-Dependent Responses. Front. Immunol. 2015, 6, 232. [Google Scholar] [CrossRef] [Green Version]
  96. Yue, J.; Tong, Y.; Xie, L.; Ma, T.; Yang, J. Genetic variant in IL-33 is associated with idiopathic recurrent miscarriage in Chinese Han population. Sci. Rep. 2016, 6, 23806. [Google Scholar] [CrossRef] [Green Version]
  97. Kaitu’u-Lino, T.J.; Tuohey, L.; Tong, S. Maternal serum interleukin-33 and soluble ST2 across early pregnancy, and their association with miscarriage. J. Reprod. Immunol. 2012, 95, 46–49. [Google Scholar] [CrossRef]
  98. Hu, W.T.; Li, M.Q.; Liu, W.; Jin, L.P.; Li, D.J.; Zhu, X.Y. IL-33 enhances proliferation and invasiveness of decidual stromal cells by up-regulation of CCL2/CCR2 via NF-κB and ERK1/2 signaling. Mol. Hum. Reprod. 2014, 20, 358–372. [Google Scholar] [CrossRef] [Green Version]
  99. Prins, J.R.; Gomez-Lopez, N.; Robertson, S.A. Interleukin-6 in pregnancy and gestational disorders. J. Reprod. Immunol. 2012, 95, 1–14. [Google Scholar] [CrossRef]
  100. Arntzen, K.J.; Lien, E.; Austgulen, R. Maternal serum levels of interleukin-6 and clinical characteristics of normal delivery at term. Acta Obstet. Gynecol. Scand. 1997, 76, 55–60. [Google Scholar] [CrossRef]
  101. Oláh, K.S.; Vince, G.S.; Neilson, J.P.; Deniz, G.; Johnson, P.M. Interleukin-6, interferon-gamma, interleukin-8, and granulocyte-macrophage colony stimulating factor levels in human amniotic fluid at term. J. Reprod. Immunol. 1996, 32, 89–98. [Google Scholar] [CrossRef] [PubMed]
  102. Steinborn, A.; Günes, H.; Röddiger, S.; Halberstadt, E. Elevated placental cytokine release, a process associated with preterm labor in the absence of intrauterine infection. Obstet. Gynecol. 1996, 88, 534–539. [Google Scholar] [CrossRef] [PubMed]
  103. Rose-John, S. Interleukin-6 Family Cytokines. Cold Spring Harb. Perspect. Biol. 2018, 10, a028415. [Google Scholar] [CrossRef] [Green Version]
  104. Sabat, R.; Grütz, G.; Warszawska, K.; Kirsch, S.; Witte, E.; Wolk, K.; Geginat, J. Biology of interleukin-10. Cytokine Growth Factor Rev. 2010, 21, 331–344. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Wang, W.J.; Hao, C.F.; Lin, Q.D. Dysregulation of macrophage activation by decidual regulatory T cells in unexplained recurrent miscarriage patients. J. Reprod. Immunol. 2011, 92, 97–102. [Google Scholar] [CrossRef]
  106. Chaouat, G.; Assal Meliani, A.; Martal, J.; Raghupathy, R.; Elliott, J.F.; Mosmann, T.; Wegmann, T.G. IL-10 prevents naturally occurring fetal loss in the CBA x DBA/2 mating combination, and local defect in IL-10 production in this abortion-prone combination is corrected by in vivo injection of IFN-tau. J. Immunol. 1995, 154, 4261–4268. [Google Scholar] [CrossRef] [Green Version]
  107. Robertson, S.A.; Care, A.S.; Skinner, R.J. Interleukin 10 regulates inflammatory cytokine synthesis to protect against lipopolysaccharide-induced abortion and fetal growth restriction in mice. Biol. Reprod. 2007, 76, 738–748. [Google Scholar] [CrossRef] [Green Version]
  108. Jenkins, C.; Roberts, J.; Wilson, R.; MacLean, M.A.; Shilito, J.; Walker, J.J. Evidence of a T(H) 1 type response associated with recurrent miscarriage. Fertil. Steril. 2000, 73, 1206–1208. [Google Scholar] [CrossRef]
  109. Hasnain, S.Z.; Tauro, S.; Das, I.; Tong, H.; Chen, A.C.; Jeffery, P.L.; McDonald, V.; Florin, T.H.; McGuckin, M.A. IL-10 promotes production of intestinal mucus by suppressing protein misfolding and endoplasmic reticulum stress in goblet cells. Gastroenterology 2013, 144, 357–368.e9. [Google Scholar] [CrossRef]
  110. Shkoda, A.; Ruiz, P.A.; Daniel, H.; Kim, S.C.; Rogler, G.; Sartor, R.B.; Haller, D. Interleukin-10 blocked endoplasmic reticulum stress in intestinal epithelial cells: Impact on chronic inflammation. Gastroenterology 2007, 132, 190–207. [Google Scholar] [CrossRef]
  111. Yang, H.; Qiu, L.; Chen, G.; Ye, Z.; Lü, C.; Lin, Q. Proportional change of CD4+CD25+ regulatory T cells in decidua and peripheral blood in unexplained recurrent spontaneous abortion patients. Fertil. Steril. 2008, 89, 656–661. [Google Scholar] [CrossRef] [PubMed]
  112. Liu, Y.S.; Wu, L.; Tong, X.H.; Wu, L.M.; He, G.P.; Zhou, G.X.; Luo, L.H.; Luan, H.B. Study on the relationship between Th17 cells and unexplained recurrent spontaneous abortion. Am. J. Reprod. Immunol. 2011, 65, 503–511. [Google Scholar] [CrossRef] [PubMed]
  113. Lee, S.K.; Kim, J.Y.; Hur, S.E.; Kim, C.J.; Na, B.J.; Lee, M.; Gilman-Sachs, A.; Kwak-Kim, J. An imbalance in interleukin-17-producing T and Foxp3⁺ regulatory T cells in women with idiopathic recurrent pregnancy loss. Hum. Reprod. 2011, 26, 2964–2971. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Nakashima, A.; Ito, M.; Shima, T.; Bac, N.D.; Hidaka, T.; Saito, S. Accumulation of IL-17-positive cells in decidua of inevitable abortion cases. Am. J. Reprod. Immunol. 2010, 64, 4–11. [Google Scholar] [CrossRef]
  115. Piccinni, M.P. T cell tolerance towards the fetal allograft. J. Reprod. Immunol. 2010, 85, 71–75. [Google Scholar] [CrossRef]
  116. Arruvito, L.; Billordo, A.; Capucchio, M.; Prada, M.E.; Fainboim, L. IL-6 trans-signaling and the frequency of CD4+FOXP3+ cells in women with reproductive failure. J. Reprod. Immunol. 2009, 82, 158–165. [Google Scholar] [CrossRef]
  117. Pasare, C.; Medzhitov, R. Toll pathway-dependent blockade of CD4+CD25+ T cell-mediated suppression by dendritic cells. Science 2003, 299, 1033–1036. [Google Scholar] [CrossRef]
  118. Corthay, A. How do regulatory T cells work? Scand. J. Immunol. 2009, 70, 326–336. [Google Scholar] [CrossRef]
  119. Vilsmaier, T.; Amann, N.; Löb, S.; Schmoeckel, E.; Kuhn, C.; Zati Zehni, A.; Meister, S.; Beyer, S.; Kolben, T.M.; Becker, J.; et al. The decidual expression of Interleukin-7 is upregulated in early pregnancy loss. Am. J. Reprod. Immunol. 2021, 86, e13437. [Google Scholar] [CrossRef]
  120. Kwak, J.Y.; Beer, A.E.; Kim, S.H.; Mantouvalos, H.P. Immunopathology of the implantation site utilizing monoclonal antibodies to natural killer cells in women with recurrent pregnancy losses. Am. J. Reprod. Immunol. 1999, 41, 91–98. [Google Scholar] [CrossRef]
  121. Haas, J.; Korporal, M.; Schwarz, A.; Balint, B.; Wildemann, B. The interleukin-7 receptor α chain contributes to altered homeostasis of regulatory T cells in multiple sclerosis. Eur. J. Immunol. 2011, 41, 845–853. [Google Scholar] [CrossRef] [PubMed]
  122. Hartgring, S.A.; Bijlsma, J.W.; Lafeber, F.P.; van Roon, J.A. Interleukin-7 induced immunopathology in arthritis. Ann. Rheum. Dis. 2006, 65 (Suppl. 3), iii69–iii74. [Google Scholar] [CrossRef] [PubMed]
  123. Chen, H.; Wang, W.; Xie, H.; Xu, X.; Wu, J.; Jiang, Z.; Zhang, M.; Zhou, L.; Zheng, S. A pathogenic role of IL- 17 at the early stage of corneal allograft rejection. Transpl. Immunol. 2009, 21, 155–161. [Google Scholar] [CrossRef] [PubMed]
  124. Yuan, X.; Paez-Cortez, J.; Schmitt-Knosalla, I.; D’Addio, F.; Mfarrej, B.; Donnarumma, M.; Habicht, A.; Clarkson, M.R.; Iacomini, J.; Glimcher, L.H.; et al. A novel role of CD4 Th17 cells in mediating cardiac allograft rejection and vasculopathy. J. Exp. Med. 2008, 205, 3133–3144. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Wang, W.J.; Liu, F.J.; Qu, H.M.; Hao, C.F.; Qu, Q.L.; Xiong, W.; Bao, H.C.; Wang, X.R. Regulation of the expression of Th17 cells and regulatory T cells by IL-27 in patients with unexplained early recurrent miscarriage. J. Reprod. Immunol. 2013, 99, 39–45. [Google Scholar] [CrossRef] [PubMed]
  126. Wu, H.X.; Jin, L.P.; Xu, B.; Liang, S.S.; Li, D.J. Decidual stromal cells recruit Th17 cells into decidua to promote proliferation and invasion of human trophoblast cells by secreting IL-17. Cell. Mol. Immunol. 2014, 11, 253–262. [Google Scholar] [CrossRef] [Green Version]
  127. Kaminski, V.L.; Ellwanger, J.H.; Matte, M.C.C.; Savaris, R.F.; Vianna, P.; Chies, J.A.B. IL-17 blood levels increase in healthy pregnancy but not in spontaneous abortion. Mol. Biol. Rep. 2018, 45, 1565–1568. [Google Scholar] [CrossRef]
  128. Xu, W.M.; Xiao, Z.N.; Wang, X.B.; Huang, Y. IL-17 Induces Fetal Loss in a CBA/J×BALB/c Mouse Model, and an Anti-IL-17 Antibody Prevents Fetal Loss in a CBA/J×DBA/2 Mouse Model. Am. J. Reprod. Immunol. 2016, 75, 51–58. [Google Scholar] [CrossRef]
  129. Lindemans, C.A.; Calafiore, M.; Mertelsmann, A.M.; O’Connor, M.H.; Dudakov, J.A.; Jenq, R.R.; Velardi, E.; Young, L.F.; Smith, O.M.; Lawrence, G.; et al. Interleukin-22 promotes intestinal-stem-cell-mediated epithelial regeneration. Nature 2015, 528, 560–564. [Google Scholar] [CrossRef] [Green Version]
  130. McGee, H.M.; Schmidt, B.A.; Booth, C.J.; Yancopoulos, G.D.; Valenzuela, D.M.; Murphy, A.J.; Stevens, S.; Flavell, R.A.; Horsley, V. IL-22 promotes fibroblast-mediated wound repair in the skin. J. Investig. Dermatol. 2013, 133, 1321–1329. [Google Scholar] [CrossRef] [Green Version]
  131. Uz, Y.H.; Murk, W.; Yetkin, C.E.; Kayisli, U.A.; Arici, A. Expression and role of interleukin-23 in human endometrium throughout the menstrual cycle and early pregnancy. J. Reprod. Immunol. 2010, 87, 21–27. [Google Scholar] [CrossRef] [PubMed]
  132. Coulomb-L’Herminé, A.; Larousserie, F.; Pflanz, S.; Bardel, E.; Kastelein, R.A.; Devergne, O. Expression of interleukin-27 by human trophoblast cells. Placenta 2007, 28, 1133–1140. [Google Scholar] [CrossRef] [PubMed]
  133. Mas, A.E.; Petitbarat, M.; Dubanchet, S.; Fay, S.; Ledée, N.; Chaouat, G. Immune regulation at the interface during early steps of murine implantation: Involvement of two new cytokines of the IL-12 family (IL-23 and IL-27) and of TWEAK. Am. J. Reprod. Immunol. 2008, 59, 323–338. [Google Scholar] [CrossRef] [PubMed]
  134. Chaouat, G.; Petitbarat, M.; Bulla, R.; Dubanchet, S.; Valdivia, K.; Ledée, N.; Steffen, T.; Jensenius, J.C.; Tedesco, F. Early regulators in abortion and implications for a preeclampsia model. J. Reprod. Immunol. 2009, 82, 131–140. [Google Scholar] [CrossRef] [PubMed]
  135. Collison, L.W.; Workman, C.J.; Kuo, T.T.; Boyd, K.; Wang, Y.; Vignali, K.M.; Cross, R.; Sehy, D.; Blumberg, R.S.; Vignali, D.A. The inhibitory cytokine IL-35 contributes to regulatory T-cell function. Nature 2007, 450, 566–569. [Google Scholar] [CrossRef]
  136. Mao, H.; Gao, W.; Ma, C.; Sun, J.; Liu, J.; Shao, Q.; Song, B.; Qu, X. Human placental trophoblasts express the immunosuppressive cytokine IL-35. Hum. Immunol. 2013, 74, 872–877. [Google Scholar] [CrossRef]
  137. Qian, J.; Zhang, N.; Lin, J.; Wang, C.; Pan, X.; Chen, L.; Li, D.; Wang, L. Distinct pattern of Th17/Treg cells in pregnant women with a history of unexplained recurrent spontaneous abortion. Biosci. Trends 2018, 12, 157–167. [Google Scholar] [CrossRef] [Green Version]
  138. Yuan, J.; Li, J.; Huang, S.Y.; Sun, X. Characterization of the subsets of human NKT-like cells and the expression of Th1/Th2 cytokines in patients with unexplained recurrent spontaneous abortion. J. Reprod. Immunol. 2015, 110, 81–88. [Google Scholar] [CrossRef]
  139. Jasper, M.J.; Tremellen, K.P.; Robertson, S.A. Reduced expression of IL-6 and IL-1alpha mRNAs in secretory phase endometrium of women with recurrent miscarriage. J. Reprod. Immunol. 2007, 73, 74–84. [Google Scholar] [CrossRef]
  140. Linjawi, S.; Li, T.C.; Tuckerman, E.M.; Blakemore, A.I.; Laird, S.M. Expression of interleukin-11 receptor alpha and interleukin-11 protein in the endometrium of normal fertile women and women with recurrent miscarriage. J. Reprod. Immunol. 2004, 64, 145–155. [Google Scholar] [CrossRef]
  141. Löb, S.; Ochmann, B.; Ma, Z.; Vilsmaier, T.; Kuhn, C.; Schmoeckel, E.; Herbert, S.L.; Kolben, T.; Wöckel, A.; Mahner, S.; et al. The role of Interleukin-18 in recurrent early pregnancy loss. J. Reprod. Immunol. 2021, 148, 103432. [Google Scholar] [CrossRef] [PubMed]
  142. Tabibzadeh, S. Ubiquitous expression of TNF-alpha/cachectin immunoreactivity in human endometrium. Am. J. Reprod. Immunol. 1991, 26, 1–4. [Google Scholar] [CrossRef] [PubMed]
  143. Hunt, J.S.; Chen, H.L.; Hu, X.L.; Tabibzadeh, S. Tumor necrosis factor-alpha messenger ribonucleic acid and protein in human endometrium. Biol. Reprod. 1992, 47, 141–147. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Philippeaux, M.M.; Piguet, P.F. Expression of tumor necrosis factor-alpha and its mRNA in the endometrial mucosa during the menstrual cycle. Am. J. Pathol. 1993, 143, 480–486. [Google Scholar]
  145. Arck, P.C.; Troutt, A.B.; Clark, D.A. Soluble receptors neutralizing TNF-alpha and IL-1 block stress-triggered murine abortion. Am. J. Reprod. Immunol. 1997, 37, 262–266. [Google Scholar] [CrossRef] [PubMed]
  146. Otun, H.A.; Lash, G.E.; Innes, B.A.; Bulmer, J.N.; Naruse, K.; Hannon, T.; Searle, R.F.; Robson, S.C. Effect of tumour necrosis factor-α in combination with interferon-γ on first trimester extravillous trophoblast invasion. J. Reprod. Immunol. 2011, 88, 1–11. [Google Scholar] [CrossRef]
  147. Lim, K.J.; Odukoya, O.A.; Ajjan, R.A.; Li, T.C.; Weetman, A.P.; Cooke, I.D. The role of T-helper cytokines in human reproduction. Fertil. Steril. 2000, 73, 136–142. [Google Scholar] [CrossRef]
  148. Von Wolff, M.; Thaler, C.J.; Strowitzki, T.; Broome, J.; Stolz, W.; Tabibzadeh, S. Regulated expression of cytokines in human endometrium throughout the menstrual cycle: Dysregulation in habitual abortion. Mol. Hum. Reprod. 2000, 6, 627–634. [Google Scholar] [CrossRef]
  149. Lin, H.; Mosmann, T.R.; Guilbert, L.; Tuntipopipat, S.; Wegmann, T.G. Synthesis of T helper 2-type cytokines at the maternal-fetal interface. J. Immunol. 1993, 151, 4562–4573. [Google Scholar]
  150. Bennett, W.A.; Lagoo-Deenadayalan, S.; Whitworth, N.S.; Brackin, M.N.; Hale, E.; Cowan, B.D. Expression and production of interleukin-10 by human trophoblast: Relationship to pregnancy immunotolerance. Early Pregnancy 1997, 3, 190–198. [Google Scholar]
  151. Zhang, A.; Fu, J.; Ning, B.; Li, D.; Sun, N.; Wei, W.; Wei, J.; Ju, X. Tolerogenic dendritic cells generated with IL-10/TGFβ1 relieve immune thrombocytopenia in mice. Thromb. Res. 2013, 132, 63–68. [Google Scholar] [CrossRef] [PubMed]
  152. Schumacher, A.; Costa, S.D.; Zenclussen, A.C. Endocrine factors modulating immune responses in pregnancy. Front. Immunol. 2014, 5, 196. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Xu, L.; Qiu, T.; Wang, Y.; Chen, Y.; Cheng, W. Expression of C-type lectin receptors and Toll-like receptors in decidua of patients with unexplained recurrent spontaneous abortion. Reprod. Fertil. Dev. 2017, 29, 1613–1624. [Google Scholar] [CrossRef]
  154. Bao, S.H.; Wang, X.P.; De Lin, Q.; Wang, W.J.; Yin, G.J.; Qiu, L.H. Decidual CD4+CD25+CD127dim/− regulatory T cells in patients with unexplained recurrent spontaneous miscarriage. Eur. J. Obstet. Gynecol. Reprod. Biol. 2011, 155, 94–98. [Google Scholar] [CrossRef]
  155. Piccinni, M.P.; Beloni, L.; Livi, C.; Maggi, E.; Scarselli, G.; Romagnani, S. Defective production of both leukemia inhibitory factor and type 2 T-helper cytokines by decidual T cells in unexplained recurrent abortions. Nat. Med. 1998, 4, 1020–1024. [Google Scholar] [CrossRef] [PubMed]
  156. Thaxton, J.E.; Sharma, S. Interleukin-10: A multi-faceted agent of pregnancy. Am. J. Reprod. Immunol. 2010, 63, 482–491. [Google Scholar] [CrossRef] [Green Version]
  157. Ticconi, C.; Pietropolli, A.; Di Simone, N.; Piccione, E.; Fazleabas, A. Endometrial Immune Dysfunction in Recurrent Pregnancy Loss. Int. J. Mol. Sci. 2019, 20, 5332. [Google Scholar] [CrossRef] [Green Version]
  158. Liu, J.; Dong, P.; Jia, N.; Wen, X.; Luo, L.; Wang, S.; Li, J. The expression of intracellular cytokines of decidual natural killer cells in unexplained recurrent pregnancy loss. J. Matern. Fetal Neonatal Med. 2022, 35, 3209–3215. [Google Scholar] [CrossRef]
  159. Liu, J.; Hao, S.; Chen, X.; Zhao, H.; Du, L.; Ren, H.; Wang, C.; Mao, H. Human placental trophoblast cells contribute to maternal-fetal tolerance through expressing IL-35 and mediating iT(R)35 conversion. Nat. Commun. 2019, 10, 4601. [Google Scholar] [CrossRef] [Green Version]
  160. Marwood, M.; Visser, K.; Salamonsen, L.A.; Dimitriadis, E. Interleukin-11 and leukemia inhibitory factor regulate the adhesion of endometrial epithelial cells: Implications in fertility regulation. Endocrinology 2009, 150, 2915–2923. [Google Scholar] [CrossRef] [Green Version]
  161. Paiva, P.; Salamonsen, L.A.; Manuelpillai, U.; Walker, C.; Tapia, A.; Wallace, E.M.; Dimitriadis, E. Interleukin-11 promotes migration, but not proliferation, of human trophoblast cells, implying a role in placentation. Endocrinology 2007, 148, 5566–5572. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Karpovich, N.; Klemmt, P.; Hwang, J.H.; McVeigh, J.E.; Heath, J.K.; Barlow, D.H.; Mardon, H.J. The production of interleukin-11 and decidualization are compromised in endometrial stromal cells derived from patients with infertility. J. Clin. Endocrinol. Metab. 2005, 90, 1607–1612. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Robb, L.; Li, R.; Hartley, L.; Nandurkar, H.H.; Koentgen, F.; Begley, C.G. Infertility in female mice lacking the receptor for interleukin 11 is due to a defective uterine response to implantation. Nat. Med. 1998, 4, 303–308. [Google Scholar] [CrossRef] [PubMed]
  164. Bilinski, P.; Roopenian, D.; Gossler, A. Maternal IL-11Ralpha function is required for normal decidua and fetoplacental development in mice. Genes Dev. 1998, 12, 2234–2243. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Maconochie, N.; Doyle, P.; Prior, S.; Simmons, R. Risk factors for first trimester miscarriage--results from a UK-population-based case-control study. BJOG 2007, 114, 170–186. [Google Scholar] [CrossRef] [PubMed]
  166. Li, L.; Liu, L.; Kou, Z.; Huo, M.; An, J.; Zhang, X. GnRH agonist treatment regulates IL-6 and IL-11 expression in endometrial stromal cells for patients with HRT regiment in frozen embryo transfer cycles. Reprod. Biol. 2022, 22, 100608. [Google Scholar] [CrossRef]
  167. Giannubilo, S.R.; Landi, B.; Pozzi, V.; Sartini, D.; Cecati, M.; Stortoni, P.; Corradetti, A.; Saccucci, F.; Tranquilli, A.L.; Emanuelli, M. The involvement of inflammatory cytokines in the pathogenesis of recurrent miscarriage. Cytokine 2012, 58, 50–56. [Google Scholar] [CrossRef]
  168. Haider, S.; Lackner, A.I.; Dietrich, B.; Kunihs, V.; Haslinger, P.; Meinhardt, G.; Maxian, T.; Saleh, L.; Fiala, C.; Pollheimer, J.; et al. Transforming growth factor-β signaling governs the differentiation program of extravillous trophoblasts in the developing human placenta. Proc. Natl. Acad. Sci. USA 2022, 119, e2120667119. [Google Scholar] [CrossRef]
  169. Clark, D.A.; Chaouat, G.; Arck, P.C.; Mittruecker, H.W.; Levy, G.A. Cytokine-dependent abortion in CBA x DBA/2 mice is mediated by the procoagulant fgl2 prothrombinase [correction of prothombinase]. J. Immunol. 1998, 160, 545–549. [Google Scholar]
  170. Knackstedt, M.; Ding, J.W.; Arck, P.C.; Hertwig, K.; Coulam, C.B.; August, C.; Lea, R.; Dudenhausen, J.W.; Gorczynski, R.M.; Levy, G.A.; et al. Activation of the novel prothrombinase, fg12, as a basis for the pregnancy complications spontaneous abortion and pre-eclampsia. Am. J. Reprod. Immunol. 2001, 46, 196–210. [Google Scholar] [CrossRef]
  171. Drake, B.L.; Head, J.R. Murine trophoblast can be killed by lymphokine-activated killer cells. J. Immunol. 1989, 143, 9–14. [Google Scholar] [PubMed]
  172. Hoshino, T.; Winkler-Pickett, R.T.; Mason, A.T.; Ortaldo, J.R.; Young, H.A. IL-13 production by NK cells: IL-13-producing NK and T cells are present in vivo in the absence of IFN-gamma. J. Immunol. 1999, 162, 51–59. [Google Scholar] [PubMed]
  173. Loza, M.J.; Perussia, B. Final steps of natural killer cell maturation: A model for type 1-type 2 differentiation? Nat. Immunol. 2001, 2, 917–924. [Google Scholar] [CrossRef] [PubMed]
  174. Peritt, D.; Robertson, S.; Gri, G.; Showe, L.; Aste-Amezaga, M.; Trinchieri, G. Differentiation of human NK cells into NK1 and NK2 subsets. J. Immunol. 1998, 161, 5821–5824. [Google Scholar] [PubMed]
  175. Warren, H.S.; Kinnear, B.F.; Phillips, J.H.; Lanier, L.L. Production of IL-5 by human NK cells and regulation of IL-5 secretion by IL-4, IL-10, and IL-12. J. Immunol. 1995, 154, 5144–5152. [Google Scholar]
  176. Higuma-Myojo, S.; Sasaki, Y.; Miyazaki, S.; Sakai, M.; Siozaki, A.; Miwa, N.; Saito, S. Cytokine profile of natural killer cells in early human pregnancy. Am. J. Reprod. Immunol. 2005, 54, 21–29. [Google Scholar] [CrossRef]
  177. Veenstra van Nieuwenhoven, A.L.; Bouman, A.; Moes, H.; Heineman, M.J.; de Leij, L.F.; Santema, J.; Faas, M.M. Cytokine production in natural killer cells and lymphocytes in pregnant women compared with women in the follicular phase of the ovarian cycle. Fertil. Steril. 2002, 77, 1032–1037. [Google Scholar] [CrossRef]
  178. Borzychowski, A.M.; Croy, B.A.; Chan, W.L.; Redman, C.W.; Sargent, I.L. Changes in systemic type 1 and type 2 immunity in normal pregnancy and pre-eclampsia may be mediated by natural killer cells. Eur. J. Immunol. 2005, 35, 3054–3063. [Google Scholar] [CrossRef]
  179. Shakhar, K.; Ben-Eliyahu, S.; Loewenthal, R.; Rosenne, E.; Carp, H. Differences in number and activity of peripheral natural killer cells in primary versus secondary recurrent miscarriage. Fertil. Steril. 2003, 80, 368–375. [Google Scholar] [CrossRef]
  180. Aoki, K.; Kajiura, S.; Matsumoto, Y.; Ogasawara, M.; Okada, S.; Yagami, Y.; Gleicher, N. Preconceptional natural-killer-cell activity as a predictor of miscarriage. Lancet 1995, 345, 1340–1342. [Google Scholar] [CrossRef]
  181. Yamada, H.; Morikawa, M.; Kato, E.H.; Shimada, S.; Kobashi, G.; Minakami, H. Pre-conceptional natural killer cell activity and percentage as predictors of biochemical pregnancy and spontaneous abortion with normal chromosome karyotype. Am. J. Reprod. Immunol. 2003, 50, 351–354. [Google Scholar] [CrossRef] [PubMed]
  182. Souza, S.S.; Ferriani, R.A.; Santos, C.M.; Voltarelli, J.C. Immunological evaluation of patients with recurrent abortion. J. Reprod. Immunol. 2002, 56, 111–121. [Google Scholar] [CrossRef] [PubMed]
  183. King, K.; Smith, S.; Chapman, M.; Sacks, G. Detailed analysis of peripheral blood natural killer (NK) cells in women with recurrent miscarriage. Hum. Reprod. 2010, 25, 52–58. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Hosseini, S.; Zarnani, A.H.; Asgarian-Omran, H.; Vahedian-Dargahi, Z.; Eshraghian, M.R.; Akbarzadeh-Pasha, Z.; Arefi, S.; Jeddi-Tehrani, M.; Shokri, F. Comparative analysis of NK cell subsets in menstrual and peripheral blood of patients with unexplained recurrent spontaneous abortion and fertile subjects. J. Reprod. Immunol. 2014, 103, 9–17. [Google Scholar] [CrossRef]
  185. Karami, N.; Boroujerdnia, M.G.; Nikbakht, R.; Khodadadi, A. Enhancement of peripheral blood CD56(dim) cell and NK cell cytotoxicity in women with recurrent spontaneous abortion or in vitro fertilization failure. J. Reprod. Immunol. 2012, 95, 87–92. [Google Scholar] [CrossRef]
  186. El-Azzamy, H.; Dambaeva, S.V.; Katukurundage, D.; Salazar Garcia, M.D.; Skariah, A.; Hussein, Y.; Germain, A.; Fernandez, E.; Gilman-Sachs, A.; Beaman, K.D.; et al. Dysregulated uterine natural killer cells and vascular remodeling in women with recurrent pregnancy losses. Am. J. Reprod. Immunol. 2018, 80, e13024. [Google Scholar] [CrossRef]
  187. Dong, P.; Wen, X.; Liu, J.; Yan, C.Y.; Yuan, J.; Luo, L.R.; Hu, Q.F.; Li, J. Simultaneous detection of decidual Th1/Th2 and NK1/NK2 immunophenotyping in unknown recurrent miscarriage using 8-color flow cytometry with FSC/Vt extended strategy. Biosci. Rep. 2017, 37, BSR20170150. [Google Scholar] [CrossRef]
  188. Fukui, A.; Funamizu, A.; Fukuhara, R.; Shibahara, H. Expression of natural cytotoxicity receptors and cytokine production on endometrial natural killer cells in women with recurrent pregnancy loss or implantation failure, and the expression of natural cytotoxicity receptors on peripheral blood natural killer cells in pregnant women with a history of recurrent pregnancy loss. J. Obstet. Gynaecol. Res. 2017, 43, 1678–1686. [Google Scholar] [CrossRef] [Green Version]
  189. Fukui, A.; Kwak-Kim, J.; Ntrivalas, E.; Gilman-Sachs, A.; Lee, S.K.; Beaman, K. Intracellular cytokine expression of peripheral blood natural killer cell subsets in women with recurrent spontaneous abortions and implantation failures. Fertil. Steril. 2008, 89, 157–165. [Google Scholar] [CrossRef]
  190. Zhu, L.; Aly, M.; Wang, H.; Karakizlis, H.; Weimer, R.; Morath, C.; Kuon, R.J.; Toth, B.; Ekpoom, N.; Opelz, G.; et al. Increased natural killer cell subsets with inhibitory cytokines and inhibitory surface receptors in patients with recurrent miscarriage and decreased or normal subsets in kidney transplant recipients late post-transplant. Clin. Exp. Immunol. 2018, 193, 241–254. [Google Scholar] [CrossRef] [Green Version]
  191. Kamoi, M.; Fukui, A.; Kwak-Kim, J.; Fuchinoue, K.; Funamizu, A.; Chiba, H.; Yokota, M.; Fukuhara, R.; Mizunuma, H. NK22 Cells in the Uterine Mid-Secretory Endometrium and Peripheral Blood of Women with Recurrent Pregnancy Loss and Unexplained Infertility. Am. J. Reprod. Immunol. 2015, 73, 557–567. [Google Scholar] [CrossRef] [PubMed]
  192. 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] [PubMed] [Green Version]
  193. Zourbas, S.; Dubanchet, S.; Martal, J.; Chaouat, G. Localization of pro-inflammatory (IL-12, IL-15) and anti-inflammatory (IL-11, IL-13) cytokines at the foetomaternal interface during murine pregnancy. Clin. Exp. Immunol. 2001, 126, 519–528. [Google Scholar] [CrossRef] [PubMed]
  194. Verma, S.; Hiby, S.E.; Loke, Y.W.; King, A. Human decidual natural killer cells express the receptor for and respond to the cytokine interleukin 15. Biol. Reprod. 2000, 62, 959–968. [Google Scholar] [CrossRef] [Green Version]
  195. Ashkar, A.A.; Black, G.P.; Wei, Q.; He, H.; Liang, L.; Head, J.R.; Croy, B.A. Assessment of requirements for IL-15 and IFN regulatory factors in uterine NK cell differentiation and function during pregnancy. J. Immunol. 2003, 171, 2937–2944. [Google Scholar] [CrossRef] [Green Version]
  196. Wilkens, J.; Male, V.; Ghazal, P.; Forster, T.; Gibson, D.A.; Williams, A.R.; Brito-Mutunayagam, S.L.; Craigon, M.; Lourenco, P.; Cameron, I.T.; et al. Uterine NK cells regulate endometrial bleeding in women and are suppressed by the progesterone receptor modulator asoprisnil. J. Immunol. 2013, 191, 2226–2235. [Google Scholar] [CrossRef] [Green Version]
  197. Barber, E.M.; Pollard, J.W. The uterine NK cell population requires IL-15 but these cells are not required for pregnancy nor the resolution of a Listeria monocytogenes infection. J. Immunol. 2003, 171, 37–46. [Google Scholar] [CrossRef]
  198. Murata, H.; Tanaka, S.; Okada, H. Immune Tolerance of the Human Decidua. J. Clin. Med. 2021, 10, 351. [Google Scholar] [CrossRef]
  199. Chegini, N.; Ma, C.; Roberts, M.; Williams, R.S.; Ripps, B.A. Differential expression of interleukins (IL) IL-13 and IL-15 throughout the menstrual cycle in endometrium of normal fertile women and women with recurrent spontaneous abortion. J. Reprod. Immunol. 2002, 56, 93–110. [Google Scholar] [CrossRef]
  200. Toth, B.; Haufe, T.; Scholz, C.; Kuhn, C.; Friese, K.; Karamouti, M.; Makrigiannakis, A.; Jeschke, U. Placental interleukin-15 expression in recurrent miscarriage. Am. J. Reprod. Immunol. 2010, 64, 402–410. [Google Scholar] [CrossRef]
  201. Yoshimoto, T.; Takeda, K.; Tanaka, T.; Ohkusu, K.; Kashiwamura, S.; Okamura, H.; Akira, S.; Nakanishi, K. IL-12 up-regulates IL-18 receptor expression on T cells, Th1 cells, and B cells: Synergism with IL-18 for IFN-gamma production. J. Immunol. 1998, 161, 3400–3407. [Google Scholar] [PubMed]
  202. Hyodo, Y.; Matsui, K.; Hayashi, N.; Tsutsui, H.; Kashiwamura, S.; Yamauchi, H.; Hiroishi, K.; Takeda, K.; Tagawa, Y.; Iwakura, Y.; et al. IL-18 up-regulates perforin-mediated NK activity without increasing perforin messenger RNA expression by binding to constitutively expressed IL-18 receptor. J. Immunol. 1999, 162, 1662–1668. [Google Scholar] [PubMed]
  203. Nakanishi, K.; Yoshimoto, T.; Tsutsui, H.; Okamura, H. Interleukin-18 regulates both Th1 and Th2 responses. Annu. Rev. Immunol. 2001, 19, 423–474. [Google Scholar] [CrossRef] [PubMed]
  204. Comba, C.; Bastu, E.; Dural, O.; Yasa, C.; Keskin, G.; Ozsurmeli, M.; Buyru, F.; Serdaroglu, H. Role of inflammatory mediators in patients with recurrent pregnancy loss. Fertil. Steril. 2015, 104, 1467–1474.e1461. [Google Scholar] [CrossRef]
  205. Chen, X.; Jiang, L.; Wang, C.C.; Huang, J.; Li, T.C. Hypoxia inducible factor and microvessels in peri-implantation endometrium of women with recurrent miscarriage. Fertil. Steril. 2016, 105, 1496–1502.e1494. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  206. Chen, X.; Liu, Y.; Cheung, W.C.; Zhao, Y.; Huang, J.; Chung, J.P.W.; Wang, C.C.; Li, T.C. Increased expression of angiogenic cytokines in CD56+ uterine natural killer cells from women with recurrent miscarriage. Cytokine 2018, 110, 272–276. [Google Scholar] [CrossRef]
  207. Ma, J.; Zhang, X.; He, G.; Yang, C. Association between TNF, IL1B, IL6, IL10 and IFNG polymorphisms and recurrent miscarriage: A case control study. Reprod. Biol. Endocrinol. 2017, 15, 83. [Google Scholar] [CrossRef]
  208. Bombell, S.; McGuire, W. Cytokine polymorphisms in women with recurrent pregnancy loss: Meta-analysis. Aust. N. Z. J. Obstet. Gynaecol. 2008, 48, 147–154. [Google Scholar] [CrossRef]
  209. Zhang, B.; Liu, T.; Wang, Z. Association of tumor necrosis factor-α gene promoter polymorphisms (-308G/A, -238G/A) with recurrent spontaneous abortion: A meta-analysis. Hum. Immunol. 2012, 73, 574–579. [Google Scholar] [CrossRef]
  210. Li, H.H.; Xu, X.H.; Tong, J.; Zhang, K.Y.; Zhang, C.; Chen, Z.J. Association of TNF-α genetic polymorphisms with recurrent pregnancy loss risk: A systematic review and meta-analysis. Reprod. Biol. Endocrinol. 2016, 14, 6. [Google Scholar] [CrossRef] [Green Version]
  211. Shi, X.; Xie, X.; Jia, Y.; Li, S. Maternal genetic polymorphisms and unexplained recurrent miscarriage: A systematic review and meta-analysis. Clin. Genet. 2017, 91, 265–284. [Google Scholar] [CrossRef]
  212. Zammiti, W.; Mtiraoui, N.; Finan, R.R.; Almawi, W.Y.; Mahjoub, T. Tumor necrosis factor alpha and lymphotoxin alpha haplotypes in idiopathic recurrent pregnancy loss. Fertil. Steril. 2009, 91, 1903–1908. [Google Scholar] [CrossRef]
  213. Finan, R.R.; Al-Irhayim, Z.; Mustafa, F.E.; Al-Zaman, I.; Mohammed, F.A.; Al-Khateeb, G.M.; Madan, S.; Issa, A.A.; Almawi, W.Y. Tumor necrosis factor-alpha polymorphisms in women with idiopathic recurrent miscarriage. J. Reprod. Immunol. 2010, 84, 186–192. [Google Scholar] [CrossRef]
  214. Liu, C.; Wang, J.; Zhou, S.; Wang, B.; Ma, X. Association between -238 but not -308 polymorphism of Tumor necrosis factor alpha (TNF-alpha)v and unexplained recurrent spontaneous abortion (URSA) in Chinese population. Reprod. Biol. Endocrinol. 2010, 8, 114. [Google Scholar] [CrossRef] [Green Version]
  215. Gupta, R.; Prakash, S.; Parveen, F.; Agrawal, S. Association of CTLA-4 and TNF-α polymorphism with recurrent miscarriage among North Indian women. Cytokine 2012, 60, 456–462. [Google Scholar] [CrossRef]
  216. Alkhuriji, A.F.; Alhimaidi, A.R.; Babay, Z.A.; Wary, A.S. The relationship between cytokine gene polymorphism and unexplained recurrent spontaneous abortion in Saudi females. Saudi Med. J. 2013, 34, 484–489. [Google Scholar]
  217. Lee, B.E.; Jeon, Y.J.; Shin, J.E.; Kim, J.H.; Choi, D.H.; Jung, Y.W.; Shim, S.H.; Lee, W.S.; Kim, N.K. Tumor necrosis factor-α gene polymorphisms in Korean patients with recurrent spontaneous abortion. Reprod. Sci. 2013, 20, 408–413. [Google Scholar] [CrossRef] [Green Version]
  218. Babbage, S.J.; Arkwright, P.D.; Vince, G.S.; Perrey, C.; Pravica, V.; Quenby, S.; Bates, M.; Hutchinson, I.V. Cytokine promoter gene polymorphisms and idiopathic recurrent pregnancy loss. J. Reprod. Immunol. 2001, 51, 21–27. [Google Scholar] [CrossRef]
  219. Daher, S.; Shulzhenko, N.; Morgun, A.; Mattar, R.; Rampim, G.F.; Camano, L.; DeLima, M.G. Associations between cytokine gene polymorphisms and recurrent pregnancy loss. J. Reprod. Immunol. 2003, 58, 69–77. [Google Scholar] [CrossRef]
  220. Pietrowski, D.; Bettendorf, H.; Keck, C.; Bürkle, B.; Unfried, G.; Riener, E.K.; Hefler, L.A.; Tempfer, C. Lack of association of TNFalpha gene polymorphisms and recurrent pregnancy loss in Caucasian women. J. Reprod. Immunol. 2004, 61, 51–58. [Google Scholar] [CrossRef]
  221. Prigoshin, N.; Tambutti, M.; Larriba, J.; Gogorza, S.; Testa, R. Cytokine gene polymorphisms in recurrent pregnancy loss of unknown cause. Am. J. Reprod. Immunol. 2004, 52, 36–41. [Google Scholar] [CrossRef]
  222. Kamali-Sarvestani, E.; Zolghadri, J.; Gharesi-Fard, B.; Sarvari, J. Cytokine gene polymorphisms and susceptibility to recurrent pregnancy loss in Iranian women. J. Reprod. Immunol. 2005, 65, 171–178. [Google Scholar] [CrossRef]
  223. Pravica, V.; Perrey, C.; Stevens, A.; Lee, J.H.; Hutchinson, I.V. A single nucleotide polymorphism in the first intron of the human IFN-gamma gene: Absolute correlation with a polymorphic CA microsatellite marker of high IFN-gamma production. Hum. Immunol. 2000, 61, 863–866. [Google Scholar] [CrossRef]
  224. Lin, M.T.; Storer, B.; Martin, P.J.; Tseng, L.H.; Gooley, T.; Chen, P.J.; Hansen, J.A. Relation of an interleukin-10 promoter polymorphism to graft-versus-host disease and survival after hematopoietic-cell transplantation. N. Engl. J. Med. 2003, 349, 2201–2210. [Google Scholar] [CrossRef] [Green Version]
  225. Temple, S.E.; Lim, E.; Cheong, K.Y.; Almeida, C.A.; Price, P.; Ardlie, K.G.; Waterer, G.W. Alleles carried at positions -819 and -592 of the IL10 promoter affect transcription following stimulation of peripheral blood cells with Streptococcus pneumoniae. Immunogenetics 2003, 55, 629–632. [Google Scholar] [CrossRef]
  226. Gu, C.; Gong, H.; Zhang, Z.; Yang, Z.; Ma, Y. Association of interleukin-10 gene promoter polymorphisms with recurrent pregnancy loss: A meta-analysis. J. Assist. Reprod. Genet. 2016, 33, 907–917. [Google Scholar] [CrossRef] [Green Version]
  227. Medica, I.; Ostojic, S.; Pereza, N.; Kastrin, A.; Peterlin, B. Association between genetic polymorphisms in cytokine genes and recurrent miscarriage--a meta-analysis. Reprod. Biomed. Online 2009, 19, 406–414. [Google Scholar] [CrossRef]
  228. Parveen, F.; Shukla, A.; Agarwal, S. Cytokine gene polymorphisms in northern Indian women with recurrent miscarriages. Fertil. Steril. 2013, 99, 433–440. [Google Scholar] [CrossRef]
  229. Peng, Z.; Lv, X.; Sun, Y.; Dai, S. Association of Interleukin-10-1082A/G Polymorphism with Idiopathic Recurrent Miscarriage: A Systematic Review and Meta-Analysis. Am. J. Reprod. Immunol. 2016, 75, 162–171. [Google Scholar] [CrossRef]
  230. Karhukorpi, J.; Laitinen, T.; Karttunen, R.; Tiilikainen, A.S. The functionally important IL-10 promoter polymorphism (-1082G-->A) is not a major genetic regulator in recurrent spontaneous abortions. Mol. Hum. Reprod. 2001, 7, 201–203. [Google Scholar] [CrossRef] [Green Version]
  231. Zammiti, W.; Mtiraoui, N.; Cochery-Nouvellon, E.; Mahjoub, T.; Almawi, W.Y.; Gris, J.C. Association of -592C/A, -819C/T and -1082A/G interleukin-10 promoter polymorphisms with idiopathic recurrent spontaneous abortion. Mol. Hum. Reprod. 2006, 12, 771–776. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  232. Kim, J.O.; Lee, W.S.; Lee, B.E.; Jeon, Y.J.; Kim, Y.R.; Jung, S.H.; Chang, S.W.; Kim, N.K. Interleukin-1beta -511T>C genetic variant contributes to recurrent pregnancy loss risk and peripheral natural killer cell proportion. Fertil. Steril. 2014, 102, 206–212.e205. [Google Scholar] [CrossRef]
  233. Saijo, Y.; Sata, F.; Yamada, H.; Konodo, T.; Kato, E.H.; Kataoka, S.; Shimada, S.; Morikawa, M.; Minakami, H.; Kishi, R. Interleukin-4 gene polymorphism is not involved in the risk of recurrent pregnancy loss. Am. J. Reprod. Immunol. 2004, 52, 143–146. [Google Scholar] [CrossRef]
  234. Gremlich, S.; Fratta, S.; Rebellato, E.; Uras, R.; Reymondin, D.; Damnon, F.; Germond, M.; Gerber, S. Interleukin-1 receptor antagonist gene (IL-1RN) polymorphism is a predictive factor of clinical pregnancy after IVF. Hum. Reprod. 2008, 23, 1200–1206. [Google Scholar] [CrossRef] [Green Version]
  235. Naeimi, S.; Ghiam, A.F.; Mojtahedi, Z.; Dehaghani, A.S.; Amani, D.; Ghaderi, A. Interleukin-18 gene promoter polymorphisms and recurrent spontaneous abortion. Eur. J. Obstet. Gynecol. Reprod. Biol. 2006, 128, 5–9. [Google Scholar] [CrossRef] [Green Version]
  236. Ostojić, S.; Volk, M.; Medica, I.; Kapović, M.; Meden-Vrtovec, H.; Peterlin, B. Polymorphisms in the interleukin-12/18 genes and recurrent spontaneous abortion. Am. J. Reprod. Immunol. 2007, 58, 403–408. [Google Scholar] [CrossRef]
  237. Yue, J.; Tong, Y.; Zhou, J.; Liu, Q.; Yang, J. Genetic variant in interleukin-18 is associated with idiopathic recurrent miscarriage in Chinese Han population. Int. J. Mol. Sci. 2015, 16, 4180–4189. [Google Scholar] [CrossRef] [Green Version]
  238. Al-Khateeb, G.M.; Sater, M.S.; Finan, R.R.; Mustafa, F.E.; Al-Busaidi, A.S.; Al-Sulaiti, M.A.; Almawi, W.Y. Analysis of interleukin-18 promoter polymorphisms and changes in interleukin-18 serum levels underscores the involvement of interleukin-18 in recurrent spontaneous miscarriage. Fertil. Steril. 2011, 96, 921–926. [Google Scholar] [CrossRef]
  239. Messaoudi, S.; Dandana, M.; Magdoud, K.; Meddeb, S.; Ben Slama, N.; Hizem, S.; Mahjoub, T. Interleukin-18 promoter polymorphisms and risk of idiopathic recurrent pregnancy loss in a Tunisian population. J. Reprod. Immunol. 2012, 93, 109–113. [Google Scholar] [CrossRef]
  240. Unfried, G.; Böcskör, S.; Endler, G.; Nagele, F.; Huber, J.C.; Tempfer, C.B. A polymorphism of the interleukin-6 gene promoter and idiopathic recurrent miscarriage. Hum. Reprod. 2003, 18, 267–270. [Google Scholar] [CrossRef]
  241. Saijo, Y.; Sata, F.; Yamada, H.; Kondo, T.; Kato, E.H.; Kishi, R. Single nucleotide polymorphisms in the promoter region of the interleukin-6 gene and the risk of recurrent pregnancy loss in Japanese women. Fertil. Steril. 2004, 81, 374–378. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  242. Demirturk, F.; Ates, O.; Gunal, O.; Bozkurt, N.; Aysal, T.; Nacar, M.C. IL-6 gene promoter polymorphisms: Genetic susceptibility to recurrent pregnancy loss. Bratisl. Lek. Listy 2014, 115, 479–482. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  243. Ma, X.; Xu, L.J.; Wang, J.; Xian, M.M.; Liu, M. Association of IL-1β and IL-6 gene polymorphisms with recurrent spontaneous abortion in a Chinese Han population. Int. J. Immunogenet. 2012, 39, 15–19. [Google Scholar] [CrossRef] [PubMed]
  244. Reid, J.G.; Simpson, N.A.; Walker, R.G.; Economidou, O.; Shillito, J.; Gooi, H.C.; Duffy, S.R.; Walker, J.J. The carriage of pro-inflammatory cytokine gene polymorphisms in recurrent pregnancy loss. Am. J. Reprod. Immunol. 2001, 45, 35–40. [Google Scholar] [CrossRef] [PubMed]
  245. Hefler, L.A.; Tempfer, C.B.; Bashford, M.T.; Unfried, G.; Zeillinger, R.; Schneeberger, C.; Koelbl, H.; Nagele, F.; Huber, J.C. Polymorphisms of the angiotensinogen gene, the endothelial nitric oxide synthase gene, and the interleukin-1beta gene promoter in women with idiopathic recurrent miscarriage. Mol. Hum. Reprod. 2002, 8, 95–100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  246. Linjawi, S.; Li, T.C.; Laird, S.; Blakemore, A. Interleukin-1 receptor antagonist and interleukin-1 beta polymorphisms in women with recurrent miscarriage. Fertil. Steril. 2005, 83, 1549–1552. [Google Scholar] [CrossRef] [PubMed]
  247. Unfried, G.; Tempfer, C.; Schneeberger, C.; Widmar, B.; Nagele, F.; Huber, J.C. Interleukin 1 receptor antagonist polymorphism in women with idiopathic recurrent miscarriage. Fertil. Steril. 2001, 75, 683–687. [Google Scholar] [CrossRef]
  248. Wang, Z.C.; Yunis, E.J.; De los Santos, M.J.; Xiao, L.; Anderson, D.J.; Hill, J.A. T helper 1-type immunity to trophoblast antigens in women with a history of recurrent pregnancy loss is associated with polymorphism of the IL1B promoter region. Genes Immun. 2002, 3, 38–42. [Google Scholar] [CrossRef]
  249. Karhukorpi, J.; Laitinen, T.; Kivelä, H.; Tiilikainen, A.; Hurme, M. IL-1 receptor antagonist gene polymorphism in recurrent spontaneous abortion. J. Reprod. Immunol. 2003, 58, 61–67. [Google Scholar] [CrossRef]
  250. Daher, S.; Mattar, R.; Gueuvoghlanian-Silva, B.Y.; Torloni, M.R. Genetic polymorphisms and recurrent spontaneous abortions: An overview of current knowledge. Am. J. Reprod. Immunol. 2012, 67, 341–347. [Google Scholar] [CrossRef]
  251. Shivers, K.Y.; Amador, N.; Abrams, L.; Hunter, D.; Jenab, S.; Quiñones-Jenab, V. Estrogen alters baseline and inflammatory-induced cytokine levels independent from hypothalamic-pituitary-adrenal axis activity. Cytokine 2015, 72, 121–129. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  252. Arck, P.; Hansen, P.J.; Mulac Jericevic, B.; Piccinni, M.P.; Szekeres-Bartho, J. Progesterone during pregnancy: Endocrine-immune cross talk in mammalian species and the role of stress. Am. J. Reprod. Immunol. 2007, 58, 268–279. [Google Scholar] [CrossRef] [PubMed]
  253. Piccinni, M.P.; Scaletti, C.; Vultaggio, A.; Maggi, E.; Romagnani, S. Defective production of LIF, M-CSF and Th2-type cytokines by T cells at fetomaternal interface is associated with pregnancy loss. J. Reprod. Immunol. 2001, 52, 35–43. [Google Scholar] [CrossRef] [PubMed]
  254. Raghupathy, R.; Al-Azemi, M. Modulation of Cytokine Production by the Dydrogesterone Metabolite Dihydrodydrogesterone. Am. J. Reprod. Immunol. 2015, 74, 419–426. [Google Scholar] [CrossRef]
  255. Lissauer, D.; Eldershaw, S.A.; Inman, C.F.; Coomarasamy, A.; Moss, P.A.; Kilby, M.D. Progesterone promotes maternal-fetal tolerance by reducing human maternal T-cell polyfunctionality and inducing a specific cytokine profile. Eur. J. Immunol. 2015, 45, 2858–2872. [Google Scholar] [CrossRef] [Green Version]
  256. AbdulHussain, G.; Azizieh, F.; Makhseed, M.; Raghupathy, R. Effects of Progesterone, Dydrogesterone and Estrogen on the Production of Th1/Th2/Th17 Cytokines by Lymphocytes from Women with Recurrent Spontaneous Miscarriage. J. Reprod. Immunol. 2020, 140, 103132. [Google Scholar] [CrossRef]
  257. Devall, A.J.; Papadopoulou, A.; Podesek, M.; Haas, D.M.; Price, M.J.; Coomarasamy, A.; Gallos, I.D. Progestogens for preventing miscarriage: A network meta-analysis. Cochrane Database Syst. Rev 2021, 4, Cd013792. [Google Scholar] [CrossRef]
  258. Coomarasamy, A.; Truchanowicz, E.G.; Rai, R. Does first trimester progesterone prophylaxis increase the live birth rate in women with unexplained recurrent miscarriages? BMJ 2011, 342, d1914. [Google Scholar] [CrossRef]
  259. Verma, P.; Verma, R.; Nair, R.R.; Budhwar, S.; Khanna, A.; Agrawal, N.R.; Sinha, R.; Birendra, R.; Rajender, S.; Singh, K. Altered crosstalk of estradiol and progesterone with Myeloid-derived suppressor cells and Th1/Th2 cytokines in early miscarriage is associated with early breakdown of maternal-fetal tolerance. Am. J. Reprod. Immunol. 2019, 81, e13081. [Google Scholar] [CrossRef]
  260. Matalka, K.Z.; Ali, D.A. Stress-induced versus preovulatory and pregnancy hormonal levels in modulating cytokine production following whole blood stimulation. Neuroimmunomodulation 2005, 12, 366–374. [Google Scholar] [CrossRef]
  261. Wang, C.; Dehghani, B.; Li, Y.; Kaler, L.J.; Vandenbark, A.A.; Offner, H. Oestrogen modulates experimental autoimmune encephalomyelitis and interleukin-17 production via programmed death 1. Immunology 2009, 126, 329–335. [Google Scholar] [CrossRef] [PubMed]
  262. Stephenson, M.D.; Kutteh, W.H.; Purkiss, S.; Librach, C.; Schultz, P.; Houlihan, E.; Liao, C. Intravenous immunoglobulin and idiopathic secondary recurrent miscarriage: A multicentered randomized placebo-controlled trial. Hum. Reprod. 2010, 25, 2203–2209. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  263. Schleussner, E.; Kamin, G.; Seliger, G.; Rogenhofer, N.; Ebner, S.; Toth, B.; Schenk, M.; Henes, M.; Bohlmann, M.K.; Fischer, T.; et al. Low-molecular-weight heparin for women with unexplained recurrent pregnancy loss: A multicenter trial with a minimization randomization scheme. Ann. Intern. Med. 2015, 162, 601–609. [Google Scholar] [CrossRef] [PubMed]
  264. Zafardoust, S.; Akhondi, M.M.; Sadeghi, M.R.; Mohammadzadeh, A.; Karimi, A.; Jouhari, S.; Ansaripour, S. Efficacy of Intrauterine Injection of Granulocyte Colony Stimulating Factor (G-CSF) on Treatment of Unexplained Recurrent Miscarriage: A Pilot RCT Study. J. Reprod. Infertil. 2017, 18, 379–385. [Google Scholar]
Figure 1. The roles of pro- and anti-inflammatory cytokines in early pregnancy. IL-6, IL-11, IL-15, and TGF-β are involved in regulating decidualization. Furthermore, IL-1β contributes to decidualization by elevating the generation of COX2 and PGE2. Furthermore, IL-1β elevates the expression of MMP, a critical regulator of trophoblast invasion. TGF-β1 reduces the invasion of trophoblast cells by enhancing kisspeptin expression. TNF-α induces trophoblast invasion through increasing IL-17 production from endometrial stromal cells. Moreover, TNF-α plays an important role in implantation by inducing VEGF generation from trophoblast cells. IFN-γ, IL-6, IL-10, IL-11, IL-1β, and IL-22 are associated with embryo implantation. In addition, IL-6, IL-10, IL-11, and IL-1β participate in placentation.
Figure 1. The roles of pro- and anti-inflammatory cytokines in early pregnancy. IL-6, IL-11, IL-15, and TGF-β are involved in regulating decidualization. Furthermore, IL-1β contributes to decidualization by elevating the generation of COX2 and PGE2. Furthermore, IL-1β elevates the expression of MMP, a critical regulator of trophoblast invasion. TGF-β1 reduces the invasion of trophoblast cells by enhancing kisspeptin expression. TNF-α induces trophoblast invasion through increasing IL-17 production from endometrial stromal cells. Moreover, TNF-α plays an important role in implantation by inducing VEGF generation from trophoblast cells. IFN-γ, IL-6, IL-10, IL-11, IL-1β, and IL-22 are associated with embryo implantation. In addition, IL-6, IL-10, IL-11, and IL-1β participate in placentation.
Ijms 24 00132 g001
Figure 2. Schematic diagram of the unbalanced Th1/Th2 cytokine network in peripheral blood of RPL patients. TNF-α can be produced by Th1 or Th2 cells. IFN-γ is secreted by Th1 cells, which induce the differentiation of Th1 cells. Moreover, TGF-β augments the differentiation of Th1 cells, which release IL-1β. In addition, IL-33 can enhance Th1 or Th2 differentiation. Th2 cells produce IL-4 and IL-13. IL-4 is known to compromise the production of Th1 cells. IL-6 is found to promote Th2 cells. IL-10 is a Th2 cytokine that inhibits the Th1 immune response.
Figure 2. Schematic diagram of the unbalanced Th1/Th2 cytokine network in peripheral blood of RPL patients. TNF-α can be produced by Th1 or Th2 cells. IFN-γ is secreted by Th1 cells, which induce the differentiation of Th1 cells. Moreover, TGF-β augments the differentiation of Th1 cells, which release IL-1β. In addition, IL-33 can enhance Th1 or Th2 differentiation. Th2 cells produce IL-4 and IL-13. IL-4 is known to compromise the production of Th1 cells. IL-6 is found to promote Th2 cells. IL-10 is a Th2 cytokine that inhibits the Th1 immune response.
Ijms 24 00132 g002
Figure 3. An illustration of the imbalanced Treg/Th17 cytokines in the periphery of RPL patients. IL-35 is predominantly produced by Treg cells. IL-7 promotes the differentiation of Th17 cells and compromises Treg cells, promoting inflammatory responses. Th17 cells secrete IL-17 and IL-22. IL-6 induces the differentiation of Th17 cells. In addition, IL-23 increases the number of Th17 cells. IL-33 contributes to the Th17 immune response. On the contrary, IL-27 impedes the expression of Th17 cells. Accordingly, aberrant expression of these cytokines in peripheral blood may cause Treg/Th17 immune imbalance, triggering the pro-inflammatory immune response and eventually leading to RPL.
Figure 3. An illustration of the imbalanced Treg/Th17 cytokines in the periphery of RPL patients. IL-35 is predominantly produced by Treg cells. IL-7 promotes the differentiation of Th17 cells and compromises Treg cells, promoting inflammatory responses. Th17 cells secrete IL-17 and IL-22. IL-6 induces the differentiation of Th17 cells. In addition, IL-23 increases the number of Th17 cells. IL-33 contributes to the Th17 immune response. On the contrary, IL-27 impedes the expression of Th17 cells. Accordingly, aberrant expression of these cytokines in peripheral blood may cause Treg/Th17 immune imbalance, triggering the pro-inflammatory immune response and eventually leading to RPL.
Ijms 24 00132 g003
Table 1. Cytokines exert anti- or pro-implantation properties.
Table 1. Cytokines exert anti- or pro-implantation properties.
Anti-Implantation
IL-6[47]
IL-17[48]
IL-23[48]
Pro-Implantation
TNF-α[49]
IFN-γ[28]
TGF-β1[50]
IL-1[51]
IL-1β[52]
IL-10[53]
IL-11[54]
IL-22[55]
IL-33[56]
Table 2. Alternations of cytokines in peripheral blood of RPL women compared with normal females.
Table 2. Alternations of cytokines in peripheral blood of RPL women compared with normal females.
CytokinesExpressionNumber of the Experimental GroupNumber of the Control GroupReference
TNF-α Increased2324[37]
IFN-γIncreased23 24 [37]
TGF-βIncreased3630 [63]
No difference32 32[64]
No difference2927 [65]
IL-33Decreased142123[66]
IL-4Increased1915[35]
No difference32 32[64]
IL-6Decreased23 24 [37]
IL-10Decreased2328[57]
Decreased1916[36]
Decreased2324[37]
Decreased3232[64]
Decreased3030[67]
IL-17Increased2020[48]
IL-22Increased4628[68]
IL-23Increased3030[67]
Increased1515[69]
IL-35Decreased6040[70]
Decreased40120[71]
Table 3. Changes of cytokines in endometrium/decidua of RPL patients compared with healthy women.
Table 3. Changes of cytokines in endometrium/decidua of RPL patients compared with healthy women.
CytokinesExpressionNumber of the Experimental GroupNumber of the Control GroupReference
TNF-αIncreased3540[137]
IFN-γIncreased1010[138]
TGF-βDecreased3540[137]
IL-1βIncreased1515[31]
IL-4Decreased1010[138]
IL-6Increased3540[137]
Decreased912[139]
IL-10Decreased1010[138]
IL-11Decreased169[140]
IL-17Increased1515[69]
IL-18Increased159[141]
IL-23Increased1515[69]
Table 4. Studies for TNF-α gene polymorphisms and RPL.
Table 4. Studies for TNF-α gene polymorphisms and RPL.
Gene PolymorphismAuthorsRaceNumber of Study and Control GroupsGenotypeTest MethodConclusions
Study GroupControl Group
−238G/AZammiti, W. et al., 2009 [212]Tunisian372 vs. 274264/88/20 a215/52/7 aPCR-RFLPThe polymorphism of the −238G/A gene was associated with the occurrence of RPL.
Finan, R. et al., 2010 [213]Bahraini Arabs204 vs. 248148/52/4 a200/48/0 aPCR-RFLP−238G/A variants were independent risk factors for RPL.
Liu, C. et al., 2010 [214]Chinese132 vs. 152128/4/0 a135/17/0 aPCRA statistical difference was exhibited in −238G/A polymorphism.
Gupta, R. et al., 2012 [215]Indian300 vs. 500121/63/16 a154/113/33 aPCR-RFLPRPL women tended to carry the G allele.
Alkhuriji, A. et al., 2013 [216]Saudis65 vs. 6557/8/0 a55/7/3 aPCRNS
Lee, B. et al., 2013 [217]Korean357 vs. 236330/26/1 a228/8/0 aPCR-RFLPTNF-α −238G > A variants elevated the incidence of RPL.
Ma, J. et al., 2017 [207]Chinese775 vs. 805732/41/2 a745/57/3 aPCR-RFLPNS
−308G/ABabbage, S. et al., 2001 [218]Caucasian43 vs. 7330/13 b56/17 bPCRNS
Daher, S. et al., 2003 [219]Brazilian48 vs. 10836/12 b89/19 bPCRNS
Pietrowski, D. et al., 2004 [220]Caucasian168 vs. 212133/33/2 a167/41/4 aPCRNS
Prigoshin, N. et al., 2004 [221]Argentinean41 vs. 5435/6 b49/5 bPCR-SSPNS
Kamali-Sarvestani, E. et al., 2005 [222]Iranian139 vs. 143117/14 b122/21 bPCRNS
Zammiti, W. et al., 2009 [212]Tunisian372 vs. 274319/39/14 a222/47/5 aPCR-RFLPNS
Finan, W. et al., 2010 [213]Bahraini Arabs204 vs. 248164/32/8 a212/32/4 aPCR-RFLPNS
Liu, C. et al., 2010 [214]Chinese132 vs. 152110/22/0 a138/13/1 aPCRNS
Gupta, R. et al., 2012 [215]Indian300 vs. 500229/62/9 a425/70/5 aPCR-RFLPThe A allele was more likely to be present in RPL women.
Alkhuriji, A. et al., 2013 [216]Saudis65 vs. 6533/24/8 a47/14/4 aPCRThe −308G > A gene polymorphisms were associated with RPL.
Lee, B. et al., 2013 [217]Korean357 vs. 236319/36/2 a213/21/2 aPCR- RFLPNS
Ma, J. et al., 2017 [207]Chinese775 vs. 805683/86/6 a726/76/3 aPCR- RFLPNS
−1031T/CFinan, R. et al., 2010 [213]Bahraini Arabs204 vs. 248152/36/16 a219/29/0 aPCR-RFLPThe frequency of the −1031C allele in RPL was significantly increased.
Lee, B. et al., 2013 [217]Korean357 vs. 236230/115/12 a191/45/0 aPCR-RFLPTNF-α − 1031T > C variants augmented the chance of experiencing RPL.
−376G/AFinan, R. et al., 2010 [213]Bahraini Arabs204 vs. 248174/20/10 a226/22/0 aPCR-RFLPPatients with RPL had a higher incidence of carrying the −376A allele.
a Genotype for TNF-α − 238G/A, GG/GA/AA; for TNF-α − 308G/A, GG/GA/AA; for TNF-α − 1031T/C, TT/TC/CC; for TNF-α − 376G/A, GG/GA/AA; b Genotype for TNF-α − 308G/A, GG/GA+AA; PCR, polymerase chain reaction; RFLP, restriction fragment length polymorphism; SSP, sequence specific primers; NS, no significance.
Table 5. Studies for IL-10 gene polymorphisms and RPL.
Table 5. Studies for IL-10 gene polymorphisms and RPL.
Gene PolymorphismAuthorsRaceNumber of Study and Control GroupsGenotypeTest MethodConclusions
Study GroupControl Group
−1082A/GBabbage, S. et al., 2001 [218]Caucasian43 vs. 738/23/12 a20/41/12 aPCRNS
Karhukorpi, J. et al., 2001 [230]Finnish38 vs. 13113/16/9 a44/64/23 aPCRNS
Daher, S. et al., 2003 [219]Brazilian43 vs. 10413/19/11 a45/43/16 aPCRNS
Kamali-Sarvestani, E. et al., 2005 [222]Iranian139 vs. 14362/41/24 a62/47/21 aPCR-RFLPNS
Zammiti, W. et al., 2006 [231]Tunisian350 vs. 20087/185/72 a54/107/39 aPCR-ASANS
Parveen, F. et al., 2013 [228]Indian200 vs. 30086/99/15 a180/108/12 aPCRThe A allele has a significant protective effect.
Kim, J. et al., 2014 [232]Korean385 vs. 232333/50/2 a198/34/0 aPCR-RFLPNS
Ma, J. et al., 2017 [207]Chinese775 vs. 805683/88/4 a685/113/7 aPCR-RFLPNS
−819C/TKamali-Sarvestani, E. et al., 2005 [222]Iranian139 vs. 14377/49/13 a61/56/15 aPCR-RFLPNS
Zammiti, W. et al., 2006 [231]Tunisian350 vs. 200182/120/48 a124/57/19 aPCR-ASAThe −819C/T polymorphisms were genetically associated with RPL.
Parveen, F. et al., 2013 [228]Indian200 vs. 30059/111/30 a122/142/36 aPCRRPL females tended to own the CT genotype.
−592C/AKamali-Sarvestani, E. et al., 2005 [222]Iranian139 vs. 14383/35/14 a61/56/15 aPCR-RFLP−592C/A polymorphism variants were one of the genetic causes of RPL.
Zammiti, W. et al., 2006 [231]Tunisian350 vs. 200206/93/51 a134/41/25 aPCR-ASANS
Parveen, F. et al., 2013 [228]Indian200 vs. 30091/79/30 a148/116/36 aPCRNS
a Genotype for IL-10 − 1082A/G, AA/AG/GG; for IL-10 − 819C/T, CC/CT/TT; for IL-10 − 592C/A, CC/CA/AA; ASA, allele-specific amplification.
Table 6. Researches about IL-18 gene polymorphisms in RPL.
Table 6. Researches about IL-18 gene polymorphisms in RPL.
Gene PolymorphismAuthors RaceNumber of Study and Control GroupsGenotypeTest MethodConclusions
Study GroupControl Group
−607C/ANaeimi, S. et al., 2006 [235]Iranian102 vs. 10337/23/42 a32/17/54 aPCRNS
Ostojic, S. et al., 2007 [236]Slovenian125 vs. 13643/68/14 a41/79/16 aPCRNS
Yue, J. et al., 2015 [237]Chinese484 vs. 46887/216/181 a79/211/178 aPCRNS
−137G/CNaeimi, S. et al., 2006 [235]Iranian102 vs. 10357/40/5 a56/39/8 aPCRNS
Ostojic, S. et al., 2007 [236]Slovenian125 vs. 13659/54/12 a62/63/11 aPCRNS
Al-Khateeb, G. et al., 2011 [238]Bahraini282 vs. 283146/98/38 a152/113/24 aPCRNS
Messaoudi, S. et al., 2012 [239]Tunisian235 vs. 235122/82/31 a126/92/19 aPCRNS
Yue, J. et al., 2015 [237]Chinese484 vs. 468338/108/38 a357/102/9 aPCR−137G/C variants had statistical relationship with RPL in additive and recessive genetic models.
−656C/AAl-Khateeb, G. et al., 2011 [238]Bahraini282 vs. 28380/144/58 a140/119/30 aPCR−656C/A variants were related to RPL.
Messaoudi, S. et al., 2012 [239]Tunisian235 vs. 23566/120/49 a114/97/24 aPCRThe genotype frequency of −656C/A was significantly correlated with the occurrence of RPL.
−119A/CAl-Khateeb, G. et al., 2011 [238]Bahraini282 vs. 283157/99/26 a155/109/25 aPCRNS
Messaoudi, S. et al., 2012 [239]Tunisian235 vs. 235132/82/21 a127/89/19 aPCRNS
−105G/AAl-Khateeb, G. et al., 2011 [238]Bahraini282 vs. 28398/94/90 a146/110/33 aPCR−105G/A variant was prominently associated with RPL.
Messaoudi, S. et al., 2012 [239]Tunisia235 vs. 23582/78/75 a120/89/26 aPCRThe genotype frequency of −105G/A was significantly different between the two groups.
Yue, J. et al., 2015 [237]Chinese484 vs. 468349/129/6 a332/128/8 aPCRNS
a Genotype for IL-18 − 607C/A, CC/CA/AA; for IL-18 − 137G/C, GG/GC/CC; for IL-18 − 656C/A, CC/CA/AA; for IL-18 − 119A/C, AA/AC/CC; for IL-18 − 105G/A, GG/GA/AA.
Table 7. Studies for IL-6 gene polymorphisms and RPL.
Table 7. Studies for IL-6 gene polymorphisms and RPL.
Gene PolymorphismAuthorsRaceNumber of Study and Control GroupsGenotypeTest MethodConclusions
Study GroupControl Group
−174G/CUnfried, G. et al., 2003 [240]White Middle-
European Caucasian women
161 vs. 12466/72/23 a43/58/23 aPCRNS
Daher, S. et al., 2003 [219]Brazilian44 vs. 10839/5 b99/9 bPCRNS
Prigoshin, N et al., 2004 [221]Argentinean38 vs. 5435/3 b49/5 bPCR-SSPNS
Saijo, Y. et al., 2004 [241]Japanese76 vs. 9376/0 c93/0 cPCRNS
Demirturk, F. et al., 2014 [242]Turkish113 vs. 11372/36/5 a100/11/2 aPCR-RFLP−174G/C polymorphisms had a relationship with an elevated incidence of RPL.
Ma, J. et al., 2017 [207]Chinese775 vs. 805484/248/43 a463/291/51 aPCR- RFLPNS
−634C/GSaijo, Y. et al., 2004 [241]Japanese76 vs. 9358/18 c56/37 cPCRWomen with the G allele were more likely to develop RPL than those with the wild-type allele C.
Ma, X. et al., 2012 [243]Chinese162 vs. 156116/46/0 a93/52/11 aPCR-RFLPThe distributions of the GG genotype and G allele were significantly decreased in RPL.
Ma, J. et al., 2017 [207]Chinese775 vs. 805554/197/24 a478/277/50 aPCR- RFLPWomen with CG and GG genotypes were less likely to develop RPL.
−572G/CDemirturk, F. et al., 2014 [242]Turkish113 vs. 11381/28/4 a88/21/4 aPCR-RFLPNS
−597G/ADemirturk, F. et al., 2014 [242]Turkish113 vs. 11396/16/1 a87/26/0 aPCR-RFLPNS
−1363G/TDemirturk, F. et al., 2014 [242]Turkish113 vs. 11395/18/0 a94/19/0 aPCR-RFLPNS
−2954G/CDemirturk, F. et al., 2014 [242]Turkish113 vs. 113107/6/0 a112/1/0 aPCR-RFLP−2954G/C polymorphism variants were related to an elevated risk of RPL.
a Genotype for IL-6 − 174G/C, GG/GC/CC; for IL-6 − 634C/G, CC/CG/GG; for IL-6 − 572G/C, GG/GC/CC; for IL-6 − 597G/A, GG/GA/AA; for IL-6 − 1363G/T, GG/GT/TT; for IL-6 − 2954G/C, GG/GC/CC; b Genotype for IL-6 − 174G/C, GG+GC/CC; c Genotype for IL-6 − 174G/C, GG/GC+CC; for IL-6 − 634C/G, CC/CG+GG.
Table 8. Studies for IL-1β, IL-1RA, and IL-1RN gene polymorphisms and RPL.
Table 8. Studies for IL-1β, IL-1RA, and IL-1RN gene polymorphisms and RPL.
Gene PolymorphismAuthorsRaceNumber of Study and Control GroupsGenotypeTest MethodConclusions
Study GroupControl Group
IL-1β + 3954C→T
on exon 5
Hefler, L. et al., 2001 [83]Caucasian131 vs. 6879/46/6 a47/16/5 aPCRNS
Reid, J. et al., 2001 [244]British17 vs. 4311/6/0 b26/14/3 bPCRNS
Ma, X. et al., 2012 [243]Chinese162 vs. 156124/38/0 c130/26/0 cPCR-RFLPNS
Ma, J. et al., 2017 [207]Chinese775 vs. 805602/168/5 c632/166/7 cPCR-RFLPNS
IL-1β − 511C/THefler, L. et al., 2002 [245]Caucasian130 vs. 6729/90/11 c20/38/9 cPCRNS
Linjawi, S. et al., 2005 [246]British206 vs. 22469/117/20 c85/110/29 cPCRNS
Ma, X. et al., 2012 [243]Chinese162 vs. 15638/84/40 c46/84/26 cPCR-RFLPNS
Kim, J. et al., 2014 [232]Korean385 vs. 23296/190/99 c39/120/73 cPCR–RFLP−511C > T polymorphism was relevant to RPL.
Ma, J. et al., 2017 [207]Chinese775 vs. 805178/384/213 c156/392/257 cPCR-RFLPNS
IL-1 RAUnfried, G. et al., 2001 [247]Austrian105 vs. 910.9/0.34/0.5 d0.97/0.11/0.5 dPCRAllele 2 was a genetic risk factor for RPL.
IL-1 RNWang, Z. et al., 2002 [248]Caucasian118 vs. 6088/26/2/2 e39/20/1/0 ePCR-RFLPNS
Karhukorpi, J. et al., 2003 [249]Finnish37 vs. 80012/19/2/4 f374/343/66/17 fPCRThe frequency of the IL-1RN*3 allele was significantly increased in RPL patients.
Linjawi, S. et al., 2005 [246]British206 vs. 25912/79/115 g17/92/150 gPCRNS
a Genotype for IL-1β + 3954, E1/E1: E1/E2: E2/E2; b Genotype for IL-1β + 3954, 1, 1/1, 2/2, 2; c Genotype for IL-1β + 3954, CC/CT/TT; for IL-1β − 511C/T, CC/CT/TT; d Genotype for IL-1 RA, 1/2/3; e Genotype for IL-1 RN, 1/2/3/4; f Genotype for IL-1 RN, 1/1: 1/2: 2/2: 1/3 or 2/3; g Genotype for IL-1 RN, 2, 2: 2, 4: 4, 4.
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

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. 2023, 24, 132. https://doi.org/10.3390/ijms24010132

AMA Style

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. International Journal of Molecular Sciences. 2023; 24(1):132. https://doi.org/10.3390/ijms24010132

Chicago/Turabian Style

Yang, Xiuhua, Yingying Tian, Linlin Zheng, Thanh Luu, and Joanne Kwak-Kim. 2023. "The Update Immune-Regulatory Role of Pro- and Anti-Inflammatory Cytokines in Recurrent Pregnancy Losses" International Journal of Molecular Sciences 24, no. 1: 132. https://doi.org/10.3390/ijms24010132

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